1
|
Yuan X, Zhao X, Wang W, Li C. Mechanosensing by Piezo1 and its implications in the kidney. Acta Physiol (Oxf) 2024; 240:e14152. [PMID: 38682304 DOI: 10.1111/apha.14152] [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: 09/21/2023] [Revised: 03/27/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
Piezo1 is an essential mechanosensitive transduction ion channel in mammals. Its unique structure makes it capable of converting mechanical cues into electrical and biological signals, modulating biological and (patho)physiological processes in a wide variety of cells. There is increasing evidence demonstrating that the piezo1 channel plays a vital role in renal physiology and disease conditions. This review summarizes the current evidence on the structure and properties of Piezo1, gating modulation, and pharmacological characteristics, with special focus on the distribution and (patho)physiological significance of Piezo1 in the kidney, which may provide insights into potential treatment targets for renal diseases involving this ion channel.
Collapse
Affiliation(s)
- Xi Yuan
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoduo Zhao
- Department of Pathology, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Weidong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chunling Li
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Nan X, Wang M, Du J, Liu Y, Cao L, Zhou J, Liu L, Li X. Single vesicle chemistry reveals partial release happens at the mechanical stress-induced exocytosis. Talanta 2024; 271:125637. [PMID: 38237284 DOI: 10.1016/j.talanta.2024.125637] [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: 12/14/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 02/24/2024]
Abstract
Neuronal activity can be modulated by mechanical stress in the central nervous system (CNS) in neurodegenerative diseases, for example Alzheimer's disease. However, the impact of mechanical stress on chemical signal transmission, especially the storage and release of neurotransmitter in neuron vesicles, has not been fully clarified. In this study, a nanotip conical carbon fiber microelectrode (CFME) and a disk CFME are placed in and on a cell, respectively. The nanotip conical CFME functions for both the mechanical stress and the quantification of transmitter storage in single vesicles, while the disk CFME is used to monitor the transmitter release during exocytosis induced by mechanical stress at the same cell. By comparing the vesicular transmitter storage with its release during mechanical stress-induced exocytosis at the same cell, we find the release ratio of transmitter in chromaffin cells varies from 27 % to 100 %, while for PC12 cells from 30 % to 100 %. Our results indicate that the exocytosis of cells responding to mechanical stress shows individual difference obviously, with a significant population exhibiting partial release mode. The variation of Ca2+ channels and mechanosensitive ion channels on cell membrane may both contribute to this variation. Our discovery not only shows mechanical stress can change the transmission of cellular chemical signals at the vesicle level, but also provides an important reference perspective for the study of nervous system regulation and nervous system diseases.
Collapse
Affiliation(s)
- Xiaoke Nan
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Mengying Wang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China
| | - Jinchang Du
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China
| | - Yuying Liu
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Lijiao Cao
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Junlan Zhou
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Luyao Liu
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China
| | - Xianchan Li
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China; Department of Preventive Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, China.
| |
Collapse
|
3
|
Ojeda-Alonso J, Calvo-Enrique L, Paricio-Montesinos R, Kumar R, Zhang MD, Poulet JFA, Ernfors P, Lewin GR. Sensory Schwann cells set perceptual thresholds for touch and selectively regulate mechanical nociception. Nat Commun 2024; 15:898. [PMID: 38320986 PMCID: PMC10847425 DOI: 10.1038/s41467-024-44845-8] [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: 12/14/2021] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
Previous work identified nociceptive Schwann cells that can initiate pain. Consistent with the existence of inherently mechanosensitive sensory Schwann cells, we found that in mice, the mechanosensory function of almost all nociceptors, including those signaling fast pain, were dependent on sensory Schwann cells. In polymodal nociceptors, sensory Schwann cells signal mechanical, but not cold or heat pain. Terminal Schwann cells also surround mechanoreceptor nerve-endings within the Meissner's corpuscle and in hair follicle lanceolate endings that both signal vibrotactile touch. Within Meissner´s corpuscles, two molecularly and functionally distinct sensory Schwann cells positive for Sox10 and Sox2 differentially modulate rapidly adapting mechanoreceptor function. Using optogenetics we show that Meissner's corpuscle Schwann cells are necessary for the perception of low threshold vibrotactile stimuli. These results show that sensory Schwann cells within diverse glio-neural mechanosensory end-organs are sensors for mechanical pain as well as necessary for touch perception.
Collapse
Affiliation(s)
- Julia Ojeda-Alonso
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Laura Calvo-Enrique
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
- Departamento de Biología Celular y Patología, Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca, Spain
| | - Ricardo Paricio-Montesinos
- Neural Circuits and Behavior, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Rakesh Kumar
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
- Pain Center, Department of Anesthesiology Washington University School of Medicine, CB 8108, 660 S. Euclid Ave., St. Louis, MO, 63110, USA
| | - Ming-Dong Zhang
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - James F A Poulet
- Neural Circuits and Behavior, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Patrik Ernfors
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden.
| | - Gary R Lewin
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- German Center for Mental Health (DZPG), partner site Berlin, Berlin, Germany.
