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 PMCID: PMC12120924 DOI: 10.1021/acs.jctc.5c00256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [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, Beijing100871, China
- School
of Medicine, Fuzhou University, Fuzhou, Fujian350108, China
| | - Chen Song
- Center
for Quantitative Biology, Peking-Tsinghua Center for Life Sciences,
Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100871, China
| |
Collapse
|
2
|
Cordeiro S, Musinszki M. Thermosensitivity of TREK K2P channels is controlled by a PKA switch and depends on the microtubular network. Pflugers Arch 2025:10.1007/s00424-025-03089-1. [PMID: 40372488 DOI: 10.1007/s00424-025-03089-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/23/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
Temperature sensing is an essential component of animal perception and enables individuals to avoid painful or lethal temperatures. Many temperature sensors in central and peripheral neurons are ion channels. Here, we focus on the thermosensitive TREK/TRAAK subfamily of K2P channels-the only known K+ selective thermosensitive channels. The C-terminal domain is essential for the temperature activation of TREK channels, but the mechanism of temperature sensation and the nature of the temperature sensor are unknown. We studied the thermosensitivity of representatives of all K2P channel subfamilies and identified TREK-1 and TREK-2 as the only thermosensitive K2P channels, while TRAAK, the third member of the mechano-gated subfamily, showed no temperature dependence. We transferred the thermosensitivity of TREK-1 to TRAAK channels by exchanging the C-termini, demonstrating that the C-terminal domain is sufficient to confer thermosensitivity. By gradually truncating the C-terminus, we isolated a specific temperature responsive element (TRE) consisting of 18 amino acids that constitutes a unique feature in mammalian thermosensitive channels. Within this TRE lie both the binding domain for microtubule associated protein 2 (MAP2) and the PKA phosphorylation site. Pharmacological disruption of the microtubular network as well as the loss of the MAP2 binding site suppressed the temperature response, and PKA activation completely abolished temperature sensitivity. Thus, the connection to the microtubular network enables the thermosensitivity of TREK channels, which is not intrinsic to the channel itself, while the PKA-mediated phosphorylation status acts as a switch that determines if TREK channels are thermosensitive at all.
Collapse
Affiliation(s)
- Sönke Cordeiro
- Institute of Physiology, Kiel University, Kiel, Germany.
| | | |
Collapse
|
3
|
Zeng Z, Chen E, Xue J. Emerging roles of mechanically activated ion channels in autoimmune disease. Autoimmun Rev 2025; 24:103813. [PMID: 40194731 DOI: 10.1016/j.autrev.2025.103813] [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: 01/21/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
Mechanically activated (MA) ion channels have rapidly gained prominence as vital conduits bridging aberrant mechanical cues in tissues with the dysregulated immune responses at the core of autoimmune diseases. Once regarded as peripheral players in inflammation, these channels, exemplified by PIEZO1, TRPV4, and specific K2P family members, now play a central role in modulating T-cell effector functions, B- cell activation and the activity of macrophages and dendritic cells. Their gating is intimately tied to physical distortions such as increased tissue stiffness, osmotic imbalances, or fluid shear, triggering a cascade of ionic fluxes that elevate proinflammatory signaling and drive tissue-destructive loops. Recognition of these channels as central mediators of mechanical stress-induced inflammation responses in autoimmune pathogenesis is rapidly expanding. In parallel, the emerging therapeutic strategies aim to restrain overactive mechanosensors or selectively harness them in affected tissues. Small molecules, peptide blockers, and gene-targeting approaches show preclinical promise, although off-target effects and the broader homeostatic roles of these channels warrant caution. This review explores how integrating mechanobiological concepts with established immunological paradigms enables a more detailed understanding of autoimmune pathogenesis. By elucidating how mechanical forces potentiate or dampen pathological immunity, we propose innovative strategies that exploit mechanosensitivity to recalibrate immune responses across a spectrum of autoimmune conditions.
Collapse
Affiliation(s)
- Zhiru Zeng
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Engeng Chen
- Department of Zhejiang Provincial Key Laboratory of Biotherapy, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Jing Xue
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
4
|
Zhou Y. Focused Ultrasound Neuromodulation to Peripheral Nerve System. Eur J Neurosci 2025; 61:e70062. [PMID: 40170299 DOI: 10.1111/ejn.70062] [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: 11/24/2024] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 04/03/2025]
Abstract
Noninvasive focused ultrasound (FUS) has been applied in the treatment of various targets. Neuromodulation using FUS is emerging as a promising therapeutic modality for the central nerve system (CNS) with the advantages of deep penetration and precise targeting in the brain. This technique can also be applied to the peripheral nerve system (PNS). The principle of FUS and the mechanisms of neromodulation on PNS are summarized. Current experimental observations on the PNS targets are introduced to show their therapeutic effects. Discussion on the limitations and perspectives of this technology illustrates the pros and cons for future development. FUS provides a noninvasive, safe, and effective modality for neurotherapeutics. Although the relevant research on PNS is much less than that on CNS, the limited studies have already shown the satisfactory performance of FUS in comparison to the FDA-approved implanted device, especially the vagus nerve stimulation (VNS). Wide applications in clinics and fast development in technology are expected in the near future.
Collapse
Affiliation(s)
- Yufeng Zhou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
- National Medical Products Administration (NMPA) Key Laboratory for Quality Evaluation of Ultrasonic Surgical Equipment, Wuhan, Hubei, China
| |
Collapse
|
5
|
Jahn H, Shyng SL, Schultz C. Lipid probes to study ion channels. Curr Opin Chem Biol 2025; 85:102581. [PMID: 39978055 DOI: 10.1016/j.cbpa.2025.102581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/22/2025]
Abstract
Lipids can have specific interaction partners and act as small molecule regulators of proteins, especially for transmembrane proteins. Transmembrane proteins, such as ion channels, can be influenced by lipids in four ways; lipids can be direct ligands, localize effector proteins or domains, affect protein-protein interaction, or change the biophysical properties of the surrounding membrane. In this article, we will give examples of how lipids directly interact with ion channels and address the complex aspect of indirect regulation via lipids of the surrounding membrane bilayer. In addition, we discuss current and propose future molecular tools and experiments elucidating the many roles lipids play in ion channel function.
Collapse
Affiliation(s)
- Helene Jahn
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA; Department of Molecular Microbiology and Immunology, School of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Show-Ling Shyng
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA.
| | - Carsten Schultz
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
6
|
Sindoni M, Grandl J. A closed-loop system for millisecond readout and control of membrane tension. Biophys J 2025:S0006-3495(25)00199-7. [PMID: 40165372 DOI: 10.1016/j.bpj.2025.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
Characterizing the function of force-gated ion channels is essential for understanding their molecular mechanisms and how they are affected by disease-causing mutations, lipids, or small molecules. Pressure-clamp electrophysiology is a method that is established and widely used to characterize the mechanical sensitivity of force-gated ion channels. However, the physical stimulus many force-gated ion channels sense is not pressure but membrane tension. Here, we further develop the approach of combining patch-clamp electrophysiology with differential interference contrast microscopy into a system that controls membrane tension in real time. The system uses machine learning object detection for millisecond analysis of membrane curvature and control of pipette pressure to produce a closed-loop membrane tension clamp. The analysis of membrane tension is fully automated and includes the propagation of experimental errors, thereby increasing throughput and reducing bias. A dynamic control program clamps membrane tension with at least 93% accuracy and 0.3 mN/m precision. Additionally, the absence of tension drift enables averaging open probabilities of ion channels with low expression and/or unitary conductance over long durations. Using this system, we apply a tension step protocol and show that TMEM63A responds to tension with a tension of half-maximal activation of T50 = 5.5 ± 0.1 mN/m. Overall, this system allows for precise and efficient generation of tension-response relationships of force-gated ion channels.
