1
|
Kamal KY, Trombetta-Lima M. Mechanotransduction and Skeletal Muscle Atrophy: The Interplay Between Focal Adhesions and Oxidative Stress. Int J Mol Sci 2025; 26:2802. [PMID: 40141444 PMCID: PMC11943188 DOI: 10.3390/ijms26062802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Mechanical unloading leads to profound musculoskeletal degeneration, muscle wasting, and weakness. Understanding the specific signaling pathways involved is essential for uncovering effective interventions. This review provides new perspectives on mechanotransduction pathways, focusing on the critical roles of focal adhesions (FAs) and oxidative stress in skeletal muscle atrophy under mechanical unloading. As pivotal mechanosensors, FAs integrate mechanical and biochemical signals to sustain muscle structural integrity. When disrupted, these complexes impair force transmission, activating proteolytic pathways (e.g., ubiquitin-proteasome system) that accelerate atrophy. Oxidative stress, driven by mitochondrial dysfunction and NADPH oxidase-2 (NOX2) hyperactivation, exacerbates muscle degeneration through excessive reactive oxygen species (ROS) production, impaired repair mechanisms, and dysregulated redox signaling. The interplay between FA dysfunction and oxidative stress underscores the complexity of muscle atrophy pathogenesis: FA destabilization heightens oxidative damage, while ROS overproduction further disrupts FA integrity, creating a self-amplifying vicious cycle. Therapeutic strategies, such as NOX2 inhibitors, mitochondrial-targeted antioxidants, and FAK-activating compounds, promise to mitigate muscle atrophy by preserving mechanotransduction signaling and restoring redox balance. By elucidating these pathways, this review advances the understanding of muscle degeneration during unloading and identifies promising synergistic therapeutic targets, emphasizing the need for combinatorial approaches to disrupt the FA-ROS feedback loop.
Collapse
Affiliation(s)
- Khaled Y. Kamal
- Department of Kinesiology, Iowa State University, Ames, IA 50011, USA
| | - Marina Trombetta-Lima
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, 9700 Groningen, The Netherlands;
| |
Collapse
|
2
|
Roy S, Pyari G, Bansal H. Theoretical analysis of low-power deep synergistic sono-optogenetic excitation of neurons by co-expressing light-sensitive and mechano-sensitive ion-channels. Commun Biol 2025; 8:379. [PMID: 40050670 PMCID: PMC11885482 DOI: 10.1038/s42003-025-07792-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025] Open
Abstract
The present challenge in neuroscience is to non-invasively exercise low-power and high-fidelity control of neurons situated deep inside the brain. Although, two-photon optogenetic excitation can activate neurons to millimeter depth with sub-cellular specificity and millisecond temporal resolution, it can also cause heating of the targeted tissue. On the other hand, sonogenetics can non-invasively modulate the cellular activity of neurons expressed with mechano-sensitive proteins in deeper areas of the brain with less spatial selectivity. We present a theoretical analysis of a synergistic sono-optogenetic method to overcome these limitations by co-expressing a mechano-sensitive (MscL-I92L) ion-channel with a light-sensitive (CoChR/ChroME2s/ChRmine) ion-channel in hippocampal neurons. It is shown that in the presence of low-amplitude subthreshold ultrasound pulses, the two-photon excitation threshold for neural spiking reduces drastically by 73% with MscL-I92L-CoChR (0.021 mW/µm2), 66% with MscL-I92L-ChroME2s (0.029 mW/µm2), and 64% with MscL-I92L-ChRmine (0.013 mW/µm2) at 5 Hz. It allows deeper excitation of up to 1.2 cm with MscL-I92L-ChRmine combination. The method is useful to design new experiments for low-power deep excitation of neurons and multimodal neuroprosthetic devices and circuits.
Collapse
Affiliation(s)
- Sukhdev Roy
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, India.
| | - Gur Pyari
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, India
| | - Himanshu Bansal
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, India
| |
Collapse
|
3
|
Hwangbo H, Chae S, Ryu D, Kim G. In situ magnetic-field-assisted bioprinting process using magnetorheological bioink to obtain engineered muscle constructs. Bioact Mater 2025; 45:417-433. [PMID: 39697238 PMCID: PMC11653149 DOI: 10.1016/j.bioactmat.2024.11.035] [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: 09/21/2024] [Revised: 11/17/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Tissue-engineered anisotropic cell constructs are promising candidates for treating volumetric muscle loss (VML). However, achieving successful cell alignment within macroscale 3D cell constructs for skeletal muscle tissue regeneration remains challenging, owing to difficulties in controlling cell arrangement within a low-viscosity hydrogel. Herein, we propose the concept of a magnetorheological bioink to manipulate the cellular arrangement within a low-viscosity hydrogel. This bioink consisted of gelatin methacrylate (GelMA), iron oxide nanoparticles, and human adipose stem cells (hASCs). The cell arrangement is regulated by the responsiveness of iron oxide nanoparticles to external magnetic fields. A bioprinting process using ring magnets was developed for in situ bioprinting, resulting in well-aligned 3D cell structures and enhanced mechanotransduction effects on hASCs. In vitro analyses revealed upregulation of cellular activities, including myogenic-related gene expression, in hASCs. When implanted into a VML mouse model, the bioconstructs improved muscle functionality and regeneration, validating the effectiveness of the proposed approach.
Collapse
Affiliation(s)
- Hanjun Hwangbo
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - SooJung Chae
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - GeunHyung Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do, 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
4
|
Jochum D, Konrad A, Lohmann LH, Cochrane D, Rittweger J, Vogel V, Warneke K. The merit of superimposed vibration for flexibility and passive stiffness: A systematic review with multilevel meta-analysis. JOURNAL OF SPORT AND HEALTH SCIENCE 2025; 14:101033. [PMID: 40021055 DOI: 10.1016/j.jshs.2025.101033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/03/2024] [Accepted: 11/14/2024] [Indexed: 03/03/2025]
Abstract
BACKGROUND Due to its high relevance in sports and rehabilitation, the exploration of interventions to further optimize flexibility becomes paramount. While stretching might be the most common way to enhance range of motion, these increases could be optimized by imposing an additional activation of the muscle, such as mechanical vibratory stimulation. While several original articles provide promising findings, contradictory results on flexibility and underlying mechanisms (e.g., stiffness), reasonable effect size (ES) pooling remains scarce. With this work we systematically reviewed the available literature to explore the possibility of potentiating flexibility, stiffness, and passive torque adaptations by superimposing mechanical vibration stimulation. METHODS A systematic search of 4 databases (Web of Science, MEDLINE, Scopus, and Cochrane Public Library) was conducted until December 2023 to identify studies comparing mechanical vibratory interventions with passive controls or the same intervention without vibration (sham) on range of motion and passive muscle stiffness in acute (immediate effects after single session) and chronic conditions (multiple sessions over a period of time). ES pooling was conducted using robust variance estimation via R to account for multiple study outcomes. Potential moderators of effects were analyzed using meta regression. RESULTS Overall, 65 studies (acute: 1162 participants, chronic: 788 participants) were included. There was moderate certainty of evidence for acute flexibility (ES = 0.71, p < 0.001) and stiffness (ES = -0.89, p = 0.006) effects of mechanical vibration treatments vs. passive controls without meaningful results against the sham condition (flexibility: ES = 0.20, p < 0.001; stiffness: ES = -0.19, p = 0.076). Similarly, moderate certainty of evidence was found for chronic vibration effects on flexibility (control: ES = 0.64, p = 0.043; sham: ES = 0.65, p < 0.001). Lack of studies and large outcome heterogeneity prevented ES pooling for underlying mechanisms. CONCLUSION Vibration improved flexibility in acute and chronic interventions compared to the stand-alone intervention, which can possibly be attributed to an accumulated mechanical stimulus through vibration. However, studies on biological mechanisms are needed to explain flexibility and stiffness effects in response to specific vibration modalities and timing.
Collapse
Affiliation(s)
- Daniel Jochum
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8006, Switzerland.
| | - Andreas Konrad
- Institute of Human Movement Science, Sport and Health, University of Graz, Graz 8010, Austria
| | - Lars H Lohmann
- Department of Human Motion Science and Exercise Physiology, Friedrich Schiller University Jena, Jena 07749, Germany
| | - Darryl Cochrane
- School of Sport, Exercise and Nutrition, College of Health, Massey University, Palmerston North 4442, New Zealand
| | - Jörn Rittweger
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne 51147, Germany; Department of Pediatrics and Adolescence Medicine, University Hospital Cologne, Cologne 50937, Germany
| | - Viola Vogel
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8006, Switzerland
| | - Konstantin Warneke
- Institute of Human Movement Science, Sport and Health, University of Graz, Graz 8010, Austria
| |
Collapse
|
5
|
Lacroix JJ, Wijerathne TD. PIEZO channels as multimodal mechanotransducers. Biochem Soc Trans 2025; 53:BST20240419. [PMID: 39936392 PMCID: PMC12010695 DOI: 10.1042/bst20240419] [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: 10/30/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/13/2025]
Abstract
All living beings experience a wide range of endogenous and exogenous mechanical forces. The ability to detect these forces and rapidly convert them into specific biological signals is essential to a wide range of physiological processes. In vertebrates, these fundamental tasks are predominantly achieved by two related mechanosensitive ion channels called PIEZO1 and PIEZO2. PIEZO channels are thought to sense mechanical forces through flexible transmembrane blade-like domains. Structural studies indeed show that these mechanosensory domains adopt a curved conformation in a resting membrane but become flattened in a membrane under tension, promoting an open state. Yet, recent studies suggest the intriguing possibility that distinct mechanical stimuli activate PIEZO channels through discrete molecular rearrangements of these domains. In addition, biological signals downstream of PIEZO channel activation vary as a function of the mechanical stimulus and of the cellular context. These unique features could explain how PIEZOs confer cells the ability to differentially interpret a complex landscape of mechanical cues.
Collapse
Affiliation(s)
- Jérôme J Lacroix
- Department of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, U.S.A
| | - Tharaka D Wijerathne
- Department of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, U.S.A
| |
Collapse
|
6
|
Li Y, Wang X, Guo J, Wang Y, Zykov V, Bodenschatz E, Gao X. Sonogenetics is a novel antiarrhythmic mechanism. CHAOS (WOODBURY, N.Y.) 2025; 35:013127. [PMID: 39792701 DOI: 10.1063/5.0224817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 12/01/2024] [Indexed: 01/12/2025]
Abstract
Arrhythmia of the heart is a dangerous and potentially fatal condition. The current widely used treatment is the implantable cardioverter defibrillator (ICD), but it is invasive and affects the patient's quality of life. The sonogenetic mechanism proposed here focuses ultrasound on a cardiac tissue, controls endogenous stretch-activated Piezo1 ion channels on the focal region's cardiomyocyte sarcolemma, and restores normal heart rhythm. In contrast to anchoring the implanted ICD lead at a fixed position in the myocardium, the size and position of the ultrasound focal region can be selected dynamically by adjusting the signals of every piezoelectric chip on the ultrasonic phased array, and it allows novel and efficient defibrillations. Based on the developed interdisciplinary electro-mechanical model of sonogenetic treatment, our analysis shows that the proposed ultrasound intensity and frequency will be safe and painless for humans and well below the limits established by the U.S. Food and Drug Administration.