| |
Collapse
|
4
|
Muir WM, Lo CL, Bell RL, Zhou FC. Multi-animal-model study reveals mutations in neural plasticity and nociception genes linked to excessive alcohol drinking. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1478-1493. [PMID: 37336636 PMCID: PMC10728351 DOI: 10.1111/acer.15131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/10/2023] [Accepted: 06/07/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND The basis for familial alcohol use disorder (AUD) remains an enigma due to various biological and societal confounds. The present study used three of the most adopted and documented rat models, combining the alcohol-preferring/non-alcohol-preferring (P/NP) lines and high alcohol-drinking/low alcohol-drinking (HAD/LAD) replicated lines, of AUD as examined through the lens of whole genomic analyses. METHODS We used complete genome sequencing of the P/NP lines and previously published sequences of the HAD/LAD replicates to enhance the discovery of variants associated with AUD and to remove confounding with genetic background and random genetic drift. Specifically, we used high-order statistical methods to search for genetic variants whose frequency changes in whole sets of gene ontologies corresponded with phenotypic changes in the direction of selection, that is, ethanol-drinking preference. RESULTS Our first finding was that in addition to variants causing translational changes, the principal genetic changes associated with drinking predisposition were silent mutations and mutations in the 3' untranslated regions (3'UTR) of genes. Neither of these types of mutations alters the amino acid sequence of the translated protein but they influence both the rate and conformation of gene transcription, including its stability and posttranslational events that alter gene efficacy. This finding argues for refocusing human genomic studies on changes in gene efficacy. Among the key ontologies identified were the central genes associated with the Na+ voltage-gated channels of neurons and glia (including the Scn1a, Scn2a, Scn2b, Scn3a, Scn7a, and Scn9a subtypes) and excitatory glutamatergic secretion (including Grm2 and Myo6), both of which are essential in neuroplasticity. In addition, we identified "Nociception or Sensory Perception of Pain," which contained variants in nociception (Arrb1, Ccl3, Ephb1) and enlist sodium (Scn1a, Scn2a, Scn2b, Scn3a, Scn7a), pain activation (Scn9a), and potassium channel (Kcna1) genes. CONCLUSION The multi-model analyses used herein reduced the confounding effects of random drift and the "founders" genetic background. The most differentiated bidirectionally selected genes across all three animal models were Scn9a, Scn1a, and Kcna, all of which are annotated in the nociception ontology. The complexity of neuroplasticity and nociception adds strength to the hypothesis that neuroplasticity and pain (physical or psychological) are prominent phenotypes genetically linked to the development of AUD.
Collapse
Affiliation(s)
- William M. Muir
- Indiana Alcohol Research Center, Indiana University School of Medicine
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Chiao-Ling Lo
- Indiana Alcohol Research Center, Indiana University School of Medicine
| | - Richard L. Bell
- Indiana Alcohol Research Center, Indiana University School of Medicine
- Stark Neuroscience Research Institute, Indianapolis, Indiana, USA
| | - Feng C. Zhou
- Indiana Alcohol Research Center, Indiana University School of Medicine
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Stark Neuroscience Research Institute, Indianapolis, Indiana, USA
| |
Collapse
|
5
|
Darkow E, Yusuf D, Rajamani S, Backofen R, Kohl P, Ravens U, Peyronnet R. Meta-Analysis of Mechano-Sensitive Ion Channels in Human Hearts: Chamber- and Disease-Preferential mRNA Expression. Int J Mol Sci 2023; 24:10961. [PMID: 37446137 DOI: 10.3390/ijms241310961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
The cardiac cell mechanical environment changes on a beat-by-beat basis as well as in the course of various cardiac diseases. Cells sense and respond to mechanical cues via specialized mechano-sensors initiating adaptive signaling cascades. With the aim of revealing new candidates underlying mechano-transduction relevant to cardiac diseases, we investigated mechano-sensitive ion channels (MSC) in human hearts for their chamber- and disease-preferential mRNA expression. Based on a meta-analysis of RNA sequencing studies, we compared the mRNA expression levels of MSC in human atrial and ventricular tissue samples from transplant donor hearts (no cardiac disease), and from patients in sinus rhythm (underlying diseases: heart failure, coronary artery disease, heart valve disease) or with atrial fibrillation. Our results suggest that a number of MSC genes are expressed chamber preferentially, e.g., CHRNE in the atria (compared to the ventricles), TRPV4 in the right atrium (compared to the left atrium), CACNA1B and KCNMB1 in the left atrium (compared to the right atrium), as well as KCNK2 and KCNJ2 in ventricles (compared to the atria). Furthermore, 15 MSC genes are differentially expressed in cardiac disease, out of which SCN9A (lower expressed in heart failure compared to donor tissue) and KCNQ5 (lower expressed in atrial fibrillation compared to sinus rhythm) show a more than twofold difference, indicative of possible functional relevance. Thus, we provide an overview of cardiac MSC mRNA expression in the four cardiac chambers from patients with different cardiac diseases. We suggest that the observed differences in MSC mRNA expression may identify candidates involved in altered mechano-transduction in the respective diseases.