Collapse
Affiliation(s)
- Michael Sindoni
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Jörg Grandl
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
7
|
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
|
8
|
Ma Q, Hernandez CC, Navratna V, Kumar A, Lee A, Mosalaganti S. Insights into the structure and modulation of human TWIK-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639014. [PMID: 40161613 PMCID: PMC11952367 DOI: 10.1101/2025.02.19.639014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The T andem of pore domain in a W eak I nward R ectifying K + channel 2 (TWIK-2; KCNK6) is a member of the Two-Pore Domain K + (K2P) channel family, which is associated with pulmonary hypertension, lung injury, and inflammation. The structure and regulatory mechanisms of TWIK-2 remain largely unknown. Here, we present the cryo-electron microscopy (cryo-EM) structure of human TWIK-2 at ~3.7 Å and highlight its conserved and unique features. Using automated whole-cell patch clamp recordings, we demonstrate that gating in TWIK-2 is voltage-dependent and insensitive to changes in the extracellular pH. We identify key residues that influence TWIK-2 activity by employing structure and sequence-guided site-directed mutagenesis and provide insights into the possible mechanism of TWIK-2 regulation. Additionally, we demonstrate the application of high-throughput automated whole-cell patch clamp platforms to screen small molecule modulators of TWIK-2. Our work serves as a foundation for designing high-throughput small molecule screening campaigns to identify specific high-affinity TWIK-2 modulators, including promising new anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Qianqian Ma
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Ciria C Hernandez
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Vikas Navratna
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Arvind Kumar
- Thermo Fisher Scientific, Waltham, Massachusetts, 02451, United States
| | - Abraham Lee
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Shyamal Mosalaganti
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, 48109, United States
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, United States
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, 48109, United States
- Department of Biophysics, College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan, 48109, United States
| |
Collapse
|
9
|
Zhu Y, Chen J, Chen C, Tang R, Xu J, Shi S, Yu X. Deciphering mechanical cues in the microenvironment: from non-malignant settings to tumor progression. Biomark Res 2025; 13:11. [PMID: 39849659 PMCID: PMC11755887 DOI: 10.1186/s40364-025-00727-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/05/2025] [Indexed: 01/25/2025] Open
Abstract
The tumor microenvironment functions as a dynamic and intricate ecosystem, comprising a diverse array of cellular and non-cellular components that precisely orchestrate pivotal tumor behaviors, including invasion, metastasis, and drug resistance. While unraveling the intricate interplay between the tumor microenvironment and tumor behaviors represents a tremendous challenge, recent research illuminates a crucial biological phenomenon known as cellular mechanotransduction. Within the microenvironment, mechanical cues like tensile stress, shear stress, and stiffness play a pivotal role by activating mechanosensitive effectors such as PIEZO proteins, integrins, and Yes-associated protein. This activation initiates cascades of intrinsic signaling pathways, effectively linking the physical properties of tissues to their physiological and pathophysiological processes like morphogenesis, regeneration, and immunity. This mechanistic insight offers a novel perspective on how the mechanical cues within the tumor microenvironment impact tumor behaviors. While the intricacies of the mechanical tumor microenvironment are yet to be fully elucidated, it exhibits distinct physical attributes from non-malignant tissues, including elevated solid stresses, interstitial hypertension, augmented matrix stiffness, and enhanced viscoelasticity. These traits exert notable influences on tumor progression and treatment responses, enriching our comprehension of the multifaceted nature of the microenvironment. Through this innovative review, we aim to provide a new lens to decipher the mechanical attributes within the tumor microenvironment from non-malignant contexts, broadening our knowledge on how these factors promote or inhibit tumor behaviors, and thus offering valuable insights to identify potential targets for anti-tumor strategies.
Collapse
Affiliation(s)
- Yicheng Zhu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jiaoshun Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chen Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
10
|
Ambattu LA, Del Rosal B, Conn CE, Yeo LY. High-frequency MHz-order vibration enables cell membrane remodeling and lipid microdomain manipulation. Biophys J 2025; 124:25-39. [PMID: 39415451 PMCID: PMC11739889 DOI: 10.1016/j.bpj.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/29/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024] Open
Abstract
We elucidate the mechanism underpinning a recently discovered phenomenon in which cells respond to MHz-order mechanostimuli. Deformations induced along the plasma membrane under these external mechanical cues are observed to decrease the membrane tension, which, in turn, drives transient and reversible remodeling of its lipid structure. In particular, the increase and consequent coalescence of ordered lipid microdomains leads to closer proximity to mechanosensitive ion channels-Piezo1, in particular-that, due to crowding, results in their activation to mobilize influx of calcium (Ca2+) ions into the cell. It is the modulation of this second messenger that is responsible for the downstream signaling and cell fates that ensue. In addition, we show that such spatiotemporal control over the membrane microdomains in cells-without necessitating biochemical factors-facilitates aggregation and association of intrinsically disordered tau proteins in neuroblastoma cells, and their transformation to pathological conditions implicated in neurodegenerative diseases, thereby paving the way for the development of therapeutic intervention strategies.
Collapse
Affiliation(s)
- Lizebona A Ambattu
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne, VIC, Australia
| | | | | | - Leslie Y Yeo
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
11
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
12
|
Duan X, Liu R, Xi Y, Tian Z. The mechanisms of exercise improving cardiovascular function by stimulating Piezo1 and TRP ion channels: a systemic review. Mol Cell Biochem 2025; 480:119-137. [PMID: 38625513 DOI: 10.1007/s11010-024-05000-5] [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: 02/08/2024] [Accepted: 03/24/2024] [Indexed: 04/17/2024]
Abstract
Mechanosensitive ion channels are widely distributed in the heart, lung, bladder and other tissues, and plays an important role in exercise-induced cardiovascular function promotion. By reviewing the PubMed databases, the results were summarized using the terms "Exercise/Sport", "Piezo1", "Transient receptor potential (TRP)" and "Cardiovascular" as the keywords, 124-related papers screened were sorted and reviewed. The results showed that: (1) Piezo1 and TRP channels play an important role in regulating blood pressure and the development of cardiovascular diseases such as atherosclerosis, myocardial infarction, and cardiac fibrosis; (2) Exercise promotes cardiac health, inhibits the development of pathological heart to heart failure, regulating the changes in the characterization of Piezo1 and TRP channels; (3) Piezo1 activates downstream signaling pathways with very broad pathways, such as AKT/eNOS, NF-κB, p38MAPK and HIPPO-YAP signaling pathways. Piezo1 and Irisin regulate nuclear localization of YAP and are hypothesized to act synergistically to regulate tissue mechanical properties of the cardiovascular system and (4) The cardioprotective effects of exercise through the TRP family are mostly accomplished through Ca2+ and involve many signaling pathways. TRP channels exert their important cardioprotective effects by reducing the TRPC3-Nox2 complex and mediating Irisin-induced Ca2+ influx through TRPV4. It is proposed that exercise stimulates the mechanosensitive cation channel Piezo1 and TRP channels, which exerts cardioprotective effects. The activation of Piezo1 and TRP channels and their downstream targets to exert cardioprotective function by exercise may provide a theoretical basis for the prevention of cardiovascular diseases and the rehabilitation of clinical patients.
Collapse
Affiliation(s)
- Xinyan Duan
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Renhan Liu
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Yue Xi
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an, 710119, China.
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an, 710119, China
| |
Collapse
|
13
|
del Rosario-Gilabert D, Valenzuela-Miralles A, Esquiva G. Advances in mechanotransduction and sonobiology: effects of audible acoustic waves and low-vibration stimulations on mammalian cells. Biophys Rev 2024; 16:783-812. [PMID: 39830129 PMCID: PMC11735818 DOI: 10.1007/s12551-024-01242-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/25/2024] [Indexed: 01/22/2025] Open
Abstract
In recent decades, research on mechanotransduction has advanced considerably, focusing on the effects of audible acoustic waves (AAWs) and low-vibration stimulation (LVS), which has propelled the field of sonobiology forward. Taken together, the current evidence demonstrates the influence of these biosignals on key cellular processes, such as growth, differentiation and migration in mammalian cells, emphasizing the determining role of specific physical parameters during stimulation, such as frequency, sound pressure level/amplitude and exposure time. These mechanical waves interact with various cellular elements, including ion channels, primary cilia, cell-cell adhesion receptors, cell-matrix and extracellular matrix proteins, and focal adhesion complexes. These components connect with the cytoskeletal fibre network, enabling the transmission of mechanical stimuli towards the nucleus. The nucleus, in turn, linked to the cytoskeleton via the linkers of the nucleoskeleton and cytoskeleton complex, acts as a mechanosensitive centre, not only responding to changes in cytoskeletal stiffness and nuclear tension but also regulating gene expression through the transcriptional co-activator YAP/TAZ and interactions between chromatin and the nuclear envelope. This intricate chain of mechanisms highlights the potential of sonobiology in various fields, including dentistry, regenerative medicine, tissue engineering and cancer research. However, progress in these fields requires the establishment of standardized measurement methodologies and biocompatible experimental setups to ensure the reproducibility of results.
Collapse
Affiliation(s)
- D. del Rosario-Gilabert
- Department of Optics, Pharmacology and Anatomy, University of Alicante, San Vicente del Raspeig, Spain
- Department of Physics, Systems Engineering and Signal Theory, University of Alicante, San Vicente del Raspeig, Spain
- Department of Computer Technology, University of Alicante, San Vicente del Raspeig, Spain
- Institute for Advanced Neuroscience of Barcelona (INAB), Barcelona, Spain
| | - A. Valenzuela-Miralles
- Department of Optics, Pharmacology and Anatomy, University of Alicante, San Vicente del Raspeig, Spain
| | - G. Esquiva
- Department of Optics, Pharmacology and Anatomy, University of Alicante, San Vicente del Raspeig, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| |
Collapse
|
14
|
Roeterink RMA, Casadevall I Solvas X, Collins DJ, Scott DJ. Force versus Response: Methods for Activating and Characterizing Mechanosensitive Ion Channels and GPCRs. Adv Healthc Mater 2024; 13:e2402167. [PMID: 39402780 DOI: 10.1002/adhm.202402167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/20/2024] [Indexed: 12/18/2024]
Abstract
Mechanotransduction is the process whereby cells convert mechanical signals into electrochemical responses, where mechanosensitive proteins mediate this interaction. To characterize these critical proteins, numerous techniques have been developed that apply forces and measure the subsequent cellular responses. While these approaches have given insight into specific aspects of many such proteins, subsequent validation and cross-comparison between techniques remain difficult given significant variations in reported activation thresholds and responses for the same protein across different studies. Accurately determining mechanosensitivity responses for various proteins, however, is essential for understanding mechanotransduction and potential physiological implications, including therapeutics. This critical review provides an assessment of current and emerging approaches used for mechanosensitive ion channel and G-Coupled Receptors (GPCRs) stimulation and measurement, with a specific focus on the ability to quantitatively measure mechanosensitive responses.