Collapse
Affiliation(s)
- Yang Li
- School of Science, Beijing University of Posts and Telecommunications, Beijing 100876, China
- School of Physics and Information Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Xingang Wang
- School of Physics and Information Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Jianzhong Guo
- School of Physics and Information Technology, Shaanxi Normal University, Xi'an 710062, China
| | - Yong Wang
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen 37077, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen 37077, Germany
| | - Vladimir Zykov
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen 37077, Germany
| | - Eberhard Bodenschatz
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen 37077, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen 37077, Germany
- Institute for Dynamics of Complex Systems, University of Göttingen, Göttingen 37075, Germany
- Laboratory of Atomic and Solid-State Physics and Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Xiang Gao
- School of Physics and Information Technology, Shaanxi Normal University, Xi'an 710062, China
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen 37077, Germany
| |
Collapse
|
7
|
Wan Y, Zhou J, Li H. The Role of Mechanosensitive Piezo Channels in Chronic Pain. J Pain Res 2024; 17:4199-4212. [PMID: 39679432 PMCID: PMC11646438 DOI: 10.2147/jpr.s490459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/28/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose of Review Mechanosensitive Piezo channels are ion channels activated by mechanical stimuli, playing a crucial role in mechanotransduction processes and mechanical hypersensitivity. When these channels are subjected to mechanical loading, membrane currents rise instantaneously, depolarizing and activating voltage-gated calcium channels. This results in an increase in intracellular Ca2+, which contributes to heightened sensitivity to mechanical stimuli. This review delves into the characteristics and mechanisms of Piezo channels in chronic pain. Recent Findings The findings suggest that Piezo channels are integral to the occurrence and development of chronic pain, including neuropathic pain, visceral pain, musculoskeletal pain, headache or orofacial pain, and inflammatory pain. Piezo channels significantly impact pain perception and transmission. These channels' critical involvement in various pain types highlights their potential as promising targets for chronic pain therapy. Summary This review discusses the role of Piezo channels in chronic pain. By understanding these pain mechanisms, new therapeutic strategies can be developed to alleviate chronic pain, offering hope for patients suffering from these debilitating conditions.
Collapse
Affiliation(s)
- Yantong Wan
- Department of Anesthesiology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, People’s Republic of China
| | - Jieshu Zhou
- Department of Anesthesiology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, People’s Republic of China
| | - Hao Li
- Department of Anesthesiology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
8
|
Cubero-Sarabia M, Kapetanaki AM, Vassalli M. Biophysical assays to test cellular mechanosensing: moving towards high throughput. Biophys Rev 2024; 16:875-882. [PMID: 39830126 PMCID: PMC11735701 DOI: 10.1007/s12551-024-01263-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Mechanosensitivity is the ability of cells to sense and respond to mechanical stimuli. In order to do this, cells are endowed with different components that allow them to react to a broad range of stimuli, such as compression or shear forces, pressure, and vibrations. This sensing process, mechanosensing, is involved in fundamental physiological mechanisms, such as stem cell differentiation and migration, but it is also central to the development of pathogenic states. Here, we review the approaches that have been proposed to quantify mechanosensation in living cells, with a specific focus on methodologies that enable higher experimental throughput. This aspect is crucial to fully understand the nuances of mechanosensation and how it impacts the physiology and pathology of living systems. We will discuss traditional methods for studying mechanosensing at the level of single cells, with particular attention to the activation of the mechanosensitive ion channel piezo1. Moreover, we will present recent attempts to push the analysis towards higher throughput.
Collapse
Affiliation(s)
| | | | - Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Glasgow, UK
| |
Collapse
|
9
|
Li H, Lin J, Lin S, Zhong H, Jiang B, Liu X, Wu W, Li W, Iranmanesh E, Zhou Z, Li W, Wang K. A bioinspired tactile scanner for computer haptics. Nat Commun 2024; 15:7632. [PMID: 39223115 PMCID: PMC11369279 DOI: 10.1038/s41467-024-51674-2] [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: 01/25/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Computer haptics (CH) is about integration of tactile sensation and rendering in Metaverse. However, unlike computer vision (CV) where both hardware infrastructure and software programs are well developed, a generic tactile data capturing device that serves the same role as what a camera does for CV, is missing. Bioinspired by electrophysiological processes in human tactile somatosensory nervous system, here we propose a tactile scanner along with a neuromorphically-engineered system, in which a closed-loop tactile acquisition and rendering (re-creation) are preliminarily achieved. Based on the architecture of afferent nerves and intelligent functions of mechano-gating and leaky integrate-and-fire models, such a tactile scanner is designed and developed by using piezoelectric transducers as axon neurons and thin film transistor (TFT)-based neuromorphic circuits to mimic synaptic behaviours and neural functions. As an example, the neuron-like tactile information of surface textures is captured and further used to render the texture friction of a virtual surface for "recreating" a "true" feeling of touch.
Collapse
Affiliation(s)
- Huimin Li
- Guangdong Province Key Laboratory of Display Material and Technology, State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology (School of Microelectronics), Sun Yat-sen University, Guangzhou, 510275, China
| | - Jianle Lin
- Guangdong Province Key Laboratory of Display Material and Technology, State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology (School of Microelectronics), Sun Yat-sen University, Guangzhou, 510275, China
| | - Shuxin Lin
- Guangdong Province Key Laboratory of Display Material and Technology, State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology (School of Microelectronics), Sun Yat-sen University, Guangzhou, 510275, China
| | - Haojie Zhong
- Guangdong Province Key Laboratory of Display Material and Technology, State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology (School of Microelectronics), Sun Yat-sen University, Guangzhou, 510275, China
| | - Bowei Jiang
- Guangdong Province Key Laboratory of Display Material and Technology, State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology (School of Microelectronics), Sun Yat-sen University, Guangzhou, 510275, China
| | - Xinghui Liu
- Shenzhen Chipwey Innovation Technologies Co. Ltd., Shenzhen, 518100, China
| | - Weisheng Wu
- Guangdong Province Key Laboratory of Display Material and Technology, State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology (School of Microelectronics), Sun Yat-sen University, Guangzhou, 510275, China
| | - Weiwei Li
- State Key Laboratory of Microelectronic Devices & Integrated Technology, Institute of Microelectronics, Chinese Academy of Sciences, Beijing, 100029, China
| | - Emad Iranmanesh
- Guangdong Province Key Laboratory of Display Material and Technology, State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology (School of Microelectronics), Sun Yat-sen University, Guangzhou, 510275, China
| | - Zhongyi Zhou
- Shenzhen Chipwey Innovation Technologies Co. Ltd., Shenzhen, 518100, China
| | - Wenjun Li
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Kai Wang
- Guangdong Province Key Laboratory of Display Material and Technology, State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology (School of Microelectronics), Sun Yat-sen University, Guangzhou, 510275, China.
| |
Collapse
|
10
|
Zhou R, Fu W, Vasylyev D, Waxman SG, Liu CJ. Ion channels in osteoarthritis: emerging roles and potential targets. Nat Rev Rheumatol 2024; 20:545-564. [PMID: 39122910 DOI: 10.1038/s41584-024-01146-0] [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] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
Osteoarthritis (OA) is a highly prevalent joint disease that causes substantial disability, yet effective approaches to disease prevention or to the delay of OA progression are lacking. Emerging evidence has pinpointed ion channels as pivotal mediators in OA pathogenesis and as promising targets for disease-modifying treatments. Preclinical studies have assessed the potential of a variety of ion channel modulators to modify disease pathways involved in cartilage degeneration, synovial inflammation, bone hyperplasia and pain, and to provide symptomatic relief in models of OA. Some of these modulators are currently being evaluated in clinical trials. This review explores the structures and functions of ion channels, including transient receptor potential channels, Piezo channels, voltage-gated sodium channels, voltage-dependent calcium channels, potassium channels, acid-sensing ion channels, chloride channels and the ATP-dependent P2XR channels in the osteoarthritic joint. The discussion spans channel-targeting drug discovery and potential clinical applications, emphasizing opportunities for further research, and underscoring the growing clinical impact of ion channel biology in OA.
Collapse
Affiliation(s)
- Renpeng Zhou
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Wenyu Fu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Dmytro Vasylyev
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
11
|
Jing X, Kotelsky A, Zhang Y, Dirksen R, Mannava S, Buckley M, Lee W. Mechano-adaptation: Exercise-driven Piezo1 & Piezo2 augmentation and chondroprotection in articular cartilage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606183. [PMID: 39211060 PMCID: PMC11361185 DOI: 10.1101/2024.08.02.606183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Chondrocytes in adult joints are mechanosensitive post-mitotic quiescent cells with robustly expressed both Piezo1 and Piezo2 ion channels. Here, we examined the mechano-adaptation and Piezo modulations in articular chondrocytes using a mouse exercise model. We first found differential expression patterns of PIEZO1 and PIEZO2 in articular chondrocytes of healthy knee joints; chondrocytes in tibial cartilage (T) exhibit significantly higher PIEZO1 and PIEZO2 than femoral chondrocytes (F). Interestingly, a few weeks of exercise caused both PIEZO1 and PIEZO2 augmentation in F and T compared to the sedentary control group. Despite the increased expression levels of these mechanosensors, chondrocytes in exercised cartilage exhibit significantly reduced mechanical susceptibility against 1mJ impact. PIEZO1 modulation was relatively more rapid than PIEZO2 channels post-exercise. We tested the exercise-induced effect using Piezo1-conditional knockout (Pz1-cKO; Agc1 CreERT2 ;Piezo1 fl/fl ). Pz1-cKO mice exhibit diminished exercise-driven chondroprotection against 1mJ impact, suggesting essential roles of Piezo1-mediated mechanotransduction for physiologic-induced cartilage matrix homeostasis. In addition, using a mouse OA model, we further found the modulated PIEZO1 in chondrocytes, consistent with reports in Ren et al., but without PIEZO2 modulations over OA progression. In summary, our data reveal the distinctly tuned Piezo1 and Piezo2 channels in chondrocytes post-exercise and post-injury, in turn modulating the mechanical susceptibility of chondrocytes. We postulate that Piezo1 is a tightly-regulated biphasic biomarker ; Piezo1 antagonism may increase cellular survival post-injury and Piezo1 (with Piezo2) agonism to promote cartilage ECM restoration.
Collapse
|
12
|
Qiao LY. Satellite Glial Cells Bridge Sensory Neuron Crosstalk in Visceral Pain and Cross-Organ Sensitization. J Pharmacol Exp Ther 2024; 390:213-221. [PMID: 38777604 PMCID: PMC11264254 DOI: 10.1124/jpet.123.002061] [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: 12/15/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Following colonic inflammation, the uninjured bladder afferent neurons are also activated. The mechanisms and pathways underlying this sensory neuron cross-activation (from injured neurons to uninjured neurons) are not fully understood. Colonic and bladder afferent neurons reside in the same spinal segments and are separated by satellite glial cells (SGCs) and extracellular matrix in dorsal root ganglia (DRG). SGCs communicate with sensory neurons in a bidirectional fashion. This review summarizes the differentially regulated genes/proteins in the injured and uninjured DRG neurons and explores the role of SGCs in regulation of sensory neuron crosstalk in visceral cross-organ sensitization. The review also highlights the paracrine pathways in mediating neuron-SGC and SGC-neuron coupling with an emphasis on the neurotrophins and purinergic systems. Finally, I discuss the results from recent RNAseq profiling of SGCs to reveal useful molecular markers for characterization, functional study, and therapeutic targets of SGCs. SIGNIFICANCE STATEMENT: Satellite glial cells (SGCs) are the largest glial subtypes in sensory ganglia and play a critical role in mediating sensory neuron crosstalk, an underlying mechanism in colon-bladder cross-sensitization. Identification of novel and unique molecular markers of SGCs can advance the discovery of therapeutic targets in treatment of chronic pain including visceral pain comorbidity.