Collapse
Affiliation(s)
- Elisa Darkow
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg∙Bad Krozingen, 79110 Freiburg im Breisgau, Germany
- Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Dilmurat Yusuf
- Bioinformatics Group, Department of Computer Science, University of Freiburg, 79110 Freiburg im Breisgau, Germany
| | - Sridharan Rajamani
- Translational Safety and Bioanalytical Sciences, Amgen Research, Amgen Inc., South San Francisco, CA 91320, USA
| | - Rolf Backofen
- Bioinformatics Group, Department of Computer Science, University of Freiburg, 79110 Freiburg im Breisgau, Germany
- Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg∙Bad Krozingen, 79110 Freiburg im Breisgau, Germany
- Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany
- Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Ursula Ravens
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg∙Bad Krozingen, 79110 Freiburg im Breisgau, Germany
- Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg∙Bad Krozingen, 79110 Freiburg im Breisgau, Germany
- Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany
| |
Collapse
|
6
|
Ovsepian SV, Waxman SG. Gene therapy for chronic pain: emerging opportunities in target-rich peripheral nociceptors. Nat Rev Neurosci 2023; 24:252-265. [PMID: 36658346 DOI: 10.1038/s41583-022-00673-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2022] [Indexed: 01/20/2023]
Abstract
With sweeping advances in precision delivery systems and manipulation of the genomes and transcriptomes of various cell types, medical biotechnology offers unprecedented selectivity for and control of a wide variety of biological processes, forging new opportunities for therapeutic interventions. This perspective summarizes state-of-the-art gene therapies enabled by recent innovations, with an emphasis on the expanding universe of molecular targets that govern the activity and function of primary sensory neurons and which might be exploited to effectively treat chronic pain.
Collapse
Affiliation(s)
- Saak V Ovsepian
- School of Science, Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, UK.
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
7
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
8
|
Primak AL, Orlov NA, Peigneur S, Tytgat J, Ignatova AA, Denisova KR, Yakimov SA, Kirpichnikov MP, Nekrasova OV, Feofanov AV. AgTx2-GFP, Fluorescent Blocker Targeting Pharmacologically Important K v1.x (x = 1, 3, 6) Channels. Toxins (Basel) 2023; 15:toxins15030229. [PMID: 36977120 PMCID: PMC10056440 DOI: 10.3390/toxins15030229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
The growing interest in potassium channels as pharmacological targets has stimulated the development of their fluorescent ligands (including genetically encoded peptide toxins fused with fluorescent proteins) for analytical and imaging applications. We report on the properties of agitoxin 2 C-terminally fused with enhanced GFP (AgTx2-GFP) as one of the most active genetically encoded fluorescent ligands of potassium voltage-gated Kv1.x (x = 1, 3, 6) channels. AgTx2-GFP possesses subnanomolar affinities for hybrid KcsA-Kv1.x (x = 3, 6) channels and a low nanomolar affinity to KcsA-Kv1.1 with moderate dependence on pH in the 7.0-8.0 range. Electrophysiological studies on oocytes showed a pore-blocking activity of AgTx2-GFP at low nanomolar concentrations for Kv1.x (x = 1, 3, 6) channels and at micromolar concentrations for Kv1.2. AgTx2-GFP bound to Kv1.3 at the membranes of mammalian cells with a dissociation constant of 3.4 ± 0.8 nM, providing fluorescent imaging of the channel membranous distribution, and this binding depended weakly on the channel state (open or closed). AgTx2-GFP can be used in combination with hybrid KcsA-Kv1.x (x = 1, 3, 6) channels on the membranes of E. coli spheroplasts or with Kv1.3 channels on the membranes of mammalian cells for the search and study of nonlabeled peptide pore blockers, including measurement of their affinity.
Collapse
Affiliation(s)
- Alexandra L Primak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Nikita A Orlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Steve Peigneur
- Toxicology and Pharmacology, Campus Gasthuisberg O&N2, University of Leuven (KU Leuven), Herestraat 49, P.O. Box 922, B-3000 Leuven, Belgium
| | - Jan Tytgat
- Toxicology and Pharmacology, Campus Gasthuisberg O&N2, University of Leuven (KU Leuven), Herestraat 49, P.O. Box 922, B-3000 Leuven, Belgium
| | - Anastasia A Ignatova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Kristina R Denisova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Sergey A Yakimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Mikhail P Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Oksana V Nekrasova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Alexey V Feofanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
9
|
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: 1.0] [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
|
10
|
Valek L, Tran BN, Tegeder I. Cold avoidance and heat pain hypersensitivity in neuronal nucleoredoxin knockout mice. Free Radic Biol Med 2022; 192:84-97. [PMID: 36126861 DOI: 10.1016/j.freeradbiomed.2022.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 08/15/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022]
Abstract
Nucleoredoxin is a thioredoxin-like oxidoreductase that mainly acts as oxidase and thereby regulates calcium calmodulin kinase Camk2a, an effector of nitric oxide mediated synaptic potentiation and nociceptive sensitization. We asked here if and how NXN affects thermal sensation and nociception in mice using pan-neuronal NXN deletion driven by Nestin-Cre, and sensory neuron specific deletion driven by Advillin-Cre. In a thermal gradient ring, where mice can freely choose the temperature of well-being, Nestin-NXN-/- mice avoided unpleasant cold temperatures. In neuropathic and inflammatory nociceptive models, Nestin-NXN-/- and Advillin-NXN-/- mice displayed subtle phenotypes of heightened heat nociception. Abnormal thermal in vivo responses were associated with heightened calcium influx upon stimulation of transient receptor channels, with heightened oxygen consumption upon disruption of the mitochondrial membrane potential and with higher density of neurite trees of primary sensory neurons of the dorsal root ganglia in cultures. The data suggest that loss of NXN's balancing redox functions leads to maladaptive changes in sensory neurons that manifest in vivo as polyneuropathy-like abnormal cold sensitivity and heat "pain".