Collapse
Affiliation(s)
- Renate M A Roeterink
- Department of Biomedical Engineering, The University of Melbourne, VIC, Parkville, Victoria, 3010, Australia
- Department of Biosystems - MeBioS, KU Leuven, Willem de Croylaan 42, Leuven, 3001, Belgium
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
| | | | - David J Collins
- Department of Biomedical Engineering, The University of Melbourne, VIC, Parkville, Victoria, 3010, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Daniel J Scott
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
15
|
Liu G, Dong BB, Devanarayana S, Chen RC, Liu Q. Emerging roles of mechanosensitive ion channels in ventilator induced lung injury: a systematic review. Front Immunol 2024; 15:1479230. [PMID: 39664395 PMCID: PMC11631737 DOI: 10.3389/fimmu.2024.1479230] [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: 08/11/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
Background The pathogenetic mechanisms of ventilator-induced lung injury (VILI) still need to be elucidated. The mechanical forces during mechanical ventilation are continually sensed and transmitted by mechanosensitive ion channels (MSICs) in pulmonary endothelial, epithelial, and immune cells. In recent years, MSICs have been shown to be involved in VILI. Methods A systematic search across PubMed, the Cochrane Library, Web of Science, and ScienceDirect was performed from inception to March 2024, and the review was conducted in accordance with PRISMA guidelines. The potential eligible studies were evaluated by two authors independently. Study characteristics, quality assessment, and potential mechanisms were analyzed. Results We included 23 eligible studies, most of which were performed with murine animals in vivo. At the in vitro level, 52% and 48% of the experiments were conducted with human or animal cells, respectively. No clinical studies were found. The most reported MSICs include Piezo channels, transient receptor potential channels, potassium channels, and stretch-activated sodium channels. Piezo1 has been the most concerned channel in the recent five years. This study found that signal pathways, such as RhoA/ROCK1, could be enhanced by cyclic stretch-activated MSICs, which contribute to VILI through dysregulated inflammation and immune responses mediated by ion transport. The review indicates the emerging role of MSICs in the pathogenesis of VILI, especially as a signal-transmitting link between mechanical stretch and pathogenesis such as inflammation, disruption of cell junctions, and edema formation. Conclusions Mechanical stretch stimulates MSICs to increase transcellular ion exchange and subsequently generates VILI through inflammation and other pathogeneses mediated by MSICs signal-transmitting pathways. These findings make it possible to identify potential therapeutic targets for the prevention of lung injury through further exploration and more studies. Systematic review registration https://inplasy.com/inplasy-2024-10-0115/, identifier INPLASY2024100115.
Collapse
Affiliation(s)
- Gang Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin-bin Dong
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shalika Devanarayana
- School of International Education, Zhengzhou University, Zhengzhou, Henan, China
| | - Rong-Chang Chen
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Shenzhen Institute of Respiratory Diseases, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Qi Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
16
|
Matsuki Y, Iwamoto M, Maki T, Takashima M, Yoshida T, Oiki S. Programmable Lipid Bilayer Tension-Control Apparatus for Quantitative Mechanobiology. ACS NANO 2024; 18:30561-30573. [PMID: 39437160 PMCID: PMC11544928 DOI: 10.1021/acsnano.4c09017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
The biological membrane is not just a platform for information processing but also a field of mechanics. The lipid bilayer that constitutes the membrane is an elastic body, generating stress upon deformation, while the membrane protein embedded therein deforms the bilayer through structural changes. Among membrane-protein interplays, various channel species act as tension-current converters for signal transduction, serving as elementary processes in mechanobiology. However, in situ studies in chaotically complex cell membranes are challenging, and characterizing the tension dependency of mechanosensitive channels remains semiquantitative owing to technical limitations. Here, we developed a programmable membrane tension-control apparatus on a lipid bilayer system. This synthetic membrane system [contact bubble bilayer (CBB)] uses pressure to drive bilayer tension changes via the Young-Laplace principle, whereas absolute bilayer tension is monitored in real-time through image analysis of the bubble geometry via the Young principle. Consequently, the mechanical nature of the system permits the implementation of closed-loop feedback control of bilayer tension (tension-clamp CBB), maintaining a constant tension for minutes and allowing stepwise tension changes within a hundred milliseconds in the tension range of 0.8 to 15 mN·m-1. We verified the system performance by examining the single-channel behavior of tension-dependent KcsA and TREK-1 potassium channels under scheduled tension time courses prescribed via visual interfaces. The result revealed steady-state activity and dynamic responses to the step tension changes, which are essential to the biophysical characterization of the channels. The apparatus explores a frontier for quantitative mechanobiology studies and promotes the development of a tension-operating experimental robot.
Collapse
Affiliation(s)
- Yuka Matsuki
- Department
of Anesthesiology and Reanimatology, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
- Life
Science Innovation Center, University of
Fukui, Fukui 910-8507, Japan
| | - Masayuki Iwamoto
- Department
of Molecular Neuroscience, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
- Life
Science Innovation Center, University of
Fukui, Fukui 910-8507, Japan
| | - Takahisa Maki
- Department
of Molecular Neuroscience, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
- Life
Science Innovation Center, University of
Fukui, Fukui 910-8507, Japan
| | - Masako Takashima
- Department
of Molecular Neuroscience, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Toshiyuki Yoshida
- Department
of Information Science, Faculty of Engineering, University of Fukui, Fukui 910-8507, Japan
| | - Shigetoshi Oiki
- Biomedical
Imaging Research Center, University of Fukui, Fukui 910-1193, Japan
| |
Collapse
|
17
|
Xiao B. Mechanisms of mechanotransduction and physiological roles of PIEZO channels. Nat Rev Mol Cell Biol 2024; 25:886-903. [PMID: 39251883 DOI: 10.1038/s41580-024-00773-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/11/2024]
Abstract
Mechanical force is an essential physical element that contributes to the formation and function of life. The discovery of the evolutionarily conserved PIEZO family, including PIEZO1 and PIEZO2 in mammals, as bona fide mechanically activated cation channels has transformed our understanding of how mechanical forces are sensed and transduced into biological activities. In this Review, I discuss recent structure-function studies that have illustrated how PIEZO1 and PIEZO2 adopt their unique structural design and curvature-based gating dynamics, enabling their function as dedicated mechanotransduction channels with high mechanosensitivity and selective cation conductivity. I also discuss our current understanding of the physiological and pathophysiological roles mediated by PIEZO channels, including PIEZO1-dependent regulation of development and functional homeostasis and PIEZO2-dominated mechanosensation of touch, tactile pain, proprioception and interoception of mechanical states of internal organs. Despite the remarkable progress in PIEZO research, this Review also highlights outstanding questions in the field.
Collapse
Affiliation(s)
- Bailong Xiao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
- Beijing Frontier Research Center of Biological Structure, Tsinghua University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
18
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
19
|
Poudel B, Vanegas JM. Structural Rearrangement of the AT1 Receptor Modulated by Membrane Thickness and Tension. J Phys Chem B 2024; 128:9470-9481. [PMID: 39298653 DOI: 10.1021/acs.jpcb.4c03325] [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: 09/22/2024]
Abstract
Membrane-embedded mechanosensitive (MS) proteins, including ion channels and G-protein coupled receptors (GPCRs), are essential for the transduction of external mechanical stimuli into biological signals. The angiotensin II type 1 (AT1) receptor plays many important roles in cardiovascular regulation and is associated with diseases such as hypertension and congestive heart failure. The membrane-mediated activation of the AT1 receptor is not well understood, despite this being one of the most widely studied GPCRs within the context of biased agonism. Here, we use extensive molecular dynamics (MD) simulations to characterize the effect of the local membrane environment on the activation of the AT1 receptor. We show that membrane thickness plays an important role in the stability of active and inactive states of the receptor, as well as the dynamic interchange between states. Furthermore, our simulation results show that membrane tension is effective in driving large-scale structural changes in the inactive state such as the outward movement of transmembrane helix 6 to stabilize intermediate active-like conformations. We conclude by comparing our simulation observations with AlphaFold 2 predictions, as a proxy to experimental structures, to provide a framework for how membrane mediated stimuli can facilitate activation of the AT1 receptor through the β-arrestin signaling pathway.
Collapse
Affiliation(s)
- Bharat Poudel
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon 97331, United States
| | - Juan M Vanegas
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon 97331, United States
| |
Collapse
|
20
|
Csáki R, Nagaraj C, Almássy J, Khozeimeh MA, Jeremic D, Olschewski H, Dobolyi A, Hoetzenecker K, Olschewski A, Enyedi P, Lengyel M. The TREK-1 potassium channel is a potential pharmacological target for vasorelaxation in pulmonary hypertension. Br J Pharmacol 2024; 181:3576-3593. [PMID: 38807478 DOI: 10.1111/bph.16426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND AND PURPOSE Pulmonary arterial hypertension (PAH) is a progressive disease in which chronic membrane potential (Em) depolarisation of the pulmonary arterial smooth muscle cells (PASMCs) causes calcium overload, a key pathological alteration. Under resting conditions, the negative Em is mainly set by two pore domain potassium (K2P) channels, of which the TASK-1 has been extensively investigated. EXPERIMENTAL APPROACH Ion channel currents and membrane potential of primary cultured human(h) PASMCs were measured using the voltage- and current clamp methods. Intracellular [Ca2+] was monitored using fluorescent microscopy. Pulmonary BP and vascular tone measurements were also performed ex vivo using a rat PAH model. KEY RESULTS TREK-1 was the most abundantly expressed K2P in hPASMCs of healthy donors and idiopathic(I) PAH patients. Background K+-current was similar in hPASMCs for both groups and significantly enhanced by the TREK activator ML-335. In donor hPASMCs, siRNA silencing or pharmacological inhibition of TREK-1 caused depolarisation, reminiscent of the electrophysiological phenotype of idiopathic PAH. ML-335 hyperpolarised donor hPASMCs and normalised the Em of IPAH hPASMCs. A close link was found between TREK-1 activity and intracellular Ca2+-signalling using a channel activator, ML-335, and an inhibitor, spadin. In the rat, ML-335 relaxed isolated pre-constricted pulmonary arteries and significantly decreased pulmonary arterial pressure in the isolated perfused lung. CONCLUSIONS AND IMPLICATIONS These data suggest that TREK-1is a key factor in Em setting and Ca2+ homeostasis of hPASMC, and therefore, essential for maintenance of a low resting pulmonary vascular tone. Thus TREK-1 may represent a new therapeutic target for PAH.