Collapse
Affiliation(s)
- Liya Y Qiao
- Department of Physiology and Biophysics, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
13
|
Cronin ME, Grandl J. Subconductance states add complexity to Piezo1 gating model. Trends Biochem Sci 2024; 49:567-568. [PMID: 38816279 PMCID: PMC11227380 DOI: 10.1016/j.tibs.2024.05.005] [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: 04/18/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/01/2024]
Abstract
Piezos are force-gated ion channels that detect and communicate membrane tension to the cell. Recent work from Ullah, Nosyreva, and colleagues characterizes partial channel openings, known as subconductance states, and develops a new gating model of Piezo1 function.
Collapse
Affiliation(s)
- Marie E Cronin
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jörg Grandl
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
14
|
Lüchtefeld I, Pivkin IV, Gardini L, Zare-Eelanjegh E, Gäbelein C, Ihle SJ, Reichmuth AM, Capitanio M, Martinac B, Zambelli T, Vassalli M. Dissecting cell membrane tension dynamics and its effect on Piezo1-mediated cellular mechanosensitivity using force-controlled nanopipettes. Nat Methods 2024; 21:1063-1073. [PMID: 38802520 PMCID: PMC11166569 DOI: 10.1038/s41592-024-02277-8] [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: 03/21/2022] [Accepted: 04/10/2024] [Indexed: 05/29/2024]
Abstract
The dynamics of cellular membrane tension and its role in mechanosensing, which is the ability of cells to respond to physical stimuli, remain incompletely understood, mainly due to the lack of appropriate tools. Here, we report a force-controlled nanopipette-based method that combines fluidic force microscopy with fluorescence imaging for precise manipulation of the cellular membrane tension while monitoring the impact on single-cell mechanosensitivity. The force-controlled nanopipette enables control of the indentation force imposed on the cell cortex as well as of the aspiration pressure applied to the plasma membrane. We show that this setup can be used to concurrently monitor the activation of Piezo1 mechanosensitive ion channels via calcium imaging. Moreover, the spatiotemporal behavior of the tension propagation is assessed with the fluorescent membrane tension probe Flipper-TR, and further dissected using molecular dynamics modeling. Finally, we demonstrate that aspiration and indentation act independently on the cellular mechanobiological machinery, that indentation induces a local pre-tension in the membrane, and that membrane tension stays confined by links to the cytoskeleton.
Collapse
Affiliation(s)
- Ines Lüchtefeld
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland.
| | - Igor V Pivkin
- Institute of Computing, Università della Svizzera Italiana, Lugano, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Lucia Gardini
- National Institute of Optics, National Research Council, Florence, Italy
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Florence, Italy
| | | | | | - Stephan J Ihle
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Andreas M Reichmuth
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Marco Capitanio
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Florence, Italy
- Physics and Astronomy Department, University of Florence, Florence, Italy
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Tomaso Zambelli
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland.
| | - Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Glasgow, UK.
| |
Collapse
|
15
|
Tiwari N, Smith C, Sharma D, Shen S, Mehta P, Qiao LY. Plp1-expresssing perineuronal DRG cells facilitate colonic and somatic chronic mechanical pain involving Piezo2 upregulation in DRG neurons. Cell Rep 2024; 43:114230. [PMID: 38743566 PMCID: PMC11234328 DOI: 10.1016/j.celrep.2024.114230] [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: 11/07/2023] [Revised: 03/06/2024] [Accepted: 04/26/2024] [Indexed: 05/16/2024] Open
Abstract
Satellite glial cells (SGCs) of dorsal root ganglia (DRGs) are activated in a variety of chronic pain conditions; however, their mediation roles in pain remain elusive. Here, we take advantage of proteolipid protein (PLP)/creERT-driven recombination in the periphery mainly occurring in SGCs of DRGs to assess the role of SGCs in the regulation of chronic mechanical hypersensitivity and pain-like responses in two organs, the distal colon and hindpaw, to test generality. We show that PLP/creERT-driven hM3Dq activation increases, and PLP/creERT-driven TrkB.T1 deletion attenuates, colon and hindpaw chronic mechanical hypersensitivity, positively associating with calcitonin gene-related peptide (CGRP) expression in DRGs and phospho-cAMP response element-binding protein (CREB) expression in the dorsal horn of the spinal cord. Activation of Plp1+ DRG cells also increases the number of small DRG neurons expressing Piezo2 and acquiring mechanosensitivity and leads to peripheral organ neurogenic inflammation. These findings unravel a role and mechanism of Plp1+ cells, mainly SGCs, in the facilitation of chronic mechanical pain and suggest therapeutic targets for pain mitigation.
Collapse
Affiliation(s)
- Namrata Tiwari
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA
| | - Cristina Smith
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA
| | - Divya Sharma
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA
| | - Shanwei Shen
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA
| | - Parshva Mehta
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA
| | - Liya Y Qiao
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA.
| |
Collapse
|
16
|
Yoshimoto K, Maki K, Adachi T, Kamei KI. Cyclic Stretching Enhances Angiocrine Signals at Liver Bud Stage from Human Pluripotent Stem Cells in Two-Dimensional Culture. Tissue Eng Part A 2024; 30:426-439. [PMID: 38062736 DOI: 10.1089/ten.tea.2023.0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024] Open
Abstract
Angiocrine signals during the development and growth of organs, including the liver, intestine, lung, and bone, are essential components of intercellular communication. The signals elicited during the liver bud stage are critical for vascularization and enhanced during the intercellular communication between the cells negative for kinase insert domain receptor (KDR) (KDR- cells) and the cells positive for KDR (KDR+ cells), which constitute the liver bud. However, the use of a human pluripotent stem cell (hPSC)-derived system has not facilitated the generation of a perfusable vascularized liver organoid that allows elucidation of liver development and has great potential for liver transplantation. This is largely owing to the lack of fundamental understanding to induce angiocrine signals in KDR- and KDR+ cells during the liver bud stage. We hypothesized that mechanical stimuli of cyclic stretching/pushing by the fetal heart adjacent to the liver bud could be the main contributor to promoting angiocrine signals in KDR- and KDR+ cells during the liver bud stage. In this study, we show that an organ-on-a-chip platform allows the emulation of an in vivo-like mechanical environment for the liver bud stage in vitro and investigate the role of cyclic mechanical stretching (cMS) to angiocrine signals in KDR- and KDR+ cells derived from hPSCs. RNA sequencing revealed that the expression of genes associated with epithelial-to-mesenchymal transition, including angiocrine signals, such as hepatocyte growth factor (HGF) and matrix metallopeptidase 9 (MMP9), were increased by cMS in cocultured KDR- and KDR+ cells. The expression and secretions of HGF and MMP9 were increased by 1.98- and 1.69-fold and 3.23- and 3.72-fold with cMS in the cocultured KDR- and KDR+ cells but were not increased by cMS in the monocultured KDR- and KDR+ cells, respectively. Finally, cMS during the liver bud stage did not lead to the dedifferentiation of hepatocytes, as the cells with cMS showed hepatic maker expression (CYP3A4, CYP3A7, ALB, and AAT) and 1.71-fold higher CYP3A activity than the cells without cMS, during 12 day-hepatocyte maturation after halting cMS. Our findings provide new insights into the mechanical factors during the liver bud stage and directions for future improvements in the engineered liver tissue.
Collapse
Affiliation(s)
- Koki Yoshimoto
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto, Japan
- Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Koichiro Maki
- Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Taiji Adachi
- Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Ken-Ichiro Kamei
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto, Japan
- Wuya College of Innovation, Shenyang Pharmaceutical University, Liaoning, China
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Liaoning, China
- Programs of Biology and Bioengineering, Divisions of Science and Engineering, New York University Abu Dhabi, Abu Dhabi, UAE
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, Brooklyn, New York, USA
| |
Collapse
|
17
|
Pitha I, Du L, Nguyen TD, Quigley H. IOP and glaucoma damage: The essential role of optic nerve head and retinal mechanosensors. Prog Retin Eye Res 2024; 99:101232. [PMID: 38110030 PMCID: PMC10960268 DOI: 10.1016/j.preteyeres.2023.101232] [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: 08/17/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 12/20/2023]
Abstract
There are many unanswered questions on the relation of intraocular pressure to glaucoma development and progression. IOP itself cannot be distilled to a single, unifying value, because IOP level varies over time, differs depending on ocular location, and can be affected by method of measurement. Ultimately, IOP level creates mechanical strain that affects axonal function at the optic nerve head which causes local extracellular matrix remodeling and retinal ganglion cell death - hallmarks of glaucoma and the cause of glaucomatous vision loss. Extracellular tissue strain at the ONH and lamina cribrosa is regionally variable and differs in magnitude and location between healthy and glaucomatous eyes. The ultimate targets of IOP-induced tissue strain in glaucoma are retinal ganglion cell axons at the optic nerve head and the cells that support axonal function (astrocytes, the neurovascular unit, microglia, and fibroblasts). These cells sense tissue strain through a series of signals that originate at the cell membrane and alter cytoskeletal organization, migration, differentiation, gene transcription, and proliferation. The proteins that translate mechanical stimuli into molecular signals act as band-pass filters - sensing some stimuli while ignoring others - and cellular responses to stimuli can differ based on cell type and differentiation state. Therefore, to fully understand the IOP signals that are relevant to glaucoma, it is necessary to understand the ultimate cellular targets of IOP-induced mechanical stimuli and their ability to sense, ignore, and translate these signals into cellular actions.
Collapse
Affiliation(s)
- Ian Pitha
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Nanomedicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Glaucoma Center of Excellence, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liya Du
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thao D Nguyen
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Harry Quigley
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Glaucoma Center of Excellence, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
18
|
He H, Zhou J, Xu X, Zhou P, Zhong H, Liu M. Piezo channels in the intestinal tract. Front Physiol 2024; 15:1356317. [PMID: 38379701 PMCID: PMC10877011 DOI: 10.3389/fphys.2024.1356317] [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: 12/15/2023] [Accepted: 01/24/2024] [Indexed: 02/22/2024] Open
Abstract
The intestine is the largest mechanosensitive organ in the human body whose epithelial cells, smooth muscle cells, neurons and enteroendocrine cells must sense and respond to various mechanical stimuli such as motility, distension, stretch and shear to regulate physiological processes including digestion, absorption, secretion, motility and immunity. Piezo channels are a newly discovered class of mechanosensitive ion channels consisting of two subtypes, Piezo1 and Piezo2. Piezo channels are widely expressed in the intestine and are involved in physiological and pathological processes. The present review summarizes the current research progress on the expression, function and regulation of Piezo channels in the intestine, with the aim of providing a reference for the future development of therapeutic strategies targeting Piezo channels.