Collapse
Affiliation(s)
- Lucie Valek
- Institute of Clinical Pharmacology, Goethe-University, Faculty of Medicine, Frankfurt, Germany
| | - Bao Ngoc Tran
- Institute of Clinical Pharmacology, Goethe-University, Faculty of Medicine, Frankfurt, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University, Faculty of Medicine, Frankfurt, Germany.
| |
Collapse
|
11
|
Delmas P, Parpaite T, Coste B. PIEZO channels and newcomers in the mammalian mechanosensitive ion channel family. Neuron 2022; 110:2713-2727. [PMID: 35907398 DOI: 10.1016/j.neuron.2022.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/25/2022] [Accepted: 07/01/2022] [Indexed: 10/16/2022]
Abstract
Many ion channels have been described as mechanosensitive according to various criteria. Most broadly defined, an ion channel is called mechanosensitive if its activity is controlled by application of a physical force. The last decade has witnessed a revolution in mechanosensory physiology at the molecular, cellular, and system levels, both in health and in diseases. Since the discovery of the PIEZO proteins as prototypical mechanosensitive channel, many proteins have been proposed to transduce mechanosensory information in mammals. However, few of these newly identified candidates have all the attributes of bona fide, pore-forming mechanosensitive ion channels. In this perspective, we will cover and discuss new data that have advanced our understanding of mechanosensation at the molecular level.
Collapse
Affiliation(s)
- Patrick Delmas
- SomatoSens, Laboratory for Cognitive Neuroscience, Aix-Marseille University, CNRS UMR 7291, Marseilles, France.
| | - Thibaud Parpaite
- SomatoSens, Laboratory for Cognitive Neuroscience, Aix-Marseille University, CNRS UMR 7291, Marseilles, France
| | - Bertrand Coste
- SomatoSens, Laboratory for Cognitive Neuroscience, Aix-Marseille University, CNRS UMR 7291, Marseilles, France
| |
Collapse
|
12
|
Yang L, Wang H, Lu W, Yang G, Lin Z, Chen R, Li H. Quantitative proteomic analysis of oxaliplatin induced peripheral neurotoxicity. J Proteomics 2022; 266:104682. [PMID: 35830924 DOI: 10.1016/j.jprot.2022.104682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/19/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022]
Abstract
Oxaliplatin (OXA)-induced peripheral neurotoxicity (OIPN) is a high-incidence and dose-dependent adverse reaction during OXA treatment. Its underlying mechanisms remain unclear, and no effective treatment or prevention therapies are currently available. Here, we employed a data independent acquisition (DIA)-based quantitative proteomic strategy to investigate the global proteome alterations in the dorsal root ganglion (DRG) tissues from mice injected with OXA for different periods. We identified 1128 differentially regulated proteins that were divided into six subclusters according to their alteration trends. Interestingly, these proteins were involved in cellular processes such as cell cycle, ribosomal stress, metabolism, and ion transport. In addition, OXA administration induced abundance changes of ion channels and proteins associated with mitochondrial function and reactive oxygen species production. Furthermore, we investigated the effects of diroximel fumarate (DRF), an FDA-approved oral fumarate drug for the treatment of relapsing forms of multiple sclerosis. Our findings showed that DRF could effectively ameliorate symptoms of OIPN and reduce the level of oxidative stress in mice. Taken together, our study systematically mapped the proteome alteration associated with the neural toxicity of OXA, and the findings could be leveraged to better understand the mechanisms of OIPN and to develop more effect treatment therapies. SIGNIFICANCE: Oxaliplatin (OXA)-induced peripheral neurotoxicity (OIPN) is a high-incidence and dose-dependent adverse reaction with unclear mechanism. Here we employed a data independent acquisition (DIA)-based quantitative proteomic strategy to explore the proteome changes in dorsal root ganglion (DRG) tissues from mice treated by OXA. The findings provided novel insights regarding the mechanisms of OIPN. For example, our data showed that OXA induced a broad disturbance in metabolism, particularly in glycolysis and amino acid metabolism. Additionally, we observed abundance changes of many ion channels and proteins associated with mitochondrial function and reactive oxygen species production. Furthermore, this study provided the first evidence for the possibility of repositioning diroximel fumarate (DRF) for treating OIPN.