Collapse
MESH Headings
- Potassium Channels, Tandem Pore Domain/antagonists & inhibitors
- Potassium Channels, Tandem Pore Domain/metabolism
- Animals
- Humans
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Male
- Rats
- Vasodilation/drug effects
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Cells, Cultured
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Female
- Rats, Sprague-Dawley
- Membrane Potentials/drug effects
- Rats, Wistar
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Calcium/metabolism
- Middle Aged
Collapse
Affiliation(s)
- Réka Csáki
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Chandran Nagaraj
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - János Almássy
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | | | - Dusan Jeremic
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Horst Olschewski
- Department of Internal Medicine, Division of Pulmonology, Medical University of Graz, Graz, Austria
| | - Alice Dobolyi
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Andrea Olschewski
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Péter Enyedi
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Miklós Lengyel
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
21
|
Garcia-Sanchez J, Lin D, Liu WW. Mechanosensitive ion channels in glaucoma pathophysiology. Vision Res 2024; 223:108473. [PMID: 39180975 PMCID: PMC11398070 DOI: 10.1016/j.visres.2024.108473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/27/2024]
Abstract
Force sensing is a fundamental ability that allows cells and organisms to interact with their physical environment. The eye is constantly subjected to mechanical forces such as blinking and eye movements. Furthermore, elevated intraocular pressure (IOP) can cause mechanical strain at the optic nerve head, resulting in retinal ganglion cell death (RGC) in glaucoma. How mechanical stimuli are sensed and affect cellular physiology in the eye is unclear. Recent studies have shown that mechanosensitive ion channels are expressed in many ocular tissues relevant to glaucoma and may influence IOP regulation and RGC survival. Furthermore, variants in mechanosensitive ion channel genes may be associated with risk for primary open angle glaucoma. These findings suggest that mechanosensitive channels may be important mechanosensors mediating cellular responses to pressure signals in the eye. In this review, we focus on mechanosensitive ion channels from three major channel families-PIEZO, two-pore potassium and transient receptor potential channels. We review the key properties of these channels, their effects on cell function and physiology, and discuss their possible roles in glaucoma pathophysiology.
Collapse
Affiliation(s)
- Julian Garcia-Sanchez
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Danting Lin
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wendy W Liu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
22
|
Hamed YMF, Ghosh B, Marshall KL. PIEZO ion channels: force sensors of the interoceptive nervous system. J Physiol 2024; 602:4777-4788. [PMID: 38456626 PMCID: PMC11845038 DOI: 10.1113/jp284077] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/08/2024] [Indexed: 03/09/2024] Open
Abstract
Many organs are designed to move: the heart pumps each second, the gastrointestinal tract squeezes and churns to digest food, and we contract and relax skeletal muscles to move our bodies. Sensory neurons of the peripheral nervous system detect signals from bodily tissues, including the forces generated by these movements, to control physiology. The processing of these internal signals is called interoception, but this is a broad term that includes a wide variety of both chemical and mechanical sensory processes. Mechanical senses are understudied, but rapid progress has been made in the last decade, thanks in part to the discovery of the mechanosensory PIEZO ion channels (Coste et al., 2010). The role of these mechanosensors within the interoceptive nervous system is the focus of this review. In defining the transduction molecules that govern mechanical interoception, we will have a better grasp of how these signals drive physiology.
Collapse
Affiliation(s)
- Yasmeen M. F. Hamed
- Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Britya Ghosh
- Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kara L. Marshall
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77030, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Lead contact
| |
Collapse
|
23
|
Sherman J, Bortz E, Antonio ES, Tseng HA, Raiff L, Han X. Ultrasound pulse repetition frequency preferentially activates different neuron populations independent of cell type. J Neural Eng 2024; 21:056008. [PMID: 39178904 PMCID: PMC11381926 DOI: 10.1088/1741-2552/ad731c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/18/2024] [Accepted: 08/23/2024] [Indexed: 08/26/2024]
Abstract
Objective. Transcranial ultrasound (US) stimulation serves as an external input to a neuron, and thus the evoked response relies on neurons' intrinsic properties. Neural activity is limited to a couple hundred hertz and often exhibits preference to input frequencies. Accordingly, US pulsed at specific physiologic pulse repetition frequencies (PRFs) may selectively engage neurons with the corresponding input frequency preference. However, most US parametric studies examine the effects of supraphysiologic PRFs. It remains unclear whether pulsing US at different physiologic PRFs could activate distinct neurons in the awake mammalian brain.Approach. We recorded cellular calcium responses of individual motor cortex neurons to US pulsed at PRFs of 10, 40, and 140 Hz in awake mice. We compared the evoked responses across these PRFs in the same neurons. To further understand the cell-type dependent effects, we categorized the recorded neurons as parvalbumin positive fast spiking interneurons or putative excitatory neurons and analyzed single-cell mechanosensitive channel expression in mice and humans using the Allen Brain Institute's RNA-sequencing databases.Main results. We discovered that many neurons were preferentially activated by only one PRF and different PRFs selectively engaged distinct neuronal populations. US-evoked cellular calcium responses exhibited the same characteristics as those naturally occurring during spiking, suggesting that US increases intrinsic neuronal activity. Furthermore, evoked responses were similar between fast-spiking inhibitory neurons and putative excitatory neurons. Thus, variation in individual neuron's cellular properties dominates US-evoked response heterogeneity, consistent with our observed cell-type independent expression patterns of mechanosensitive channels across individual neurons in mice and humans. Finally, US transiently increased network synchrony without producing prolonged over-synchronization that could be detrimental to neural circuit functions.Significance. These results highlight the feasibility of activating distinct neuronal subgroups by varying PRF and the potential to improve neuromodulation effects by combining physiologic PRFs.
Collapse
Affiliation(s)
- Jack Sherman
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- Department of Pharmacology and Experimental Therapeutics, Boston University, Boston, MA, United States of America
| | - Emma Bortz
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| | - Erynne San Antonio
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| | - Hua-an Tseng
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| | - Laura Raiff
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| |
Collapse
|
24
|
Astashkin AV, Gyawali YP, Jiang T, Zhang H, Feng C. Analyzing the FMN-heme interdomain docking interactions in neuronal and inducible NOS isoforms by pulsed EPR experiments and conformational distribution modeling. J Biol Inorg Chem 2024; 29:611-623. [PMID: 39136772 PMCID: PMC11390318 DOI: 10.1007/s00775-024-02068-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024]
Abstract
Nitric oxide synthases (NOSs), a family of flavo-hemoproteins with relatively rigid domains linked by flexible regions, require optimal FMN domain docking to the heme domain for efficient interdomain electron transfer (IET). To probe the FMN-heme interdomain docking, the magnetic dipole interactions between the FMN semiquinone radical (FMNH•) and the low-spin ferric heme centers in oxygenase/FMN (oxyFMN) constructs of neuronal and inducible NOS (nNOS and iNOS, respectively) were measured using the relaxation-induced dipolar modulation enhancement (RIDME) technique. The FMNH• RIDME data were analyzed using the mesoscale Monte Carlo calculations of conformational distributions of NOS, which were improved to account for the native degrees of freedom of the amino acid residues constituting the flexible interdomain tethers. This combined computational and experimental analysis allowed for the estimation of the stabilization energies and populations of the docking complexes of calmodulin (CaM) and the FMN domain with the heme domain. Moreover, combining the five-pulse and scaled four-pulse RIDME data into a single trace has significantly reduced the uncertainty in the estimated docking probabilities. The obtained FMN-heme domain docking energies for nNOS and iNOS were similar (-3.8 kcal/mol), in agreement with the high degree of conservation of the FMN-heme domain docking interface between the NOS isoforms. In spite of the similar energetics, the FMN-heme domain docking probabilities in nNOS and iNOS oxyFMN were noticeably different (~ 0.19 and 0.23, respectively), likely due to differences in the lengths of the FMN-heme interdomain tethers and the docking interface topographies. The analysis based on the IET theory and RIDME experiments indicates that the variations in conformational dynamics may account for half of the difference in the FMN-heme IET rates between the two NOS isoforms.
Collapse
Affiliation(s)
- Andrei V Astashkin
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, 85721, USA
| | | | - Ting Jiang
- College of Pharmacy, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Haikun Zhang
- College of Pharmacy, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Changjian Feng
- College of Pharmacy, University of New Mexico, Albuquerque, NM, 87131, USA.