Collapse
Affiliation(s)
- Haolong He
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jingying Zhou
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xuan Xu
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Pinxi Zhou
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Huan Zhong
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Acupuncture and Moxibustion Bioinformatics, Education Department of Hunan Province, Changsha, Hunan, China
| | - Mi Liu
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Acupuncture and Moxibustion Bioinformatics, Education Department of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
19
|
Jammal Salameh L, Bitzenhofer SH, Hanganu-Opatz IL, Dutschmann M, Egger V. Blood pressure pulsations modulate central neuronal activity via mechanosensitive ion channels. Science 2024; 383:eadk8511. [PMID: 38301001 DOI: 10.1126/science.adk8511] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/11/2023] [Indexed: 02/03/2024]
Abstract
The transmission of the heartbeat through the cerebral vascular system causes intracranial pressure pulsations. We discovered that arterial pressure pulsations can directly modulate central neuronal activity. In a semi-intact rat brain preparation, vascular pressure pulsations elicited correlated local field oscillations in the olfactory bulb mitral cell layer. These oscillations did not require synaptic transmission but reflected baroreceptive transduction in mitral cells. This transduction was mediated by a fast excitatory mechanosensitive ion channel and modulated neuronal spiking activity. In awake animals, the heartbeat entrained the activity of a subset of olfactory bulb neurons within ~20 milliseconds. Thus, we propose that this fast, intrinsic interoceptive mechanism can modulate perception-for example, during arousal-within the olfactory bulb and possibly across various other brain areas.
Collapse
Affiliation(s)
- Luna Jammal Salameh
- Neurophysiology Group, Zoological Institute, Regensburg University, 93040 Regensburg, Germany
| | - Sebastian H Bitzenhofer
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Mathias Dutschmann
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Veronica Egger
- Neurophysiology Group, Zoological Institute, Regensburg University, 93040 Regensburg, Germany
| |
Collapse
|
20
|
Mierke CT. Extracellular Matrix Cues Regulate Mechanosensing and Mechanotransduction of Cancer Cells. Cells 2024; 13:96. [PMID: 38201302 PMCID: PMC10777970 DOI: 10.3390/cells13010096] [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: 11/12/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
Extracellular biophysical properties have particular implications for a wide spectrum of cellular behaviors and functions, including growth, motility, differentiation, apoptosis, gene expression, cell-matrix and cell-cell adhesion, and signal transduction including mechanotransduction. Cells not only react to unambiguously mechanical cues from the extracellular matrix (ECM), but can occasionally manipulate the mechanical features of the matrix in parallel with biological characteristics, thus interfering with downstream matrix-based cues in both physiological and pathological processes. Bidirectional interactions between cells and (bio)materials in vitro can alter cell phenotype and mechanotransduction, as well as ECM structure, intentionally or unintentionally. Interactions between cell and matrix mechanics in vivo are of particular importance in a variety of diseases, including primarily cancer. Stiffness values between normal and cancerous tissue can range between 500 Pa (soft) and 48 kPa (stiff), respectively. Even the shear flow can increase from 0.1-1 dyn/cm2 (normal tissue) to 1-10 dyn/cm2 (cancerous tissue). There are currently many new areas of activity in tumor research on various biological length scales, which are highlighted in this review. Moreover, the complexity of interactions between ECM and cancer cells is reduced to common features of different tumors and the characteristics are highlighted to identify the main pathways of interaction. This all contributes to the standardization of mechanotransduction models and approaches, which, ultimately, increases the understanding of the complex interaction. Finally, both the in vitro and in vivo effects of this mechanics-biology pairing have key insights and implications for clinical practice in tumor treatment and, consequently, clinical translation.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Science, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
| |
Collapse
|
21
|
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
|
22
|
Mao F, Yang W. How Merkel cells transduce mechanical stimuli: A biophysical model of Merkel cells. PLoS Comput Biol 2023; 19:e1011720. [PMID: 38117763 PMCID: PMC10732429 DOI: 10.1371/journal.pcbi.1011720] [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: 07/02/2023] [Accepted: 11/27/2023] [Indexed: 12/22/2023] Open
Abstract
Merkel cells combine with Aβ afferents, producing slowly adapting type 1(SA1) responses to mechanical stimuli. However, how Merkel cells transduce mechanical stimuli into neural signals to Aβ afferents is still unclear. Here we develop a biophysical model of Merkel cells for mechanical transduction by incorporating main ingredients such as Ca2+ and K+ voltage-gated channels, Piezo2 channels, internal Ca2+ stores, neurotransmitters release, and cell deformation. We first validate our model with several experiments. Then we reveal that Ca2+ and K+ channels on the plasma membrane shape the depolarization of membrane potentials, further regulating the Ca2+ transients in the cells. We also show that Ca2+ channels on the plasma membrane mainly inspire the Ca2+ transients, while internal Ca2+ stores mainly maintain the Ca2+ transients. Moreover, we show that though Piezo2 channels are rapidly adapting mechanical-sensitive channels, they are sufficient to inspire sustained Ca2+ transients in Merkel cells, which further induce the release of neurotransmitters for tens of seconds. Thus our work provides a model that captures the membrane potentials and Ca2+ transients features of Merkel cells and partly explains how Merkel cells transduce the mechanical stimuli by Piezo2 channels.
Collapse
Affiliation(s)
- Fangtao Mao
- Research Center for Humanoid Sensing, Intelligent Perception Research Institute of Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Wenzhen Yang
- Research Center for Humanoid Sensing, Intelligent Perception Research Institute of Zhejiang Lab, Hangzhou, Zhejiang, China
| |
Collapse
|
23
|
Zhou Z, Martinac B. Mechanisms of PIEZO Channel Inactivation. Int J Mol Sci 2023; 24:14113. [PMID: 37762415 PMCID: PMC10531961 DOI: 10.3390/ijms241814113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
PIEZO channels PIEZO1 and PIEZO2 are the newly identified mechanosensitive, non-selective cation channels permeable to Ca2+. In higher vertebrates, PIEZO1 is expressed ubiquitously in most tissues and cells while PIEZO2 is expressed more specifically in the peripheral sensory neurons. PIEZO channels contribute to a wide range of biological behaviors and developmental processes, therefore driving significant attention in the effort to understand their molecular properties. One prominent property of PIEZO channels is their rapid inactivation, which manifests itself as a decrease in channel open probability in the presence of a sustained mechanical stimulus. The lack of the PIEZO channel inactivation is linked to various mechanopathologies emphasizing the significance of studying this PIEZO channel property and the factors affecting it. In the present review, we discuss the mechanisms underlying the PIEZO channel inactivation, its modulation by the interaction of the channels with lipids and/or proteins, and how the changes in PIEZO inactivation by the channel mutations can cause a variety of diseases in animals and humans.
Collapse
Affiliation(s)
- Zijing Zhou
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Darlinghurst, NSW 2010, Australia;
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Darlinghurst, NSW 2010, Australia;
- St Vincent’s Clinical School, University of New South Wales, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
24
|
Ishizawa R, Estrada JA, Kim HK, Hotta N, Fukazawa A, Iwamoto GA, Smith SA, Vongpatanasin W, Mizuno M. Neural discharge of muscle afferents and pressor response to mechanical stimulation are associated with muscle deformation velocity in rats. Am J Physiol Regul Integr Comp Physiol 2023; 325:R13-R20. [PMID: 37067428 PMCID: PMC10259846 DOI: 10.1152/ajpregu.00327.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 04/18/2023]
Abstract
Skeletal muscle reflexes play a crucial role in determining the magnitude of the cardiovascular response to exercise. However, evidence supporting an association between the magnitude of the pressor response and the velocity of muscle deformation has remained to be elucidated. Thus, we investigated the impact of different muscle deformation rates on the neural discharge of muscle afferents and pressor and sympathetic responses in Sprague-Dawley rats. In an ex vivo muscle-nerve preparation, action potentials elicited by sinusoidal mechanical stimuli (137 mN) at different frequencies (0.01, 0.05, 0.1, 0.2, and 0.25 Hz) were recorded in mechanosensitive group III and IV fibers. The afferent response magnitude to sine-wave stimulation significantly varied at different frequencies (ANOVA, P = 0.01). Specifically, as compared with 0.01 Hz (0.83 ± 0.96 spikes/s), the response magnitudes were significantly greater at 0.20 Hz (4.07 ± 5.04 spikes/s, P = 0.031) and 0.25 Hz (4.91 ± 5.30 spikes/s, P = 0.014). In an in vivo decerebrated rat preparation, renal sympathetic nerve activity (RSNA) and mean arterial pressure (MAP) responses to passive stretch (1 kg) of hindlimb skeletal muscle at different velocities of loading (slow, medium, and fast) were measured. Pressor responses to passive stretch were significantly associated with the velocity of muscle deformation (ANOVA, P < 0.001). The MAP response to fast stretch (Δ 56 ± 12 mmHg) was greater than slow (Δ 33 ± 11 mmHg, P = 0.006) or medium (Δ 30 ± 11 mmHg, P < 0.001) stretch. Likewise, the RSNA response was related to deformation velocity (ANOVA, P = 0.024). These findings suggest that the muscle neural afferent discharge and the cardiovascular response to mechanical stimulation are associated with muscle deformation velocity.
Collapse
Affiliation(s)
- Rie Ishizawa
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Juan A Estrada
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Han-Kyul Kim
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Norio Hotta
- College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Ayumi Fukazawa
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Gary A Iwamoto
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Scott A Smith
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Wanpen Vongpatanasin
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Masaki Mizuno
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
25
|
Wijerathne TD, Ozkan AD, Lacroix JJ. Microscopic mechanism of PIEZO1 activation by pressure-induced membrane stretch. J Gen Physiol 2023; 155:213842. [PMID: 36715688 PMCID: PMC9930135 DOI: 10.1085/jgp.202213260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/16/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
Mechanosensitive PIEZO1 ion channels open in response to membrane stretch. Yet, the underlying microscopic mechanism of this activation remains unknown. To probe this mechanism, we used cell-attached pressure-clamp recordings to measure single channel currents at different steady-state negative pipette pressures, spanning the full range of the channel's pressure sensitivity. Pressure-dependent activation occurs through a sharp reduction of the mean shut duration and through a moderate increase of the mean open duration. Across all tested pressures, the distribution of open and shut dwell times best follows sums of two and three exponential components, respectively. As the magnitude of the pressure stimulus increases, the time constants of most of these exponential components gradually change, in opposite directions for open and shut dwell times, and to a similar extent. In addition, while the relative amplitudes of fast and slow components remain unchanged for open intervals, they fully reverse for shut intervals, further reducing the mean shut duration. Using two-dimensional dwell time analysis, Markov-chain modeling, and simulations, we identified a minimal five-states model which recapitulates essential characteristics of single channel data, including microscopic reversibility, correlations between adjacent open and shut intervals, and asymmetric modulation of dwell times by pressure. This study identifies a microscopic mechanism for the activation of PIEZO1 channels by pressure-induced membrane stretch and deepens our fundamental understanding of mechanotransduction by a vertebrate member of the PIEZO channel family.