Collapse
Affiliation(s)
- Linlin Yang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Hua Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Wanting Lu
- General Surgery Department and Neurology Department, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Gangqi Yang
- General Surgery Department and Neurology Department, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Zian Lin
- Ministry of Education Key Laboratory of Analytical Science for Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Ruibing Chen
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China.
| | - Hongyan Li
- General Surgery Department and Neurology Department, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China.
| |
Collapse
|
13
|
Lv YY, Wang H, Fan HT, Xu T, Xin WJ, Guo RX. SUMOylation of Kir7.1 participates in neuropathic pain through regulating its membrane expression in spinal cord neurons. CNS Neurosci Ther 2022; 28:1259-1267. [PMID: 35633059 PMCID: PMC9253747 DOI: 10.1111/cns.13871] [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: 01/22/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 11/28/2022] Open
Abstract
Aims Potassium (K+) channels have been demonstrated to play a prominent involvement in nociceptive processing. Kir7.1, the newest members of the Kir channel family, has not been extensively studied in the CNS, and its function remains largely unknown. The present study investigated the role of spinal Kir7.1 in the development of pathological pain. Methods and Results Neuropathic pain was induced by spared nerve injury (SNI). The mechanical sensitivity was assessed by von Frey test. Immunofluorescence staining assay revealed that Kir7.1 was predominantly expressed in spinal neurons but not astrocytes or microglia in normal rats. Western blot results showed that SNI markedly decreased the total and membrane expression of Kir7.1 in the spinal dorsal horn accompanied by mechanical hypersensitivity. Blocking Kir7.1 with the specific antagonist ML418 or knockdown kir7.1 by siRNA led to mechanical allodynia. Co‐IP results showed that the spinal kir7.1 channels were decorated by SUMO‐1 but not SUMO‐2/3, and Kir7.1 SUMOylation was upregulated following SNI. Moreover, inhibited SUMOylation by GA (E1 inhibitor) or 2‐D08 (UBC9 inhibitor) can increase the spinal surface Kir7.1 expression. Conclusion SUMOylation of the Kir7.1 in the spinal cord might contribute to the development of SNI‐induced mechanical allodynia by decreasing the Kir7.1 surface expression in rats.
Collapse
Affiliation(s)
- You-You Lv
- Department of Anesthesiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Han Wang
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hai-Ting Fan
- Department of Anesthesiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Ting Xu
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wen-Jun Xin
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Rui-Xian Guo
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
14
|
Mercado-Perez A, Beyder A. Gut feelings: mechanosensing in the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2022; 19:283-296. [PMID: 35022607 PMCID: PMC9059832 DOI: 10.1038/s41575-021-00561-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 12/11/2022]
Abstract
The primary function of the gut is to procure nutrients. Synchronized mechanical activities underlie nearly all its endeavours. Coordination of mechanical activities depends on sensing of the mechanical forces, in a process called mechanosensation. The gut has a range of mechanosensory cells. They function either as specialized mechanoreceptors, which convert mechanical stimuli into coordinated physiological responses at the organ level, or as non-specialized mechanosensory cells that adjust their function based on the mechanical state of their environment. All major cell types in the gastrointestinal tract contain subpopulations that act as specialized mechanoreceptors: epithelia, smooth muscle, neurons, immune cells, and others. These cells are tuned to the physical properties of the surrounding tissue, so they can discriminate mechanical stimuli from the baseline mechanical state. The importance of gastrointestinal mechanosensation has long been recognized, but the latest discoveries of molecular identities of mechanosensors and technical advances that resolve the relevant circuitry have poised the field to make important intellectual leaps. This Review describes the mechanical factors relevant for normal function, as well as the molecules, cells and circuits involved in gastrointestinal mechanosensing. It concludes by outlining important unanswered questions in gastrointestinal mechanosensing.
Collapse
Affiliation(s)
- Arnaldo Mercado-Perez
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA
- Medical Scientist Training Program (MSTP), Mayo Clinic, Rochester, MN, USA
| | - Arthur Beyder
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA.
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
15
|
Sun H, Patil MJ, Ru F, Meeker S, Undem BJ. K
V
1/D‐type potassium channels inhibit the excitability of bronchopulmonary vagal afferent nerves. J Physiol 2022; 600:2953-2971. [PMID: 35430729 PMCID: PMC9203938 DOI: 10.1113/jp282803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract The KV1/D‐type potassium current (ID) is an important determinant of neuronal excitability. This study explored whether and how ID channels regulate the activation of bronchopulmonary vagal afferent nerves. The single‐neuron RT‐PCR assay revealed that nearly all mouse bronchopulmonary nodose neurons expressed the transcripts of α‐dendrotoxin (α‐DTX)‐sensitive, ID channel‐forming KV1.1, KV1.2 and/or KV1.6 α‐subunits, with the expression of KV1.6 being most prevalent. Patch‐clamp recordings showed that ID, defined as the α‐DTX‐sensitive K+ current, activated at voltages slightly more negative than the resting membrane potential in lung‐specific nodose neurons and displayed little inactivation at subthreshold voltages. Inhibition of ID channels by α‐DTX depolarized the lung‐specific nodose neurons and caused an increase in input resistance, decrease in rheobase, as well as increase in action potential number and firing frequency in response to suprathreshold current steps. Application of α‐DTX to the lungs via trachea in the mouse ex vivo vagally innervated trachea–lungs preparation led to action potential discharges in nearly half of bronchopulmonary nodose afferent nerve fibres, including nodose C‐fibres, as detected by the two‐photon microscopic Ca2+ imaging technique and extracellular electrophysiological recordings. In conclusion, ID channels act as a critical brake on the activation of bronchopulmonary vagal afferent nerves by stabilizing the membrane potential, counterbalancing the subthreshold depolarization and promoting the adaptation of action potential firings. Down‐regulation of ID channels, as occurs in various inflammatory diseases, may contribute to the enhanced C‐fibre activity in airway diseases that are associated with excessive coughing, dyspnoea, and reflex bronchospasm and secretions. Key points The α‐dendrotoxin (α‐DTX)‐sensitive D‐type K+ current (ID) is an important determinant of neuronal excitability. Nearly all bronchopulmonary nodose afferent neurons in the mouse express ID and the transcripts of α‐DTX‐sensitive, ID channel‐forming KV1.1, KV1.2 and/or KV1.6 α‐subunits. Inhibition of ID channels by α‐DTX depolarizes the bronchopulmonary nodose neurons, reduces the minimal depolarizing current needed to evoke an action potential (AP) and increases AP number and AP firing frequency in response to suprathreshold stimulations. Application of α‐DTX to the lungs ex vivo elicits AP discharges in about half of bronchopulmonary nodose C‐fibre terminals.