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
25
|
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
|
26
|
Cox CD, Poole K, Martinac B. Re-evaluating TRP channel mechanosensitivity. Trends Biochem Sci 2024; 49:693-702. [PMID: 38851904 DOI: 10.1016/j.tibs.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 06/10/2024]
Abstract
Transient receptor potential (TRP) channels are implicated in a wide array of mechanotransduction processes. However, a question remains whether TRP channels directly sense mechanical force, thus acting as primary mechanotransducers. We use several recent examples to demonstrate the difficulty in definitively ascribing mechanosensitivity to TRP channel subfamilies. Ultimately, despite being implicated in an ever-growing list of mechanosignalling events in most cases limited robust or reproducible evidence supports the contention that TRP channels act as primary transducers of mechanical forces. They either (i) possess unique and as yet unspecified structural or local requirements for mechanosensitivity; or (ii) act as mechanoamplifiers responding downstream of the activation of a primary mechanotransducer that could include Ca2+-permeable mechanosensitive (MS) channels or other potentially unidentified mechanosensors.
Collapse
Affiliation(s)
- Charles D Cox
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, 2052, Australia; Victor Chang Cardiac Research Institute, Sydney, Darlinghurst, NSW, 2010, Australia
| | - Kate Poole
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, 2052, Australia.
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Sydney, Darlinghurst, NSW, 2010, Australia.
| |
Collapse
|
27
|
Bechard E, Arel E, Bride J, Louradour J, Bussy X, Elloumi A, Vigor C, Soule P, Oger C, Galano JM, Durand T, Le Guennec JY, Moha-Ou-Maati H, Demion M. Activation of hTREK-1 by polyunsaturated fatty acids involves direct interaction. Sci Rep 2024; 14:15244. [PMID: 38956407 PMCID: PMC11220079 DOI: 10.1038/s41598-024-66192-w] [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: 12/21/2023] [Accepted: 06/28/2024] [Indexed: 07/04/2024] Open
Abstract
TREK-1 is a mechanosensitive channel activated by polyunsaturated fatty acids (PUFAs). Its activation is supposed to be linked to changes in membrane tension following PUFAs insertion. Here, we compared the effect of 11 fatty acids and ML402 on TREK-1 channel activation using the whole cell and the inside-out configurations of the patch-clamp technique. Firstly, TREK-1 activation by PUFAs is variable and related to the variable constitutive activity of TREK-1. We observed no correlation between TREK-1 activation and acyl chain length or number of double bonds suggesting that the bilayer-couple hypothesis cannot explain by itself the activation of TREK-1 by PUFAs. The membrane fluidity measurement is not modified by PUFAs at 10 µM. The spectral shift analysis in TREK-1-enriched microsomes indicates a KD,TREK1 at 44 µM of C22:6 n-3. PUFAs display the same activation and reversible kinetics than the direct activator ML402 and activate TREK-1 in both whole-cell and inside-out configurations of patch-clamp suggesting that the binding site of PUFAs is accessible from both sides of the membrane, as for ML402. Finally, we proposed a two steps mechanism: first, insertion into the membrane, with no fluidity or curvature modifications at 10 µM, and then interaction with TREK-1 channel to open it.
Collapse
Affiliation(s)
- Emilie Bechard
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Elodie Arel
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Jamie Bride
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Julien Louradour
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Xavier Bussy
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Anis Elloumi
- IBMM, Université de Montpellier, UMR CNRS 5247, ENSCM, Montpellier, France
| | - Claire Vigor
- IBMM, Université de Montpellier, UMR CNRS 5247, ENSCM, Montpellier, France
| | | | - Camille Oger
- IBMM, Université de Montpellier, UMR CNRS 5247, ENSCM, Montpellier, France
| | - Jean-Marie Galano
- IBMM, Université de Montpellier, UMR CNRS 5247, ENSCM, Montpellier, France
| | - Thierry Durand
- IBMM, Université de Montpellier, UMR CNRS 5247, ENSCM, Montpellier, France
| | - Jean-Yves Le Guennec
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Hamid Moha-Ou-Maati
- IGF, Université de Montpellier, UMR CNRS 5203, Inserm 1191, Montpellier, France
- INM, Inserm U1298, Montpellier, France
| | - Marie Demion
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France.
| |
Collapse
|
28
|
Li X, Zhang G, Zheng C, Tian Y. Enhanced Transient Response of Ultrasound Neuromodulation Mediated by Large Conductance Mechanosensitive Ion Channel. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40040126 DOI: 10.1109/embc53108.2024.10782025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Ultrasound neuromodulation, a burgeoning field aimed at enhancing brain function and behavior, relies on the intricate electrophysiological responses of neurons driven by ion channel dynamics. However, achieving targeted and precise modulation with ultrasound poses a significant challenge. A hypothesis is that mechanosensitive (MS) ion channels may play a pivotal role in achieving the desired specificity in ultrasound effects. Our study involved augmenting MscL-G22S channel expression in specific areas of rats' hippocampus (HP) and primary visual cortex (V1) via AAV virus delivery. We recorded local field potentials (LFPs) to assess the modulation effect and identified distinct evoked-related potentials (ERPs) in regions overexpressing MscL-G22S channels under anesthesia. Notable variations in the amplitude of negative peaks were observed in the ERPs between HP and V1, highlighting a nuanced response to ultrasound stimulation. These results highlight the complexity and promise of MS channel-mediated neuromodulation, offering critical insights into the development of targeted ultrasound interventions to improve brain function.
Collapse
|
29
|
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
|
30
|
Pérez-Mitta G, Sezgin Y, Wang W, MacKinnon R. Freestanding bilayer microscope for single-molecule imaging of membrane proteins. SCIENCE ADVANCES 2024; 10:eado4722. [PMID: 38905330 PMCID: PMC11192074 DOI: 10.1126/sciadv.ado4722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/16/2024] [Indexed: 06/23/2024]
Abstract
Integral membrane proteins (IMPs) constitute a large fraction of organismal proteomes, playing fundamental roles in physiology and disease. Despite their importance, the mechanisms underlying dynamic features of IMPs, such as anomalous diffusion, protein-protein interactions, and protein clustering, remain largely unknown due to the high complexity of cell membrane environments. Available methods for in vitro studies are insufficient to study IMP dynamics systematically. This publication introduces the freestanding bilayer microscope (FBM), which combines the advantages of freestanding bilayers with single-particle tracking. The FBM, based on planar lipid bilayers, enables the study of IMP dynamics with single-molecule resolution and unconstrained diffusion. This paper benchmarks the FBM against total internal reflection fluorescence imaging on supported bilayers and is used here to estimate ion channel open probability and to examine the diffusion behavior of an ion channel in phase-separated bilayers. The FBM emerges as a powerful tool to examine membrane protein/lipid organization and dynamics to understand cell membrane processes.
Collapse
Affiliation(s)
- Gonzalo Pérez-Mitta
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Yeliz Sezgin
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | | | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| |
Collapse
|
31
|
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
|
32
|
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: 14] [Impact Index Per Article: 14.0] [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
|
33
|
Jojoa-Cruz S, Dubin AE, Lee WH, Ward AB. Structure-guided mutagenesis of OSCAs reveals differential activation to mechanical stimuli. eLife 2024; 12:RP93147. [PMID: 38592763 PMCID: PMC11003742 DOI: 10.7554/elife.93147] [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: 04/10/2024] Open
Abstract
The dimeric two-pore OSCA/TMEM63 family has recently been identified as mechanically activated ion channels. Previously, based on the unique features of the structure of OSCA1.2, we postulated the potential involvement of several structural elements in sensing membrane tension (Jojoa-Cruz et al., 2018). Interestingly, while OSCA1, 2, and 3 clades are activated by membrane stretch in cell-attached patches (i.e. they are stretch-activated channels), they differ in their ability to transduce membrane deformation induced by a blunt probe (poking). Here, in an effort to understand the domains contributing to mechanical signal transduction, we used cryo-electron microscopy to solve the structure of Arabidopsis thaliana (At) OSCA3.1, which, unlike AtOSCA1.2, only produced stretch- but not poke-activated currents in our initial characterization (Murthy et al., 2018). Mutagenesis and electrophysiological assessment of conserved and divergent putative mechanosensitive features of OSCA1.2 reveal a selective disruption of the macroscopic currents elicited by poking without considerable effects on stretch-activated currents (SAC). Our results support the involvement of the amphipathic helix and lipid-interacting residues in the membrane fenestration in the response to poking. Our findings position these two structural elements as potential sources of functional diversity within the family.
Collapse
Affiliation(s)
- Sebastian Jojoa-Cruz
- Department of Integrative Structural and Computational Biology, Scripps ResearchLa JollaUnited States
| | - Adrienne E Dubin
- Department of Integrative Structural and Computational Biology, Scripps ResearchLa JollaUnited States
- Department of Neuroscience, Scripps ResearchLa JollaUnited States
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, Scripps ResearchLa JollaUnited States
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, Scripps ResearchLa JollaUnited States
| |
Collapse
|
34
|
Zhang M, Tang S, Wang X, Fang S, Li Y. Mechanosensitive channel MscL gating transitions coupling with constriction point shift. Protein Sci 2024; 33:e4965. [PMID: 38501596 PMCID: PMC10949393 DOI: 10.1002/pro.4965] [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: 10/25/2023] [Revised: 02/23/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
The mechanosensitive channel of large conductance (MscL) acts as an "emergency release valve" that protects bacterial cells from acute hypoosmotic stress, and it serves as a paradigm for studying the mechanism underlying the transduction of mechanical forces. MscL gating is proposed to initiate with an expansion without opening, followed by subsequent pore opening via a number of intermediate substates, and ends in a full opening. However, the details of gating process are still largely unknown. Using in vivo viability assay, single channel patch clamp recording, cysteine cross-linking, and tryptophan fluorescence quenching approach, we identified and characterized MscL mutants with different occupancies of constriction region in the pore domain. The results demonstrated the shifts of constriction point along the gating pathway towards cytoplasic side from residue G26, though G22, to L19 upon gating, indicating the closed-expanded transitions coupling of the expansion of tightly packed hydrophobic constriction region to conduct the initial ion permeation in response to the membrane tension. Furthermore, these transitions were regulated by the hydrophobic and lipidic interaction with the constricting "hot spots". Our data reveal a new resolution of the transitions from the closed to the opening substate of MscL, providing insights into the gating mechanisms of MscL.