Collapse
Affiliation(s)
- Tharaka D Wijerathne
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, CA, USA
| | - Alper D Ozkan
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, CA, USA
| | - Jérôme J Lacroix
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, CA, USA
| |
Collapse
|
26
|
Hsiao MY, Liao D, Xiang G, Zhong P. Intercellular Calcium Waves and Permeability Change Induced by Vertically Deployed Surface Acoustic Waves in a Human Cerebral Microvascular Endothelial Cell Line (hCMEC/D3) Monolayer. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1153-1163. [PMID: 36764884 PMCID: PMC10050144 DOI: 10.1016/j.ultrasmedbio.2022.12.019] [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: 08/12/2022] [Revised: 11/28/2022] [Accepted: 12/29/2022] [Indexed: 05/11/2023]
Abstract
OBJECTIVE The ultrasound-mediated blood-brain barrier (BBB) opening with microbubbles has been widely employed, while recent studies also indicate the possibility that ultrasound alone can open the BBB through a direct mechanical effect. However, the exact mechanisms through which ultrasound interacts with the BBB and whether it can directly trigger intracellular signaling and a permeability change in the BBB endothelium remain unclear. METHODS Vertically deployed surface acoustic waves (VD-SAWs) were applied on a human cerebral microvascular endothelial cell line (hCMEC/D3) monolayer using a 33-MHz interdigital transducer that exerts shear stress-predominant stimulation. The intracellular calcium response was measured by fluorescence imaging, and the permeability of the hCMEC/D3 monolayer was assessed by transendothelial electrical resistance (TEER). DISCUSSION At a certain intensity threshold, VD-SAWs induced an intracellular calcium surge that propagated to adjacent cells as intercellular calcium waves. VD-SAWs induced a TEER decrease in a pulse repetition frequency-dependent manner, thereby suggesting possible involvement of the mechanosensitive ion channels. CONCLUSION The unique VD-SAW system enables more physiological mechanical stimulation of the endothelium monolayer. Moreover, it can be easily combined with other measurement devices, providing a useful platform for further mechanistic studies on ultrasound-mediated BBB opening.
Collapse
Affiliation(s)
- Ming-Yen Hsiao
- Department of Physical Medicine and Rehabilitation, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Taipei, Taiwan.
| | - Defei Liao
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Gaoming Xiang
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Pei Zhong
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| |
Collapse
|
27
|
Madar J, Tiwari N, Smith C, Sharma D, Shen S, Elmahdi A, Qiao LY. Piezo2 regulates colonic mechanical sensitivity in a sex specific manner in mice. Nat Commun 2023; 14:2158. [PMID: 37061508 PMCID: PMC10105732 DOI: 10.1038/s41467-023-37683-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/27/2023] [Indexed: 04/17/2023] Open
Abstract
The mechanosensitive ion channel Piezo2 in mucosa and primary afferents transduces colonic mechanical sensation. Here we show that chemogenetic activation or nociceptor-targeted deletion of Piezo2 is sufficient to regulate colonic mechanical sensitivity in a sex dependent manner. Clozapine N-oxide-induced activation of Piezo2;hM3Dq-expressing sensory neurons evokes colonic hypersensitivity in male mice, and causes dyspnea in female mice likely due to effects on lung sensory neurons. Activation of Piezo2-expressing colonic afferent neurons also induces colonic hypersensitivity in male but not female mice. Piezo2 levels in nociceptive neurons are higher in female than in male mice. We also show that Piezo2 conditional deletion from nociceptive neurons increases body weight growth, slows colonic transits, and reduces colonic mechanosensing in female but not male mice. Piezo2 deletion blocks colonic hypersensitivity in male but not female mice. These results suggest that Piezo2 in nociceptive neurons mediates innocuous colonic mechanosensing in female mice and painful sensation in male mice, suggesting a sexual dimorphism of Piezo2 function in the colonic sensory system.
Collapse
Affiliation(s)
- Jonathan Madar
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Namrata Tiwari
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Cristina Smith
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Divya Sharma
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Shanwei Shen
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Alsiddig Elmahdi
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Liya Y Qiao
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
28
|
Wang C, Ramahdita G, Genin G, Huebsch N, Ma Z. Dynamic mechanobiology of cardiac cells and tissues: Current status and future perspective. BIOPHYSICS REVIEWS 2023; 4:011314. [PMID: 37008887 PMCID: PMC10062054 DOI: 10.1063/5.0141269] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/08/2023] [Indexed: 03/31/2023]
Abstract
Mechanical forces impact cardiac cells and tissues over their entire lifespan, from development to growth and eventually to pathophysiology. However, the mechanobiological pathways that drive cell and tissue responses to mechanical forces are only now beginning to be understood, due in part to the challenges in replicating the evolving dynamic microenvironments of cardiac cells and tissues in a laboratory setting. Although many in vitro cardiac models have been established to provide specific stiffness, topography, or viscoelasticity to cardiac cells and tissues via biomaterial scaffolds or external stimuli, technologies for presenting time-evolving mechanical microenvironments have only recently been developed. In this review, we summarize the range of in vitro platforms that have been used for cardiac mechanobiological studies. We provide a comprehensive review on phenotypic and molecular changes of cardiomyocytes in response to these environments, with a focus on how dynamic mechanical cues are transduced and deciphered. We conclude with our vision of how these findings will help to define the baseline of heart pathology and of how these in vitro systems will potentially serve to improve the development of therapies for heart diseases.
Collapse
Affiliation(s)
| | - Ghiska Ramahdita
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | | | | | - Zhen Ma
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
29
|
Young MN, Sindoni MJ, Lewis AH, Zauscher S, Grandl J. The energetics of rapid cellular mechanotransduction. Proc Natl Acad Sci U S A 2023; 120:e2215747120. [PMID: 36795747 PMCID: PMC9974467 DOI: 10.1073/pnas.2215747120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/18/2023] [Indexed: 02/17/2023] Open
Abstract
Cells throughout the human body detect mechanical forces. While it is known that the rapid (millisecond) detection of mechanical forces is mediated by force-gated ion channels, a detailed quantitative understanding of cells as sensors of mechanical energy is still lacking. Here, we combine atomic force microscopy with patch-clamp electrophysiology to determine the physical limits of cells expressing the force-gated ion channels (FGICs) Piezo1, Piezo2, TREK1, and TRAAK. We find that, depending on the ion channel expressed, cells can function either as proportional or nonlinear transducers of mechanical energy and detect mechanical energies as little as ~100 fJ, with a resolution of up to ~1 fJ. These specific energetic values depend on cell size, channel density, and cytoskeletal architecture. We also make the surprising discovery that cells can transduce forces either nearly instantaneously (<1 ms) or with a substantial time delay (~10 ms). Using a chimeric experimental approach and simulations, we show how such delays can emerge from channel-intrinsic properties and the slow diffusion of tension in the membrane. Overall, our experiments reveal the capabilities and limits of cellular mechanosensing and provide insights into molecular mechanisms that different cell types may employ to specialize for their distinct physiological roles.
Collapse
Affiliation(s)
- Michael N. Young
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
| | - Michael J. Sindoni
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
| | - Amanda H. Lewis
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
| | - Stefan Zauscher
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC27710
| | - Jörg Grandl
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
| |
Collapse
|
30
|
Khatib NS, Monsen J, Ahmed S, Huang Y, Hoey DA, Nowlan NC. Mechanoregulatory role of TRPV4 in prenatal skeletal development. SCIENCE ADVANCES 2023; 9:eade2155. [PMID: 36696489 PMCID: PMC9876556 DOI: 10.1126/sciadv.ade2155] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/22/2022] [Indexed: 06/17/2023]
Abstract
Biophysical cues are essential for guiding skeletal development, but the mechanisms underlying the mechanical regulation of cartilage and bone formation are unknown. TRPV4 is a mechanically sensitive ion channel involved in cartilage and bone cell mechanosensing, mutations of which lead to skeletal developmental pathologies. We tested the hypothesis that loading-driven prenatal skeletal development is dependent on TRPV4 activity. We first establish that mechanically stimulating mouse embryo hindlimbs cultured ex vivo stimulates knee cartilage growth, morphogenesis, and expression of TRPV4, which localizes to areas of high biophysical stimuli. We then demonstrate that loading-driven joint cartilage growth and shape are dependent on TRPV4 activity, mediated via control of cell proliferation and matrix biosynthesis, indicating a mechanism by which mechanical loading could direct growth and morphogenesis during joint formation. We conclude that mechanoregulatory pathways initiated by TRPV4 guide skeletal development; therefore, TRPV4 is a valuable target for the development of skeletal regenerative and repair strategies.
Collapse
Affiliation(s)
- Nidal S. Khatib
- Department of Bioengineering, Imperial College London, London, UK
| | - James Monsen
- Department of Bioengineering, Imperial College London, London, UK
| | - Saima Ahmed
- Department of Bioengineering, Imperial College London, London, UK
| | - Yuming Huang
- Department of Bioengineering, Imperial College London, London, UK
| | - David A. Hoey
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Niamh C. Nowlan
- Department of Bioengineering, Imperial College London, London, UK
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Mechanical and Materials Engineering, University College Dublin, Dublin, Ireland
- UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
31
|
Ziolkowski LH, Gracheva EO, Bagriantsev SN. Mechanotransduction events at the physiological site of touch detection. eLife 2023; 12:84179. [PMID: 36607222 PMCID: PMC9833821 DOI: 10.7554/elife.84179] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/21/2022] [Indexed: 01/07/2023] Open
Abstract
Afferents of peripheral mechanoreceptors innervate the skin of vertebrates, where they detect physical touch via mechanically gated ion channels (mechanotransducers). While the afferent terminal is generally understood to be the primary site of mechanotransduction, the functional properties of mechanically activated (MA) ionic current generated by mechanotransducers at this location remain obscure. Until now, direct evidence of MA current and mechanically induced action potentials in the mechanoreceptor terminal has not been obtained. Here, we report patch-clamp recordings from the afferent terminal innervating Grandry (Meissner) corpuscles in the bill skin of a tactile specialist duck. We show that mechanical stimulation evokes MA current in the afferent with fast kinetics of activation and inactivation during the dynamic phases of the mechanical stimulus. These responses trigger rapidly adapting firing in the afferent detected at the terminal and in the afferent fiber outside of the corpuscle. Our findings elucidate the initial electrogenic events of touch detection in the mechanoreceptor nerve terminal.
Collapse
Affiliation(s)
- Luke H Ziolkowski
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HavenUnited States
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HavenUnited States,Department of Neuroscience, Yale University School of MedicineNew HavenUnited States,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of MedicineNew HavenUnited States,Kavli Institute for Neuroscience, Yale University School of MedicineNew HavenUnited States
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HavenUnited States
| |
Collapse
|
32
|
Mao F, Yang Y, Jiang H. Electromechanical model for object roughness perception during finger sliding. Biophys J 2022; 121:4740-4747. [PMID: 36116008 PMCID: PMC9748192 DOI: 10.1016/j.bpj.2022.09.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/02/2022] [Accepted: 09/13/2022] [Indexed: 12/15/2022] Open
Abstract
Touch allows us to gather abundant information in the world around us. However, how sensory cells embedded in the fingers convey texture information into their firing patterns is still poorly understood. Here, we develop an electromechanical model for roughness perception by incorporating main ingredients such as voltage-gated ion channels, active ion pumps, mechanosensitive channels, and cell deformation. The model reveals that sensory cells can convey texture wavelengths into the period of their firing patterns as the finger slides across object surfaces, but they can only convey a limited range of texture wavelengths. We also show that an increase in sliding speed broadens the decoding wavelength range at the cost of reduction of lower perception limits. Thus, a smaller sliding speed and a bigger contact force may be needed to successfully discern a smooth surface, consistent with previous psychophysical observations. Moreover, we show that cells with slowly adapting mechanosensitive channels can still fire action potentials under static loadings, indicating that slowly adapting mechanosensitive channels may contribute to the perception of coarse textures under static touch. Our work thus provides a new theoretical framework to study roughness perception and may have important implications for the design of electronic skin, artificial touch, and haptic interfaces.