Our novel finding that ID channels act as a critical brake on the activation of bronchopulmonary vagal afferent nerves suggests that their down‐regulation, as occurs in various inflammatory diseases, may contribute to the enhanced C‐fibre activity in airway inflammation associated with excessive respiratory symptoms.
Collapse
Affiliation(s)
- Hui Sun
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Mayur J. Patil
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Fei Ru
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Sonya Meeker
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Bradley J. Undem
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| |
Collapse
|
16
|
Perez-Flores MC, Verschooten E, Lee JH, Kim HJ, Joris PX, Yamoah EN. Intrinsic mechanical sensitivity of mammalian auditory neurons as a contributor to sound-driven neural activity. eLife 2022; 11:74948. [PMID: 35266451 PMCID: PMC8942473 DOI: 10.7554/elife.74948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/09/2022] [Indexed: 11/18/2022] Open
Abstract
Mechanosensation – by which mechanical stimuli are converted into a neuronal signal – is the basis for the sensory systems of hearing, balance, and touch. Mechanosensation is unmatched in speed and its diverse range of sensitivities, reaching its highest temporal limits with the sense of hearing; however, hair cells (HCs) and the auditory nerve (AN) serve as obligatory bottlenecks for sounds to engage the brain. Like other sensory neurons, auditory neurons use the canonical pathway for neurotransmission and millisecond-duration action potentials (APs). How the auditory system utilizes the relatively slow transmission mechanisms to achieve ultrafast speed, and high audio-frequency hearing remains an enigma. Here, we address this paradox and report that the mouse, and chinchilla, AN are mechanically sensitive, and minute mechanical displacement profoundly affects its response properties. Sound-mimicking sinusoidal mechanical and electrical current stimuli affect phase-locked responses. In a phase-dependent manner, the two stimuli can also evoke suppressive responses. We propose that mechanical sensitivity interacts with synaptic responses to shape responses in the AN, including frequency tuning and temporal phase locking. Combining neurotransmission and mechanical sensation to control spike patterns gives the mammalian AN a secondary receptor role, an emerging theme in primary neuronal functions.
Collapse
Affiliation(s)
| | - Eric Verschooten
- Laboratory of Auditory Neurophysiology, University of Leuven, Leuven, Belgium
| | | | | | - Philip X Joris
- Laboratory of Auditory Neurophysiology, University of Leuven, Leuven, Belgium
| | | |
Collapse
|
17
|
Kouraki A, Doherty M, Fernandes GS, Zhang W, Walsh DA, Kelly A, Valdes AM. Different genes may be involved in distal and local sensitisation: a genome-wide gene-based association study and meta-analysis. Eur J Pain 2021; 26:740-753. [PMID: 34958702 PMCID: PMC9303629 DOI: 10.1002/ejp.1902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/11/2021] [Accepted: 12/25/2021] [Indexed: 11/22/2022]
Abstract
Background Neuropathic pain symptoms and signs of increased pain sensitization in osteoarthritis (OA) patients may explain persistent pain after total joint replacement (TJR). Therefore, identifying genetic markers associated with pain sensitization and neuropathic‐like pain phenotypes could be clinically important in identifying targets for early intervention. Methods We performed a genome‐wide gene‐based association study (GWGAS) using pressure pain detection thresholds (PPTs) from distal pain‐free sites (anterior tibia), a measure of distal sensitization, and from proximal pain‐affected sites (lateral joint line), a measure of local sensitization, in 320 knee OA participants from the Knee Pain and related health in the Community (KPIC) cohort. We next performed gene‐based fixed‐effects meta‐analysis of PPTs and a neuropathic‐like pain phenotype using genome‐wide association study (GWAS) data from KPIC and from an independent cohort of 613 post‐TJR participants, respectively. Results The most significant genes associated with distal and local sensitization were OR5B3 and BRDT, respectively. We also found previously identified neuropathic pain‐associated genes—KCNA1, MTOR, ADORA1 and SCN3B—associated with PPT at the anterior tibia and an inflammatory pain gene—PTAFR—associated with PPT at the lateral joint line. Meta‐analysis results of anterior tibia and neuropathic‐like pain phenotypes revealed genes associated with bone morphogenesis, neuro‐inflammation, obesity, type 2 diabetes, cardiovascular disease and cognitive function. Conclusions Overall, our results suggest that different biological processes might be involved in distal and local sensitization, and common genetic mechanisms might be implicated in distal sensitization and neuropathic‐like pain. Future studies are needed to replicate these findings. Significance To the best of our knowledge, this is the first GWAS for pain sensitization and the first gene‐based meta‐analysis of pain sensitization and neuropathic‐like pain. Higher pain sensitization and neuropathic pain symptoms are associated with persistent pain after surgery hence, identifying genetic biomarkers and molecular pathways associated with these traits is clinically relevant.