Collapse
Affiliation(s)
- Mingfeng Zhang
- Department of Cell Biology, College of MedicineJiaxing UniversityJiaxingChina
- School of Life ScienceWestlake UniversityHangzhouChina
- School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Siyang Tang
- School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Xiaomin Wang
- Department of Cell Biology, College of MedicineJiaxing UniversityJiaxingChina
| | - Sanhua Fang
- Core FacilitiesZhejiang University School of MedicineHangzhouChina
| | - Yuezhou Li
- Department of Cell Biology, College of MedicineJiaxing UniversityJiaxingChina
| |
Collapse
|
35
|
Sherman J, Bortz E, Antonio ES, Tseng HA, Raiff L, Han X. Ultrasound pulse repetition frequency preferentially activates different neuron populations independent of cell type. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.25.586645. [PMID: 38585918 PMCID: PMC10996595 DOI: 10.1101/2024.03.25.586645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Transcranial ultrasound activates mechanosensitive cellular signaling and modulates neural dynamics. Given that intrinsic neuronal activity is limited to a couple hundred hertz and often exhibits frequency preference, we examined whether pulsing ultrasound at physiologic pulse repetition frequencies (PRFs) could selectively influence neuronal activity in the mammalian brain. We performed calcium imaging of individual motor cortex neurons, while delivering 0.35 MHz ultrasound at PRFs of 10, 40, and 140 Hz in awake mice. We found that most neurons were preferentially activated by only one of the three PRFs, highlighting unique cellular effects of physiologic PRFs. Further, ultrasound evoked responses were similar between excitatory neurons and parvalbumin positive interneurons regardless of PRFs, indicating that individual cell sensitivity dominates ultrasound-evoked effects, consistent with the heterogeneous mechanosensitive channel expression we found across single neurons in mice and humans. These results highlight the feasibility of tuning ultrasound neuromodulation effects through varying PRFs.
Collapse
|
36
|
Davis MJ, Zawieja SD. Pacemaking in the lymphatic system. J Physiol 2024. [PMID: 38520402 DOI: 10.1113/jp284752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/08/2024] [Indexed: 03/25/2024] Open
Abstract
Lymphatic collecting vessels exhibit spontaneous phasic contractions that are critical for lymph propulsion and tissue fluid homeostasis. This rhythmic activity is driven by action potentials conducted across the lymphatic muscle cell (LMC) layer to produce entrained contractions. The contraction frequency of a lymphatic collecting vessel displays exquisite mechanosensitivity, with a dynamic range from <1 to >20 contractions per minute. A myogenic pacemaker mechanism intrinsic to the LMCs was initially postulated to account for pressure-dependent chronotropy. Further interrogation into the cellular constituents of the lymphatic vessel wall identified non-muscle cell populations that shared some characteristics with interstitial cells of Cajal, which have pacemaker functions in the gastrointestinal and lower urinary tracts, thus raising the possibility of a non-muscle cell pacemaker. However, recent genetic knockout studies in mice support LMCs and a myogenic origin of the pacemaker activity. LMCs exhibit stochastic, but pressure-sensitive, sarcoplasmic reticulum calcium release (puffs and waves) from IP3R1 receptors, which couple to the calcium-activated chloride channel Anoctamin 1, causing depolarisation. The resulting electrical activity integrates across the highly coupled lymphatic muscle electrical syncytia through connexin 45 to modulate diastolic depolarisation. However, multiple other cation channels may also contribute to the ionic pacemaking cycle. Upon reaching threshold, a voltage-gated calcium channel-dependent action potential fires, resulting in a nearly synchronous calcium global calcium flash within the LMC layer to drive an entrained contraction. This review summarizes the key ion channels potentially responsible for the pressure-dependent chronotropy of lymphatic collecting vessels and various mechanisms of IP3R1 regulation that could contribute to frequency tuning.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Scott D Zawieja
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, USA
| |
Collapse
|
37
|
Jojoa-Cruz S, Dubin AE, Lee WH, Ward A. Structure-guided mutagenesis of OSCAs reveals differential activation to mechanical stimuli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.03.560740. [PMID: 37873218 PMCID: PMC10592937 DOI: 10.1101/2023.10.03.560740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The dimeric two-pore OSCA/TMEM63 family has recently been identified as mechanically activated ion channels. Previously, based on the unique features of the structure of OSCA1.2, we postulated the potential involvement of several structural elements in sensing membrane tension1. Interestingly, while OSCA1, 2, and 3 clades are activated by membrane stretch in cell-attached patches (i.e., they are stretch-activated channels), they differ in their ability to transduce membrane deformation induced by a blunt probe (poking). In an effort to understand the domains contributing to mechanical signal transduction, we used cryo-electron microscopy to solve the structure of Arabidopsis thaliana (At) OSCA3.1, which, unlike AtOSCA1.2, only produced stretch- but not poke-activated currents in our initial characterization2. Mutagenesis and electrophysiological assessment of conserved and divergent putative mechanosensitive features of OSCA1.2 reveal a selective disruption of the macroscopic currents elicited by poking without considerable effects on stretch-activated currents (SAC). Our results support the involvement of the amphipathic helix and lipid-interacting residues in the membrane fenestration in the response to poking. Our findings position these two structural elements as potential sources of functional diversity within the family.
Collapse
Affiliation(s)
- Sebastian Jojoa-Cruz
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California 92037, USA
| | - Adrienne E. Dubin
- Department of Neuroscience, Scripps Research, La Jolla, California 92037, USA
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California 92037, USA
| | - Andrew Ward
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California 92037, USA
| |
Collapse
|
38
|
Petersen EN, Pavel MA, Hansen SS, Gudheti M, Wang H, Yuan Z, Murphy KR, Ja W, Ferris HA, Jorgensen E, Hansen SB. Mechanical activation of TWIK-related potassium channel by nanoscopic movement and rapid second messenger signaling. eLife 2024; 12:RP89465. [PMID: 38407149 PMCID: PMC10942622 DOI: 10.7554/elife.89465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024] Open
Abstract
Rapid conversion of force into a biological signal enables living cells to respond to mechanical forces in their environment. The force is believed to initially affect the plasma membrane and then alter the behavior of membrane proteins. Phospholipase D2 (PLD2) is a mechanosensitive enzyme that is regulated by a structured membrane-lipid site comprised of cholesterol and saturated ganglioside (GM1). Here we show stretch activation of TWIK-related K+ channel (TREK-1) is mechanically evoked by PLD2 and spatial patterning involving ordered GM1 and 4,5-bisphosphate (PIP2) clusters in mammalian cells. First, mechanical force deforms the ordered lipids, which disrupts the interaction of PLD2 with the GM1 lipids and allows a complex of TREK-1 and PLD2 to associate with PIP2 clusters. The association with PIP2 activates the enzyme, which produces the second messenger phosphatidic acid (PA) that gates the channel. Co-expression of catalytically inactive PLD2 inhibits TREK-1 stretch currents in a biological membrane. Cellular uptake of cholesterol inhibits TREK-1 currents in culture and depletion of cholesterol from astrocytes releases TREK-1 from GM1 lipids in mouse brain. Depletion of the PLD2 ortholog in flies results in hypersensitivity to mechanical force. We conclude PLD2 mechanosensitivity combines with TREK-1 ion permeability to elicit a mechanically evoked response.
Collapse
Affiliation(s)
- E Nicholas Petersen
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
- Scripps Research Skaggs Graduate School of Chemical and Biological Science, The Scripps Research Institute, Scripps,JupiterUnited States
| | - Mahmud Arif Pavel
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
| | - Samuel S Hansen
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
| | - Manasa Gudheti
- Division of Endocrinology and Metabolism, Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Hao Wang
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
- Scripps Research Skaggs Graduate School of Chemical and Biological Science, The Scripps Research Institute, Scripps,JupiterUnited States
| | - Zixuan Yuan
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
- Scripps Research Skaggs Graduate School of Chemical and Biological Science, The Scripps Research Institute, Scripps,JupiterUnited States
| | - Keith R Murphy
- Department of Neuroscience, The Scripps Research Institute, ScrippsJupiterUnited States
- Center on Aging,The Scripps Research Institute, ScrippsJupiterUnited States
| | - William Ja
- Department of Neuroscience, The Scripps Research Institute, ScrippsJupiterUnited States
- Center on Aging,The Scripps Research Institute, ScrippsJupiterUnited States
| | - Heather A Ferris
- Division of Endocrinology and Metabolism, Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Erik Jorgensen
- Department of Biology, Howard Hughes Medical Institute, University of UtahSalt Lake CityUnited States
| | - Scott B Hansen
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
| |
Collapse
|
39
|
Abstract
Mechanical forces influence different cell types in our bodies. Among the earliest forces experienced in mammals is blood movement in the vascular system. Blood flow starts at the embryonic stage and ceases when the heart stops. Blood flow exposes endothelial cells (ECs) that line all blood vessels to hemodynamic forces. ECs detect these mechanical forces (mechanosensing) through mechanosensors, thus triggering physiological responses such as changes in vascular diameter. In this review, we focus on endothelial mechanosensing and on how different ion channels, receptors, and membrane structures detect forces and mediate intricate mechanotransduction responses. We further highlight that these responses often reflect collaborative efforts involving several mechanosensors and mechanotransducers. We close with a consideration of current knowledge regarding the dysregulation of endothelial mechanosensing during disease. Because hemodynamic disruptions are hallmarks of cardiovascular disease, studying endothelial mechanosensing holds great promise for advancing our understanding of vascular physiology and pathophysiology.