Collapse
Affiliation(s)
- Fangtao Mao
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, CAS Center for Excellence in Complex System Mechanics, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui, China
| | - Yuehua Yang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, CAS Center for Excellence in Complex System Mechanics, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui, China.
| | - Hongyuan Jiang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, CAS Center for Excellence in Complex System Mechanics, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
33
|
Song S, Zhang H, Wang X, Chen W, Cao W, Zhang Z, Shi C. The role of mechanosensitive Piezo1 channel in diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 172:39-49. [PMID: 35436566 DOI: 10.1016/j.pbiomolbio.2022.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 06/14/2023]
Abstract
Mechanotransduction is associated with organ development and homoeostasis. Piezo1 and Piezo2 are novel mechanosensitive ion channels (MSCs) in mammals. MSCs are membrane proteins that are critical for the mechanotransduction of living cells. Current studies have demonstrated that the Piezo protein family not only functions in volume regulation, cellular migration, proliferation, and apoptosis but is also important for human diseases of various systems. The complete loss of Piezo1 and Piezo2 function is fatal in the embryonic period. This review summarizes the role of Piezo1 in diseases of different systems and perspectives potential treatments related to Piezo1 for these diseases.
Collapse
Affiliation(s)
- Siqi Song
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Hong Zhang
- Department of Cardiac Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Xiaoya Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Wei Chen
- Department of Urology, The Affiliated Xinqiao Hospital, The Third Military Medical University, Chongqing, 400038, China
| | - Wenxuan Cao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Zhe Zhang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao 266071, Shandong Province, China.
| | - Chunying Shi
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China.
| |
Collapse
|
34
|
Young M, Lewis AH, Grandl J. Physics of mechanotransduction by Piezo ion channels. J Gen Physiol 2022; 154:213231. [PMID: 35593732 PMCID: PMC9127981 DOI: 10.1085/jgp.202113044] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/28/2022] [Accepted: 04/29/2022] [Indexed: 12/26/2022] Open
Abstract
Piezo ion channels are sensors of mechanical forces and mediate a wide range of physiological mechanotransduction processes. More than a decade of intense research has elucidated much of the structural and mechanistic principles underlying Piezo gating and its roles in physiology, although wide gaps of knowledge continue to exist. Here, we review the forces and energies involved in mechanical activation of Piezo ion channels and their functional modulation by other chemical and physical stimuli including lipids, voltage, and temperature. We compare the three predominant mechanisms likely to explain Piezo activation—the force-from-lipids mechanism, the tether model, and the membrane footprint theory. Additional sections shine light on how Piezo ion channels may affect each other through spatial clustering and functional cooperativity, and how substantial functional heterogeneity of Piezo ion channels arises as a byproduct of the precise physical environment each channel experiences. Finally, our review concludes by pointing out major research questions and technological limitations that future research can address.
Collapse
Affiliation(s)
- Michael Young
- Department of Neurobiology, Duke University Medical Center, Durham, NC
| | - Amanda H Lewis
- Department of Neurobiology, Duke University Medical Center, Durham, NC
| | - Jörg Grandl
- Department of Neurobiology, Duke University Medical Center, Durham, NC
| |
Collapse
|
35
|
Nagel M, Chesler AT. PIEZO2 ion channels in proprioception. Curr Opin Neurobiol 2022; 75:102572. [PMID: 35689908 DOI: 10.1016/j.conb.2022.102572] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/28/2022] [Accepted: 05/06/2022] [Indexed: 12/18/2022]
Abstract
PIEZO2 is a stretch-gated ion channel that is expressed at high levels in somatosensory neurons. Humans with rare mutations in the PIEZO2 gene have profound mechanosensory deficits that include a loss of the sense of proprioception. These striking phenotypes match those seen in conditional knockout mouse models demonstrating the highly conserved function for this gene. Here, we review the ramifications of loss of PIEZO2 function on normal daily activities and what studies like these have revealed about proprioception at the molecular and cellular level. Additionally, we highlight recent work that has uncovered the surprising functional and molecular diversity of proprioceptors. Together, these findings pioneer a path toward determining how the detection of mechanosensory input from muscles and tendons is used to control posture and refine motor performance.
Collapse
Affiliation(s)
- Maximilian Nagel
- Sensory Cells and Circuits Section, National Center for Complementary and Integrative Health, 35 Convent Drive, Bethesda, MD, 20892, USA
| | - Alexander T Chesler
- Sensory Cells and Circuits Section, National Center for Complementary and Integrative Health, 35 Convent Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
36
|
Zhang Z, Sa G, Wang Z, Wei Z, Zheng L, Zhang R, Zhu X, Yang X. Piezo1 and IFT88 synergistically regulate mandibular condylar chondrocyte differentiation under cyclic tensile strain. Tissue Cell 2022; 76:101781. [PMID: 35279604 DOI: 10.1016/j.tice.2022.101781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/02/2022] [Accepted: 03/05/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVE(S) Mandibular condyle chondrocytes (MCCs) are exposed to various mechanical environments. Primary cilia, as a carrier for ion channels, can sense mechanical signals. Intraflagellar transport protein 88 (IFT88) is crucial for the assembly and function of primary cilia. Piezo1 is a mechanically activated ion channel that mediates mechanical signal transduction. This study aimed to identify the possible synergistic effect between Piezo1 and IFT88 in MCC differentiation during mechanical conduction. MATERIALS AND METHODS Confocal immunofluorescence staining was used to reveal the Piezo1 localization. Small interfering RNA (siRNA) technology was used to knock down the expression levels of Piezo1 and IFT88. The chondrogenic differentiation ability of MCCs was evaluated by Alcian blue staining, and the early differentiation ability was evaluated by Western blot of SOX9 and COL2A1. RESULTS Confocal immunofluorescence results showed that Piezo1 localized in the root of primary cilia. Without cyclic tensile strain (CTS) stimuli, Alcian blue staining showed that Piezo1 knockdown had a marginal effect on the chondrogenic differentiation of MCCs, while IFT88 knockdown inhibited the chondrogenic differentiation. The protein levels of SOX9 and COL2A1 decreased significantly with CTS stimuli. However, these protein levels were restored when Piezo1 was knocked down. In addition, IFT88 knockdown decreased the protein level of Piezo1 with or without CTS. CONCLUSION Piezo1 and IFT88 might play a synergistic role in regulating MCC differentiation under CTS stimuli.
Collapse
Affiliation(s)
- Zhuoyu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Guoliang Sa
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China; Department of Orthognathic & Cleft Lip and Palate Plastic Surgery, Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Zhuo Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Zequan Wei
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China; Hebei Med Univ, Hosp 3, Dept Oral & Maxillofacial Surg, Shijiazhuang, Hebei, PR China
| | - Liwu Zheng
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Ruochen Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Xinbiao Zhu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Xuewen Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, PR China; Department of Orthognathic & Cleft Lip and Palate Plastic Surgery, Hospital of Stomatology, Wuhan University, Wuhan, PR China.
| |
Collapse
|
37
|
Li X, Hu J, Zhao X, Li J, Chen Y. Piezo channels in the urinary system. Exp Mol Med 2022; 54:697-710. [PMID: 35701561 PMCID: PMC9256749 DOI: 10.1038/s12276-022-00777-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/25/2022] [Accepted: 02/16/2022] [Indexed: 12/24/2022] Open
Abstract
The Piezo channel family, including Piezo1 and Piezo2, includes essential mechanosensitive transduction molecules in mammals. Functioning in the conversion of mechanical signals to biological signals to regulate a plethora of physiological processes, Piezo channels, which have a unique homotrimeric three-blade propeller-shaped structure, utilize a cap-motion and plug-and-latch mechanism to gate their ion-conducting pathways. Piezo channels have a wide range of biological roles in various human systems, both in vitro and in vivo. Currently, there is a lack of comprehensive understanding of their antagonists and agonists, and therefore further investigation is needed. Remarkably, increasingly compelling evidence demonstrates that Piezo channel function in the urinary system is important. This review article systematically summarizes the existing evidence of the importance of Piezo channels, including protein structure, mechanogating mechanisms, and pharmacological characteristics, with a particular focus on their physiological and pathophysiological roles in the urinary system. Collectively, this review aims to provide a direction for future clinical applications in urinary system diseases.
Collapse
Affiliation(s)
- Xu Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Junwei Hu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xuedan Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Juanjuan Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yuelai Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
38
|
He J, Li B, Han S, Zhang Y, Liu K, Yi S, Liu Y, Xiu M. Drosophila as a Model to Study the Mechanism of Nociception. Front Physiol 2022; 13:854124. [PMID: 35418874 PMCID: PMC8996152 DOI: 10.3389/fphys.2022.854124] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/28/2022] [Indexed: 12/15/2022] Open
Abstract
Nociception refers to the process of encoding and processing noxious stimuli, which allow animals to detect and avoid potentially harmful stimuli. Several types of stimuli can trigger nociceptive sensory transduction, including thermal, noxious chemicals, and harsh mechanical stimulation that depend on the corresponding nociceptors. In view of the high evolutionary conservation of the mechanisms that govern nociception from Drosophila melanogaster to mammals, investigation in the fruit fly Drosophila help us understand how the sensory nervous system works and what happen in nociception. Here, we present an overview of currently identified conserved genetics of nociception, the nociceptive sensory neurons responsible for detecting noxious stimuli, and various assays for evaluating different nociception. Finally, we cover development of anti-pain drug using fly model. These comparisons illustrate the value of using Drosophila as model for uncovering nociception mechanisms, which are essential for identifying new treatment goals and developing novel analgesics that are applicable to human health.
Collapse
Affiliation(s)
- Jianzheng He
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China
| | - Botong Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shuzhen Han
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yuan Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Kai Liu
- College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Simeng Yi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yongqi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China
- *Correspondence: Yongqi Liu,
| | - Minghui Xiu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
- Minghui Xiu,
| |
Collapse
|
39
|
Shinge SAU, Zhang D, Din AU, Yu F, Nie Y. Emerging Piezo1 signaling in inflammation and atherosclerosis; a potential therapeutic target. Int J Biol Sci 2022; 18:923-941. [PMID: 35173527 PMCID: PMC8771847 DOI: 10.7150/ijbs.63819] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/08/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose of Review: Atherosclerosis is the principal cause of cardiovascular diseases (CVDs) which are the major cause of death worldwide. Mechanical force plays an essential role in cardiovascular health and disease. To bring the awareness of mechanosensitive Piezo1 role in atherosclerosis and its therapeutic potentials we review recent literature to highlight its involvement in various mechanisms of the disease. Recent Findings: Recent studies reported Piezo1 channel as a sensor, and transducer of various mechanical forces into biochemical signals, which affect various cellular activities such as proliferation, migration, apoptosis and vascular remodeling including immune/inflammatory mechanisms fundamental phenomenon in atherogenesis. Summary: Numerous evidences suggest Piezo1 as a player in different mechanisms of cell biology, including immune/inflammatory and other cellular mechanisms correlated with atherosclerosis. This review discusses mechanistic insight about this matter and highlights the drugability and therapeutic potentials consistent with emerging functions Piezo1 in various mechanisms of atherosclerosis. Based on the recent works, we suggest Piezo1 as potential therapeutic target and a valid candidate for future research. Therefore, a deeper exploration of Piezo1 biology and translation towards the clinic will be a novel strategy for treating atherosclerosis and other CVDs.