Collapse
Affiliation(s)
- A Kouraki
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - M Doherty
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,Versus Arthritis Centre for Sports, Exercise and Osteoarthritis, University of Nottingham, Nottingham, NG7 2UH, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - G S Fernandes
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 6EH, United Kingdom
| | - W Zhang
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,Versus Arthritis Centre for Sports, Exercise and Osteoarthritis, University of Nottingham, Nottingham, NG7 2UH, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - D A Walsh
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,Versus Arthritis Centre for Sports, Exercise and Osteoarthritis, University of Nottingham, Nottingham, NG7 2UH, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - A Kelly
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - A M Valdes
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| |
Collapse
|
18
|
Alles SRA, Smith PA. Peripheral Voltage-Gated Cation Channels in Neuropathic Pain and Their Potential as Therapeutic Targets. FRONTIERS IN PAIN RESEARCH 2021; 2:750583. [PMID: 35295464 PMCID: PMC8915663 DOI: 10.3389/fpain.2021.750583] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
The persistence of increased excitability and spontaneous activity in injured peripheral neurons is imperative for the development and persistence of many forms of neuropathic pain. This aberrant activity involves increased activity and/or expression of voltage-gated Na+ and Ca2+ channels and hyperpolarization activated cyclic nucleotide gated (HCN) channels as well as decreased function of K+ channels. Because they display limited central side effects, peripherally restricted Na+ and Ca2+ channel blockers and K+ channel activators offer potential therapeutic approaches to pain management. This review outlines the current status and future therapeutic promise of peripherally acting channel modulators. Selective blockers of Nav1.3, Nav1.7, Nav1.8, Cav3.2, and HCN2 and activators of Kv7.2 abrogate signs of neuropathic pain in animal models. Unfortunately, their performance in the clinic has been disappointing; some substances fail to meet therapeutic end points whereas others produce dose-limiting side effects. Despite this, peripheral voltage-gated cation channels retain their promise as therapeutic targets. The way forward may include (i) further structural refinement of K+ channel activators such as retigabine and ASP0819 to improve selectivity and limit toxicity; use or modification of Na+ channel blockers such as vixotrigine, PF-05089771, A803467, PF-01247324, VX-150 or arachnid toxins such as Tap1a; the use of Ca2+ channel blockers such as TTA-P2, TTA-A2, Z 944, ACT709478, and CNCB-2; (ii) improving methods for assessing "pain" as opposed to nociception in rodent models; (iii) recognizing sex differences in pain etiology; (iv) tailoring of therapeutic approaches to meet the symptoms and etiology of pain in individual patients via quantitative sensory testing and other personalized medicine approaches; (v) targeting genetic and biochemical mechanisms controlling channel expression using anti-NGF antibodies such as tanezumab or re-purposed drugs such as vorinostat, a histone methyltransferase inhibitor used in the management of T-cell lymphoma, or cercosporamide a MNK 1/2 inhibitor used in treatment of rheumatoid arthritis; (vi) combination therapy using drugs that are selective for different channel types or regulatory processes; (vii) directing preclinical validation work toward the use of human or human-derived tissue samples; and (viii) application of molecular biological approaches such as clustered regularly interspaced short palindromic repeats (CRISPR) technology.
Collapse
Affiliation(s)
- Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Peter A Smith
- Department of Pharmacology, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
19
|
Lewis KJ. Osteocyte calcium signaling - A potential translator of mechanical load to mechanobiology. Bone 2021; 153:116136. [PMID: 34339908 DOI: 10.1016/j.bone.2021.116136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/25/2021] [Accepted: 07/27/2021] [Indexed: 10/20/2022]
Abstract
Osteocytes are embedded dendritic bone cells; by virtue of their position in bone tissue, ability to coordinate bone building osteoblasts and resorbing osteoclasts, and sensitivity to tissue level mechanical loading, they serve as the resident bone mechanosensor. The mechanisms osteocytes use to change mechanical loading into biological signals that drive tissue level changes has been well studied over the last 30 years, however the ways loading parameters are encoded at the cellular level are still not fully understood. Calcium signaling is a first messenger signal exhibited by osteocytes in response to mechanical forces. A body of work interrogating the mechanisms of osteocyte calcium signaling exists and is presently expanding, presenting the opportunity to better understand the relationship between calcium signaling characteristics and tuned osteocyte responses to tissue level strain features (e.g. magnitude, duration, frequency). This review covers the history of osteocyte load induced calcium signaling and highlights potential cellular mechanisms used by osteocytes to turn details about loading parameters into biological events.