Collapse
Affiliation(s)
- Xin Rui Lim
- Department of Pharmacology, Larner College of Medicine and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA;
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA;
| |
Collapse
|
40
|
Bassetto CAZ, Pfeffermann J, Yadav R, Strassgschwandtner S, Glasnov T, Bezanilla F, Pohl P. Photolipid excitation triggers depolarizing optocapacitive currents and action potentials. Nat Commun 2024; 15:1139. [PMID: 38326372 PMCID: PMC10850502 DOI: 10.1038/s41467-024-45403-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/22/2024] [Indexed: 02/09/2024] Open
Abstract
Optically-induced changes in membrane capacitance may regulate neuronal activity without requiring genetic modifications. Previously, they mainly relied on sudden temperature jumps due to light absorption by membrane-associated nanomaterials or water. Yet, nanomaterial targeting or the required high infrared light intensities obstruct broad applicability. Now, we propose a very versatile approach: photolipids (azobenzene-containing diacylglycerols) mediate light-triggered cellular de- or hyperpolarization. As planar bilayer experiments show, the respective currents emerge from millisecond-timescale changes in bilayer capacitance. UV light changes photolipid conformation, which awards embedding plasma membranes with increased capacitance and evokes depolarizing currents. They open voltage-gated sodium channels in cells, generating action potentials. Blue light reduces the area per photolipid, decreasing membrane capacitance and eliciting hyperpolarization. If present, mechanosensitive channels respond to the increased mechanical membrane tension, generating large depolarizing currents that elicit action potentials. Membrane self-insertion of administered photolipids and focused illumination allows cell excitation with high spatiotemporal control.
Collapse
Affiliation(s)
- Carlos A Z Bassetto
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, 60637, USA
| | - Juergen Pfeffermann
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstraße 40, 4020, Linz, Austria
| | - Rohit Yadav
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstraße 40, 4020, Linz, Austria
| | | | - Toma Glasnov
- Institute of Chemistry, Karl-Franzens-University, Graz, Austria
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, 60637, USA.
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.
| | - Peter Pohl
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstraße 40, 4020, Linz, Austria.
| |
Collapse
|
41
|
Xia K, Chen X, Wang W, Liu Q, Zhao M, Ma J, Jia H. Roles of mechanosensitive ion channels in immune cells. Heliyon 2024; 10:e23318. [PMID: 38148826 PMCID: PMC10750075 DOI: 10.1016/j.heliyon.2023.e23318] [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: 09/16/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Mechanosensitive ion channels are a class of membrane-integrated proteins that convert externalmechanical forces, including stretching, pressure, gravity, and osmotic pressure changes, some of which can be caused by pathogen invasion, into electrical and chemical signals transmitted to the cytoplasm. In recent years, with the identification of many of these channels, their roles in the initiation and progression of many diseases have been gradually revealed. Multiple studies have shown that mechanosensitive ion channels regulate the proliferation, activation, and inflammatory responses of immune cells by being expressed on the surface of immune cells and further responding to mechanical forces. Nonetheless, further clarification is required regarding the signaling pathways of immune-cell pattern-recognition receptors and on the impact of microenvironmental changes and mechanical forces on immune cells. This review summarizes the roles of mechanosensitive ion channels in immune cells.
Collapse
Affiliation(s)
- Kexin Xia
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Xiaolin Chen
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Wenyan Wang
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Qianwen Liu
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Mai Zhao
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Road 100, Shanghai, 200080, China
| | - Jiacheng Ma
- The Department of Information Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, 999077, China
| | - Hao Jia
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry & Molecular Cellular Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
42
|
Qin PP, Jin M, Xia AW, Li AS, Lin TT, Liu Y, Kan RL, Zhang BB, Kranz GS. The effectiveness and safety of low-intensity transcranial ultrasound stimulation: A systematic review of human and animal studies. Neurosci Biobehav Rev 2024; 156:105501. [PMID: 38061596 DOI: 10.1016/j.neubiorev.2023.105501] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/07/2023] [Accepted: 12/02/2023] [Indexed: 12/26/2023]
Abstract
Low-intensity transcranial ultrasound stimulation (LITUS) is a novel non-invasive neuromodulation technique. We conducted a systematic review to evaluate current evidence on the efficacy and safety of LITUS neuromodulation. Five databases were searched from inception to May 31, 2023. Randomized controlled human trials and controlled animal studies were included. The neuromodulation effects of LITUS on clinical or pre-clinical, neurophysiological, neuroimaging, histological and biochemical outcomes, and adverse events were summarized. In total, 11 human studies and 44 animal studies were identified. LITUS demonstrated therapeutic efficacy in neurological disorders, psychiatric disorders, pain, sleep disorders and hypertension. LITUS-related changes in neuronal structure and cortical activity were found. From histological and biochemical perspectives, prominent findings included suppressing the inflammatory response and facilitating neurogenesis. No adverse effects were reported in controlled animal studies included in our review, while reversible headache, nausea, and vomiting were reported in a few human subjects. Overall, LITUS alleviates various symptoms and modulates associated brain circuits without major side effects. Future research needs to establish a solid therapeutic framework for LITUS.
Collapse
Affiliation(s)
- Penny Ping Qin
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Minxia Jin
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China; Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Adam Weili Xia
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Ami Sinman Li
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Tim Tianze Lin
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Yuchen Liu
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Rebecca Laidi Kan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Bella Bingbing Zhang
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Georg S Kranz
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China; Mental Health Research Center (MHRC), The Hong Kong Polytechnic University, Hong Kong, SAR, China; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
43
|
Mitrokhin V, Bilichenko A, Kazanski V, Schobik R, Shileiko S, Revkova V, Kalsin V, Kamkina O, Kamkin A, Mladenov M. Transcriptomic profile of the mechanosensitive ion channelome in human cardiac fibroblasts. Exp Biol Med (Maywood) 2023; 248:2341-2350. [PMID: 38158807 PMCID: PMC10903254 DOI: 10.1177/15353702231218488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/27/2023] [Indexed: 01/03/2024] Open
Abstract
Human cardiac fibroblasts (HCFs) have mRNA transcripts that encode different mechanosensitive ion channels and channel regulatory proteins whose functions are not known yet. The primary goal of this work was to define the mechanosensitive ion channelome of HCFs. The most common type of cationic channel is the transient receptor potential (TRP) family, which is followed by the TWIK-related K+ channel (TREK), transmembrane protein 63 (TMEM63), and PIEZO channel (PIEZO) families. In the sodium-dependent NON-voltage-gated channel (SCNN) subfamily, only SCNN1D was shown to be highly expressed. Particular members of the acid-sensing ion channel (ASIC) (ASIC1 and ASIC3) subfamilies were also significantly expressed. The transcripts per kilobase million (TPMs) for Piezo 2 were almost 100 times less abundant than those for Piezo 1. The tandem of P domains in a weak inward rectifying K+ channel (TWIK)-2 channel, TWIK-related acid-sensitive K+ channel (TASK)-5, TASK-1, and the TWIK-related K1 (TREK-1) channel were the four most prevalent types in the K2P subfamily. The highest expression in the TRPP subfamily was found for PKD2 and PKD1, while in the TRPM subfamily, it was found for TRPM4, TRPM7, and TRPM3. TRPV2, TRPV4, TRPV3, and TRPV6 (all members of the TRPV subfamily) were also substantially expressed. A strong expression of the TRPC1, TRPC4, TRPC6, and TRPC2 channels and all members of the TRPML subfamily (MCOLN1, MCOLN2, and MCOLN3) was also shown. In terms of the transmembrane protein 16 (TMEM16) family, the HCFs demonstrated significant expression of the TMEM16H, TMEM16F, TMEM16J, TMEM16A, and TMEM16G channels. TMC3 is the most expressed channel in HCFs of all known members of the transmembrane channel-like protein (TMC) family. This analysis of the mechanosensitive ionic channel transcriptome in HCFs: (1) agrees with previously documented findings that all currently identified mechanosensitive channels play a significant and well recognized physiological function in elucidating the mechanosensitive characteristics of HCFs; (2) supports earlier preliminary reports that point to the most common expression of the TRP mechanosensitive family in HCFs; and (3) points to other new mechanosensitive channels (TRPC1, TRPC2, TWIK-2, TMEM16A, ASIC1, and ASIC3).