Collapse
Affiliation(s)
- Shafiu A. Umar Shinge
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
| | - Daifang Zhang
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Clinical Research Center, Southwest Medical University, Luzhou, Sichuan PRC
| | - Ahmad Ud Din
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan PRC
| | - FengXu Yu
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, Sichuan PRC
| | - YongMei Nie
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, Sichuan PRC
| |
Collapse
|
40
|
Foote AG, Lungova V, Thibeault SL. Piezo1-expressing vocal fold epithelia modulate remodeling via effects on self-renewal and cytokeratin differentiation. Cell Mol Life Sci 2022; 79:591. [PMID: 36376494 PMCID: PMC9663367 DOI: 10.1007/s00018-022-04622-6] [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: 06/15/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
Mechanoreceptors are implicated as functional afferents within mucosa of the airways and the recent discovery of mechanosensitive channels Piezo1 and Piezo2 has proved essential for cells of various mechanically sensitive tissues. However, the role for Piezo1/2 in vocal fold (VF) mucosal epithelia, a cell that withstands excessive biomechanical insult, remains unknown. The purpose of this study was to test the hypothesis that Piezo1 is required for VF mucosal repair pathways of epithelial cell injury. Utilizing a sonic hedgehog (shh) Cre line for epithelial-specific ablation of Piezo1/2 mechanoreceptors, we investigated 6wk adult VF mucosa following naphthalene exposure for repair strategies at 1, 3, 7 and 14 days post-injury (dpi). PIEZO1 localized to differentiated apical epithelia and was paramount for epithelial remodeling events. Injury to wildtype epithelium was most appreciated at 3 dpi. Shhcre/+; Piezo1loxP/loxP, Piezo2 loxP/+ mutant epithelium exhibited severe cell/nuclear defects compared to injured controls. Conditional ablation of Piezo1 and/or Piezo2 to uninjured VF epithelium did not result in abnormal phenotypes across P0, P15 and 6wk postnatal stages compared to heterozygote and control tissue. Results demonstrate a role for Piezo1-expressing VF epithelia in regulating self-renewal via effects on p63 transcription and YAP subcellular translocation-altering cytokeratin differentiation.
Collapse
Affiliation(s)
- Alexander G. Foote
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of Wisconsin-Madison, Wisconsin, USA
| | - Vlasta Lungova
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of Wisconsin-Madison, Wisconsin, USA
| | - Susan L. Thibeault
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of Wisconsin-Madison, Wisconsin, USA
| |
Collapse
|
41
|
Zhang K, Wang L, Liu Z, Geng B, Teng Y, Liu X, Yi Q, Yu D, Chen X, Zhao D, Xia Y. Mechanosensory and mechanotransductive processes mediated by ion channels in articular chondrocytes: Potential therapeutic targets for osteoarthritis. Channels (Austin) 2021; 15:339-359. [PMID: 33775217 PMCID: PMC8018402 DOI: 10.1080/19336950.2021.1903184] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
Articular cartilage consists of an extracellular matrix including many proteins as well as embedded chondrocytes. Articular cartilage formation and function are influenced by mechanical forces. Hind limb unloading or simulated microgravity causes articular cartilage loss, suggesting the importance of the healthy mechanical environment in articular cartilage homeostasis and implying a significant role of appropriate mechanical stimulation in articular cartilage degeneration. Mechanosensitive ion channels participate in regulating the metabolism of articular chondrocytes, including matrix protein production and extracellular matrix synthesis. Mechanical stimuli, including fluid shear stress, stretch, compression and cell swelling and decreased mechanical conditions (such as simulated microgravity) can alter the membrane potential and regulate the metabolism of articular chondrocytes via transmembrane ion channel-induced ionic fluxes. This process includes Ca2+ influx and the resulting mobilization of Ca2+ that is due to massive released Ca2+ from stores, intracellular cation efflux and extracellular cation influx. This review brings together published information on mechanosensitive ion channels, such as stretch-activated channels (SACs), voltage-gated Ca2+ channels (VGCCs), large conductance Ca2+-activated K+ channels (BKCa channels), Ca2+-activated K+ channels (SKCa channels), voltage-activated H+ channels (VAHCs), acid sensing ion channels (ASICs), transient receptor potential (TRP) family channels, and piezo1/2 channels. Data based on epithelial sodium channels (ENaCs), purinergic receptors and N-methyl-d-aspartate (NMDA) receptors are also included. These channels mediate mechanoelectrical physiological processes essential for converting physical force signals into biological signals. The primary channel-mediated effects and signaling pathways regulated by these mechanosensitive ion channels can influence the progression of osteoarthritis during the mechanosensory and mechanoadaptive process of articular chondrocytes.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Lifu Wang
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Zhongcheng Liu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Bin Geng
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Yuanjun Teng
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Xuening Liu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Qiong Yi
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Dechen Yu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Xiangyi Chen
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Dacheng Zhao
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Yayi Xia
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| |
Collapse
|
42
|
Lewis AH, Grandl J. Piezo1 ion channels inherently function as independent mechanotransducers. eLife 2021; 10:70988. [PMID: 34711306 PMCID: PMC8555984 DOI: 10.7554/elife.70988] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/07/2021] [Indexed: 01/25/2023] Open
Abstract
Piezo1 is a mechanically activated ion channel involved in sensing forces in various cell types and tissues. Cryo-electron microscopy has revealed that the Piezo1 structure is bowl-shaped and capable of inducing membrane curvature via its extended footprint, which indirectly suggests that Piezo1 ion channels may bias each other’s spatial distribution and interact functionally. Here, we use cell-attached patch-clamp electrophysiology and pressure-clamp stimulation to functionally examine large numbers of membrane patches from cells expressing Piezo1 endogenously at low levels and cells overexpressing Piezo1 at high levels. Our data, together with stochastic simulations of Piezo1 spatial distributions, show that both at endogenous densities (1–2 channels/μm2), and at non-physiological densities (10–100 channels/μm2) predicted to cause substantial footprint overlap, Piezo1 density has no effect on its pressure sensitivity or open probability in the nominal absence of membrane tension. The results suggest that Piezo channels, at densities likely to be physiologically relevant, inherently behave as independent mechanotransducers. We propose that this property is essential for cells to transduce forces homogeneously across the entire cell membrane. Cells can sense a range of mechanical forces both inside and outside the body, such as the stroke of a fingertip or the filling of a lung. Pores on the surface of the cell called Piezo channels open up in response to this pressure. This allows ions to flood in to the cell and trigger a series of biochemical reactions that alter the cell’s behavior. Piezo channels have a unique bowl-like structure that transforms the shape of the cell surface around them, potentially affecting how nearby proteins behave. Previous research had suggested that these channels might be unevenly distributed across the cell surface, and were predicted to modify each other’s behaviors when tightly packed together. This cooperative response would have a significant impact on how cells sense mechanical force. To investigate if this was the case, Lewis and Grandl studied a mouse cell called Neuro2A which naturally produces Piezo ion channels. In the experiment, pressure was applied to different parts of the cell and the electric current generated by ions moving across the surface was recorded: the higher the electrical activity, the more ion channels present. This showed that Piezo channels are randomly distributed across the cell surface and do not tend to cluster together. The same Neuro2A cells were then engineered to produce up to one hundred times more Piezo proteins. Despite the channels being more densely packed together, how they responded to mechanical force remained the same. These results suggest that Piezo channels act independently and are not influenced by close proximity to one another. Lewis and Grandl propose that this property may ensure that all parts of the cell surface react to mechanical force in the same way. Further work is needed to see if this finding applies to other cell types that produce Piezo proteins.
Collapse
Affiliation(s)
- Amanda H Lewis
- Department of Neurobiology, Duke University Medical Center, Durham, United States
| | - Jörg Grandl
- Department of Neurobiology, Duke University Medical Center, Durham, United States
| |
Collapse
|
43
|
Turner D, Kang C, Mesirca P, Hong J, Mangoni ME, Glukhov AV, Sah R. Electrophysiological and Molecular Mechanisms of Sinoatrial Node Mechanosensitivity. Front Cardiovasc Med 2021; 8:662410. [PMID: 34434970 PMCID: PMC8382116 DOI: 10.3389/fcvm.2021.662410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/24/2021] [Indexed: 01/01/2023] Open
Abstract
The understanding of the electrophysiological mechanisms that underlie mechanosensitivity of the sinoatrial node (SAN), the primary pacemaker of the heart, has been evolving over the past century. The heart is constantly exposed to a dynamic mechanical environment; as such, the SAN has numerous canonical and emerging mechanosensitive ion channels and signaling pathways that govern its ability to respond to both fast (within second or on beat-to-beat manner) and slow (minutes) timescales. This review summarizes the effects of mechanical loading on the SAN activity and reviews putative candidates, including fast mechanoactivated channels (Piezo, TREK, and BK) and slow mechanoresponsive ion channels [including volume-regulated chloride channels and transient receptor potential (TRP)], as well as the components of mechanochemical signal transduction, which may contribute to SAN mechanosensitivity. Furthermore, we examine the structural foundation for both mechano-electrical and mechanochemical signal transduction and discuss the role of specialized membrane nanodomains, namely, caveolae, in mechanical regulation of both membrane and calcium clock components of the so-called coupled-clock pacemaker system responsible for SAN automaticity. Finally, we emphasize how these mechanically activated changes contribute to the pathophysiology of SAN dysfunction and discuss controversial areas necessitating future investigations. Though the exact mechanisms of SAN mechanosensitivity are currently unknown, identification of such components, their impact into SAN pacemaking, and pathological remodeling may provide new therapeutic targets for the treatment of SAN dysfunction and associated rhythm abnormalities.
Collapse
Affiliation(s)
- Daniel Turner
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Chen Kang
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Juan Hong
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Alexey V Glukhov
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Rajan Sah
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
44
|
Decoding mechanical cues by molecular mechanotransduction. Curr Opin Cell Biol 2021; 72:72-80. [PMID: 34218181 DOI: 10.1016/j.ceb.2021.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/18/2021] [Accepted: 05/28/2021] [Indexed: 12/29/2022]
Abstract
Cells are exposed to a variety of mechanical cues, including forces from their local environment and physical properties of the tissue. These mechanical cues regulate a vast number of cellular processes, relying on a repertoire of mechanosensors that transduce forces into biochemical pathways through mechanotransduction. Forces can act on different parts of the cell, carry information regarding magnitude and direction, and have distinct temporal profiles. Thus, the specific cellular response to mechanical forces is dependent on the ability of cells to sense and transduce these physical parameters. In this review, we will highlight recent findings that provide insights into the mechanisms by which different mechanosensors decode mechanical cues and how their coordinated response determines the cellular outcomes.
Collapse
|
45
|
Abstract
Mechanosensation is the ability to detect dynamic mechanical stimuli (e.g., pressure, stretch, and shear stress) and is essential for a wide variety of processes, including our sense of touch on the skin. How touch is detected and transduced at the molecular level has proved to be one of the great mysteries of sensory biology. A major breakthrough occurred in 2010 with the discovery of a family of mechanically gated ion channels that were coined PIEZOs. The last 10 years of investigation have provided a wealth of information about the functional roles and mechanisms of these molecules. Here we focus on PIEZO2, one of the two PIEZO proteins found in humans and other mammals. We review how work at the molecular, cellular, and systems levels over the past decade has transformed our understanding of touch and led to unexpected insights into other types of mechanosensation beyond the skin.
Collapse
Affiliation(s)
- Marcin Szczot
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland 20892, USA; .,Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, 583 30 Linköping, Sweden
| | - Alec R Nickolls
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Ruby M Lam
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland 20892, USA; .,NIH-Brown University Graduate Program in Neuroscience, Providence, Rhode Island 02912, USA
| | - Alexander T Chesler
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland 20892, USA; .,National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
46
|
Iskander A, Bilgi C, Naftalovich R, Hacihaliloglu I, Berkman T, Naftalovich D, Pahlevan N. The Rheology of the Carotid Sinus: A Path Toward Bioinspired Intervention. Front Bioeng Biotechnol 2021; 9:678048. [PMID: 34178967 PMCID: PMC8222608 DOI: 10.3389/fbioe.2021.678048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/05/2021] [Indexed: 11/30/2022] Open
Abstract
The association between blood viscosity and pathological conditions involving a number of organ systems is well known. However, how the body measures and maintains appropriate blood viscosity is not well-described. The literature endorsing the function of the carotid sinus as a site of baroreception can be traced back to some of the earliest descriptions of digital pressure on the neck producing a drop in blood delivery to the brain. For the last 30 years, improved computational fluid dynamic (CFD) simulations of blood flow within the carotid sinus have demonstrated a more nuanced understanding of the changes in the region as it relates to changes in conventional metrics of cardiovascular function, including blood pressure. We suggest that the unique flow patterns within the carotid sinus may make it an ideal site to transduce flow data that can, in turn, enable real-time measurement of blood viscosity. The recent characterization of the PIEZO receptor family in the sinus vessel wall may provide a biological basis for this characterization. When coupled with other biomarkers of cardiovascular performance and descriptions of the blood rheology unique to the sinus region, this represents a novel venue for bioinspired design that may enable end-users to manipulate and optimize blood flow.
Collapse
Affiliation(s)
- Andrew Iskander
- Department of Anesthesiology, Westchester Medical Center, New York Medical College, Valhalla, NY, United States
| | - Coskun Bilgi
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, United States
| | - Rotem Naftalovich
- Department of Anesthesiology, New Jersey Medical School, University Hospital, Rutgers University, Newark, NJ, United States.,Medical Corps of the U.S. Army, U.S. Army Medical Department, Fort Sam Houston, San Antonio, TX, United States
| | - Ilker Hacihaliloglu
- Department of Biomedical Engineering, Rutgers School of Engineering, Rutgers University, Piscataway, NJ, United States
| | - Tolga Berkman
- Department of Anesthesiology, New Jersey Medical School, University Hospital, Rutgers University, Newark, NJ, United States
| | - Daniel Naftalovich
- Department of Computational and Mathematical Sciences, California Institute of Technology, Pasadena, CA, United States.,Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Niema Pahlevan
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, United States.,Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
47
|
Kim TH, Jeon WY, Ji Y, Park EJ, Yoon DS, Lee NH, Park SM, Mandakhbayar N, Lee JH, Lee HH, Kim HW. Electricity auto-generating skin patch promotes wound healing process by activation of mechanosensitive ion channels. Biomaterials 2021; 275:120948. [PMID: 34157562 DOI: 10.1016/j.biomaterials.2021.120948] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/20/2021] [Accepted: 05/29/2021] [Indexed: 12/14/2022]
Abstract
Electricity constitutes a natural biophysical component that preserves tissue homeostasis and modulates many biological processes, including the repair of damaged tissues. Wound healing involves intricate cellular events, such as inflammation, angiogenesis, matrix synthesis, and epithelialization whereby multiple cell types sense the environmental cues to rebuild the structure and functions. Here, we report that electricity auto-generating glucose-responsive enzymatic-biofuel-cell (EBC) skin patch stimulates the wound healing process. Rat wounded-skin model and in vitro cell cultures showed that EBC accelerated wound healing by modulating inflammation while stimulating angiogenesis, fibroblast fuctionality and matrix synthesis. Of note, EBC-activated cellular bahaviors were linked to the signalings involved with calcium influx, which predominantly dependent on the mechanosensitive ion channels, primarily Piezo1. Inhibition of Piezo1-receptor impaired the EBC-induced key functions of both fibroblasts and endothelial cells in the wound healing. This study highlights the significant roles of electricity played in wound healing through activated mechanosensitive ion channels and the calcium influx, and suggests the possibility of the electricity auto-generating EBC-based skin patch for use as a wound healing device.
Collapse
Affiliation(s)
- Tae-Hyun Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Won-Yong Jeon
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; School of Chemical Engineering, Biomedical Institute for Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yunseong Ji
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Eun Ju Park
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Institute of Materials Research and Engineering (IMRE), A*STAR, 2 Fusionopolis Way, #08-03 Innovis, 138634, Singapore
| | - Dong Suk Yoon
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Na-Hyun Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Sung-Min Park
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Cheonan, 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea.
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Cheonan, 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
48
|
Jakob D, Klesen A, Allegrini B, Darkow E, Aria D, Emig R, Chica AS, Rog-Zielinska EA, Guth T, Beyersdorf F, Kari FA, Proksch S, Hatem SN, Karck M, Künzel SR, Guizouarn H, Schmidt C, Kohl P, Ravens U, Peyronnet R. Piezo1 and BK Ca channels in human atrial fibroblasts: Interplay and remodelling in atrial fibrillation. J Mol Cell Cardiol 2021; 158:49-62. [PMID: 33974928 DOI: 10.1016/j.yjmcc.2021.05.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 04/18/2021] [Accepted: 05/04/2021] [Indexed: 12/17/2022]
Abstract
AIMS Atrial Fibrillation (AF) is an arrhythmia of increasing prevalence in the aging populations of developed countries. One of the important indicators of AF is sustained atrial dilatation, highlighting the importance of mechanical overload in the pathophysiology of AF. The mechanisms by which atrial cells, including fibroblasts, sense and react to changing mechanical forces, are not fully elucidated. Here, we characterise stretch-activated ion channels (SAC) in human atrial fibroblasts and changes in SAC- presence and activity associated with AF. METHODS AND RESULTS Using primary cultures of human atrial fibroblasts, isolated from patients in sinus rhythm or sustained AF, we combine electrophysiological, molecular and pharmacological tools to identify SAC. Two electrophysiological SAC- signatures were detected, indicative of cation-nonselective and potassium-selective channels. Using siRNA-mediated knockdown, we identified the cation-nonselective SAC as Piezo1. Biophysical properties of the potassium-selective channel, its sensitivity to calcium, paxilline or iberiotoxin (blockers), and NS11021 (activator), indicated presence of calcium-dependent 'big potassium channels' (BKCa). In cells from AF patients, Piezo1 activity and mRNA expression levels were higher than in cells from sinus rhythm patients, while BKCa activity (but not expression) was downregulated. Both Piezo1-knockdown and removal of extracellular calcium from the patch pipette resulted in a significant reduction of BKCa current during stretch. No co-immunoprecipitation of Piezo1 and BKCa was detected. CONCLUSIONS Human atrial fibroblasts contain at least two types of ion channels that are activated during stretch: Piezo1 and BKCa. While Piezo1 is directly stretch-activated, the increase in BKCa activity during mechanical stimulation appears to be mainly secondary to calcium influx via SAC such as Piezo1. During sustained AF, Piezo1 is increased, while BKCa activity is reduced, highlighting differential regulation of both channels. Our data support the presence and interplay of Piezo1 and BKCa in human atrial fibroblasts in the absence of physical links between the two channel proteins.
Collapse
Affiliation(s)
- Dorothee Jakob
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Alexander Klesen
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Benoit Allegrini
- CNRS University Cote d'Azur laboratory Institut Biology Valrose, Nice, France
| | - Elisa Darkow
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Diana Aria
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany; G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Operative Dentistry and Periodontology, Medical Center - University of Freiburg, Germany
| | - Ramona Emig
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany; CIBSS Centre for Integrative Biological Signalling Studies, Faculty of Biology, University of Freiburg, Germany
| | - Ana Simon Chica
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Eva A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Tim Guth
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Friedhelm Beyersdorf
- Faculty of Medicine, University of Freiburg, Germany; Department of Cardiovascular Surgery, University Heart Center Freiburg Bad Krozingen, Medical Center - University of Freiburg, Germany
| | - Fabian A Kari
- Faculty of Medicine, University of Freiburg, Germany; Department of Cardiovascular Surgery, University Heart Center Freiburg Bad Krozingen, Medical Center - University of Freiburg, Germany
| | - Susanne Proksch
- Faculty of Medicine, University of Freiburg, Germany; G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Operative Dentistry and Periodontology, Medical Center - University of Freiburg, Germany
| | - Stéphane N Hatem
- Sorbonne University, Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière Hospital, INSERM UMR_S1166, Cardiology department, Institute of Cardiometabolism and Nutrition-ICAN, Paris, France
| | - Matthias Karck
- Department of Cardiac Surgery, University of Heidelberg, Germany
| | - Stephan R Künzel
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Hélène Guizouarn
- CNRS University Cote d'Azur laboratory Institut Biology Valrose, Nice, France
| | - Constanze Schmidt
- Department of Cardiology, University of Heidelberg, Germany; DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim, University of Heidelberg, Germany
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany; CIBSS Centre for Integrative Biological Signalling Studies, Faculty of Biology, University of Freiburg, Germany
| | - Ursula Ravens
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
49
|
Cellular transduction of mechanical oscillations in plants by the plasma-membrane mechanosensitive channel MSL10. Proc Natl Acad Sci U S A 2021; 118:1919402118. [PMID: 33372153 DOI: 10.1073/pnas.1919402118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Plants spend most of their life oscillating around 1-3 Hz due to the effect of the wind. Therefore, stems and foliage experience repetitive mechanical stresses through these passive movements. However, the mechanism of the cellular perception and transduction of such recurring mechanical signals remains an open question. Multimeric protein complexes forming mechanosensitive (MS) channels embedded in the membrane provide an efficient system to rapidly convert mechanical tension into an electrical signal. So far, studies have mostly focused on nonoscillatory stretching of these channels. Here, we show that the plasma-membrane MS channel MscS-LIKE 10 (MSL10) from the model plant Arabidopsis thaliana responds to pulsed membrane stretching with rapid activation and relaxation kinetics in the range of 1 s. Under sinusoidal membrane stretching MSL10 presents a greater activity than under static stimulation. We observed this amplification mostly in the range of 0.3-3 Hz. Above these frequencies the channel activity is very close to that under static conditions. With a localization in aerial organs naturally submitted to wind-driven oscillations, our results suggest that the MS channel MSL10, and by extension MS channels sharing similar properties, represents a molecular component allowing the perception of oscillatory mechanical stimulations by plants.
Collapse
|
50
|
Stewart L, Turner NA. Channelling the Force to Reprogram the Matrix: Mechanosensitive Ion Channels in Cardiac Fibroblasts. Cells 2021; 10:990. [PMID: 33922466 PMCID: PMC8145896 DOI: 10.3390/cells10050990] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/13/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac fibroblasts (CF) play a pivotal role in preserving myocardial function and integrity of the heart tissue after injury, but also contribute to future susceptibility to heart failure. CF sense changes to the cardiac environment through chemical and mechanical cues that trigger changes in cellular function. In recent years, mechanosensitive ion channels have been implicated as key modulators of a range of CF functions that are important to fibrotic cardiac remodelling, including cell proliferation, myofibroblast differentiation, extracellular matrix turnover and paracrine signalling. To date, seven mechanosensitive ion channels are known to be functional in CF: the cation non-selective channels TRPC6, TRPM7, TRPV1, TRPV4 and Piezo1, and the potassium-selective channels TREK-1 and KATP. This review will outline current knowledge of these mechanosensitive ion channels in CF, discuss evidence of the mechanosensitivity of each channel, and detail the role that each channel plays in cardiac remodelling. By better understanding the role of mechanosensitive ion channels in CF, it is hoped that therapies may be developed for reducing pathological cardiac remodelling.
Collapse
Affiliation(s)
| | - Neil A. Turner
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, UK;
| |
Collapse
|