Collapse
Affiliation(s)
- Vadim Mitrokhin
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Andrei Bilichenko
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Viktor Kazanski
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Roman Schobik
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Stanislav Shileiko
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Veronika Revkova
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Vladimir Kalsin
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Olga Kamkina
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Andre Kamkin
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Mitko Mladenov
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia
| |
Collapse
|
44
|
Hori A, Fukazawa A, Katanosaka K, Mizuno M, Hotta N. Mechanosensitive channels in the mechanical component of the exercise pressor reflex. Auton Neurosci 2023; 250:103128. [PMID: 37925831 DOI: 10.1016/j.autneu.2023.103128] [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: 06/18/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023]
Abstract
The cardiovascular response is appropriately regulated during exercise to meet the metabolic demands of the active muscles. The exercise pressor reflex is a neural feedback mechanism through thin-fiber muscle afferents activated by mechanical and metabolic stimuli in the active skeletal muscles. The mechanical component of this reflex is referred to as skeletal muscle mechanoreflex. Its initial step requires mechanotransduction mediated by mechanosensors, which convert mechanical stimuli into biological signals. Recently, various mechanosensors have been identified, and their contributions to muscle mechanoreflex have been actively investigated. Nevertheless, the mechanosensitive channels responsible for this muscular reflex remain largely unknown. This review discusses progress in our understanding of muscle mechanoreflex under healthy conditions, focusing on mechanosensitive channels.
Collapse
Affiliation(s)
- Amane Hori
- College of Life and Health Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan; Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo 102-8472, Japan; Department of Applied Clinical Research, UT Southwestern Medical Center, Dallas, TX 75390-9174, USA
| | - Ayumi Fukazawa
- Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo 102-8472, Japan; Department of Applied Clinical Research, UT Southwestern Medical Center, Dallas, TX 75390-9174, USA
| | - Kimiaki Katanosaka
- College of Life and Health Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan
| | - Masaki Mizuno
- Department of Applied Clinical Research, UT Southwestern Medical Center, Dallas, TX 75390-9174, USA
| | - Norio Hotta
- College of Life and Health Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan.
| |
Collapse
|
45
|
Cepkenovic B, Friedland F, Noetzel E, Maybeck V, Offenhäusser A. Single-neuron mechanical perturbation evokes calcium plateaus that excite and modulate the network. Sci Rep 2023; 13:20669. [PMID: 38001109 PMCID: PMC10673841 DOI: 10.1038/s41598-023-47090-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Mechanical stimulation is a promising means to non-invasively excite and modulate neuronal networks with a high spatial resolution. Despite the thorough characterization of the initiation mechanism, whether or how mechanical responses disperse into non-target areas remains to be discovered. Our in vitro study demonstrates that a single-neuron deformation evokes responses that propagate to about a third of the untouched neighbors. The responses develop via calcium influx through mechanosensitive channels and regeneratively propagate through the neuronal ensemble via gap junctions. Although independent of action potentials and synapses, mechanical responses reliably evoke membrane depolarizations capable of inducing action potentials both in the target and neighbors. Finally, we show that mechanical stimulation transiently potentiates the responding assembly for further inputs, as both gain and excitability are transiently increased exclusively in neurons that respond to a neighbor's mechanical stimulation. The findings indicate a biological component affecting the spatial resolution of mechanostimulation and point to a cross-talk in broad-network mechanical stimulations. Since giga-seal formation in patch-clamp produces a similar mechanical stimulus on the neuron, our findings inform which neuroscientific questions could be reliably tackled with patch-clamp and what recovery post-gigaseal formation is necessary.
Collapse
Affiliation(s)
- Bogdana Cepkenovic
- Institute of Biological Information Processing: Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße 1, 52428, Jülich, Germany
- RWTH Aachen University, Templergraben 55, 52062, Aachen, Germany
| | - Florian Friedland
- Institute of Biological Information Processing: Mechanobiology (IBI-2), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße 1, 52428, Jülich, Germany
| | - Erik Noetzel
- Institute of Biological Information Processing: Mechanobiology (IBI-2), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße 1, 52428, Jülich, Germany
| | - Vanessa Maybeck
- Institute of Biological Information Processing: Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße 1, 52428, Jülich, Germany.
| | - Andreas Offenhäusser
- Institute of Biological Information Processing: Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße 1, 52428, Jülich, Germany
- RWTH Aachen University, Templergraben 55, 52062, Aachen, Germany
| |
Collapse
|
46
|
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
|
47
|
Abstract
All cells in the body are exposed to physical force in the form of tension, compression, gravity, shear stress, or pressure. Cells convert these mechanical cues into intracellular biochemical signals; this process is an inherent property of all cells and is essential for numerous cellular functions. A cell's ability to respond to force largely depends on the array of mechanical ion channels expressed on the cell surface. Altered mechanosensing impairs conscious senses, such as touch and hearing, and unconscious senses, like blood pressure regulation and gastrointestinal (GI) activity. The GI tract's ability to sense pressure changes and mechanical force is essential for regulating motility, but it also underlies pain originating in the GI tract. Recent identification of the mechanically activated ion channels Piezo1 and Piezo2 in the gut and the effects of abnormal ion channel regulation on cellular function indicate that these channels may play a pathogenic role in disease. Here, we discuss our current understanding of mechanically activated Piezo channels in the pathogenesis of pancreatic and GI diseases, including pancreatitis, diabetes mellitus, irritable bowel syndrome, GI tumors, and inflammatory bowel disease. We also describe how Piezo channels could be important targets for treating GI diseases.
Collapse
|
48
|
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
|
49
|
Liu WW, Kinzy TG, Cooke Bailey JN, Xu Z, Hysi P, Wiggs JL. Mechanosensitive ion channel gene survey suggests potential roles in primary open angle glaucoma. Sci Rep 2023; 13:15871. [PMID: 37741866 PMCID: PMC10517927 DOI: 10.1038/s41598-023-43072-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023] Open
Abstract
Although glaucoma is a disease modulated by eye pressure, the mechanisms of pressure sensing in the eye are not well understood. Here, we investigated associations between mechanosensitive ion channel gene variants and primary open-angle glaucoma (POAG). Common (minor allele frequency > 5%) single nucleotide polymorphisms located within the genomic regions of 20 mechanosensitive ion channel genes in the K2P, TMEM63, PIEZO and TRP channel families were assessed using genotype data from the NEIGHBORHOOD consortium of 3853 cases and 33,480 controls. Rare (minor allele frequency < 1%) coding variants were assessed using exome array genotyping data for 2606 cases and 2606 controls. Association with POAG was analyzed using logistic regression adjusting for age and sex. Two rare PIEZO1 coding variants with protective effects were identified in the NEIGHBOR dataset: R1527H, (OR 0.17, P = 0.0018) and a variant that alters a canonical splice donor site, g.16-88737727-C-G Hg38 (OR 0.38, P = 0.02). Both variants showed similar effects in the UK Biobank and the R1527H also in the FinnGen database. Several common variants also reached study-specific thresholds for association in the NEIGHBORHOOD dataset. These results identify novel variants in several mechanosensitive channel genes that show associations with POAG, suggesting that these channels may be potential therapeutic targets.
Collapse
Affiliation(s)
- Wendy W Liu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, 2370 Watson Court, Palo Alto, CA, 94303, USA.
| | - Tyler G Kinzy
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Jessica N Cooke Bailey
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Zihe Xu
- Department of Ophthalmology, King's College London, St. Thomas' Hospital, London, UK
| | - Pirro Hysi
- Department of Ophthalmology, King's College London, St. Thomas' Hospital, London, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, St. Thomas' Hospital, London, UK
| | - Janey L Wiggs
- Massachusetts Eye and Ear, Harvard Medical School Boston, Boston, MA, USA
| |
Collapse
|
50
|
Li C, Liu P, Zhi Y, Zhai Y, Liu Z, Gao L, Jiang L. Ultra-mechanosensitive Chloride Ion Transport through Bioinspired High-Density Elastomeric Nanochannels. J Am Chem Soc 2023; 145:19098-19106. [PMID: 37603884 DOI: 10.1021/jacs.3c07675] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Mechanosensitive ion channels play crucial roles in physiological activities, where small mechanical stimuli induce the membrane tension, trigger the ion channels' deformation, and are further transformed into significant electrochemical signals. Artificial ion channels with stiff moduli have been developed to mimic mechanosensory behaviors, exhibiting an electrochemical response by the high-pressure-induced flow. However, fabricating flexible mechanosensitive channels capable of regulating specific ion transporting upon dramatic deformation has remained a challenge. Here, we demonstrate bioinspired high-density elastomeric channels self-assembled by polyisoprene-b-poly4-vinylpyridine, which exhibit ultra-mechanosensitive chloride ion transport resulting from nanochannel deformation. The PI-formed continuous elastic matrix can transmit external forces into internal tensions, while P4VP forms transmembrane chloride channels that undergo dramatic deformation and respond to mechanical stimuli. The integrated and flexible chloride channels present a dramatic and stable electrochemical signal toward a low pressure of 0.2 mbar. This research first demonstrates the artificial mechanosensory chloride channels, which could provide a promising avenue for designing flexible and responsive channel systems.
Collapse
Affiliation(s)
- Chao Li
- Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, P. R. China
- Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Pengxiang Liu
- Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, P. R. China
| | - Yafang Zhi
- Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, P. R. China
| | - Yi Zhai
- Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, P. R. China
| | - Zhiwen Liu
- Oxford Instrument Technology China, Beijing 100034, P. R China
| | - Longcheng Gao
- Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, P. R. China
| | - Lei Jiang
- Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, P. R. China
- Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| |
Collapse
|