Collapse
Affiliation(s)
- Karl J Lewis
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America.
| |
Collapse
|
20
|
Hao J, Brosse L, Bonnet C, Ducrocq M, Padilla F, Penalba V, Desplat A, Ruel J, Delmas P. The widely used antihistamine mepyramine causes topical pain relief through direct blockade of nociceptor sodium channels. FASEB J 2021; 35:e22025. [PMID: 34758144 DOI: 10.1096/fj.202100976rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 11/11/2022]
Abstract
Mepyramine, a first-generation antihistamine targeting the histamine H(1) receptor, was extensively prescribed to patients suffering from allergic reactions and urticaria. Serious adverse effects, especially in case of overdose, were frequently reported, including drowsiness, impaired thinking, convulsion, and coma. Many of these side effects were associated with the blockade of histaminergic or cholinergic receptors. Here we show that mepyramine directly inhibits a variety of voltage-gated sodium channels, including the Tetrodotoxin-sensitive isoforms and the main isoforms (Nav1.7, Nav1.8, and Nav1.9) of nociceptors. Estimated IC50 were within the range of drug concentrations detected in poisoned patients. Mepyramine inhibited sodium channels through fast- or slow-inactivated state preference depending on the isoform. Moreover, mepyramine inhibited the firing responses of C- and Aβ-type nerve fibers in ex vivo skin-nerve preparations. Locally applied mepyramine had analgesic effects on the scorpion toxin-induced excruciating pain and produced pain relief in acute, inflammatory, and chronic pain models. Collectively, these data provide evidence that mepyramine has the potential to be developed as a topical analgesic agent.
Collapse
Affiliation(s)
- Jizhe Hao
- Laboratoire de Neurosciences Cognitives, UMR 7291, CNRS, Aix-Marseille-Université, Marseille Cedex 15, France
| | - Lucie Brosse
- Laboratoire de Neurosciences Cognitives, UMR 7291, CNRS, Aix-Marseille-Université, Marseille Cedex 15, France
| | - Caroline Bonnet
- Laboratoire de Neurosciences Cognitives, UMR 7291, CNRS, Aix-Marseille-Université, Marseille Cedex 15, France
| | - Myriam Ducrocq
- Laboratoire de Neurosciences Cognitives, UMR 7291, CNRS, Aix-Marseille-Université, Marseille Cedex 15, France
| | - Françoise Padilla
- Laboratoire de Neurosciences Cognitives, UMR 7291, CNRS, Aix-Marseille-Université, Marseille Cedex 15, France
| | - Virginie Penalba
- Laboratoire de Neurosciences Cognitives, UMR 7291, CNRS, Aix-Marseille-Université, Marseille Cedex 15, France
| | - Angélique Desplat
- Laboratoire de Neurosciences Cognitives, UMR 7291, CNRS, Aix-Marseille-Université, Marseille Cedex 15, France
| | - Jérôme Ruel
- Laboratoire de Neurosciences Cognitives, UMR 7291, CNRS, Aix-Marseille-Université, Marseille Cedex 15, France
| | - Patrick Delmas
- Laboratoire de Neurosciences Cognitives, UMR 7291, CNRS, Aix-Marseille-Université, Marseille Cedex 15, France
| |
Collapse
|
21
|
Studying Independent Kcna6 Knock-out Mice Reveals Toxicity of Exogenous LacZ to Central Nociceptor Terminals and Differential Effects of Kv1.6 on Acute and Neuropathic Pain Sensation. J Neurosci 2021; 41:9141-9162. [PMID: 34544832 DOI: 10.1523/jneurosci.0187-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 11/21/2022] Open
Abstract
The potassium channel Kv1.6 has recently been implicated as a major modulatory channel subunit expressed in primary nociceptors. Furthermore, its expression at juxtaparanodes of myelinated primary afferents is induced following traumatic nerve injury as part of an endogenous mechanism to reduce hyperexcitability and pain-related hypersensitivity. In this study, we compared two mouse models of constitutive Kv1.6 knock-out (KO) achieved by different methods: traditional gene trap via homologous recombination and CRISPR-mediated excision. Both Kv1.6 KO mouse lines exhibited an unexpected reduction in sensitivity to noxious heat stimuli, to differing extents: the Kv1.6 mice produced via gene trap had a far more significant hyposensitivity. These mice (Kcna6lacZ ) expressed the bacterial reporter enzyme LacZ in place of Kv1.6 as a result of the gene trap mechanism, and we found that their central primary afferent presynaptic terminals developed a striking neurodegenerative phenotype involving accumulation of lipid species, development of "meganeurites," and impaired transmission to dorsal horn wide dynamic range neurons. The anatomic defects were absent in CRISPR-mediated Kv1.6 KO mice (Kcna6 -/-) but were present in a third mouse model expressing exogenous LacZ in nociceptors under the control of a Nav1.8-promoted Cre recombinase. LacZ reporter enzymes are thus intrinsically neurotoxic to sensory neurons and may induce pathologic defects in transgenic mice, which has confounding implications for the interpretation of gene KOs using lacZ Nonetheless, in Kcna6 -/- mice not affected by LacZ, we demonstrated a significant role for Kv1.6 regulating acute noxious thermal sensitivity, and both mechanical and thermal pain-related hypersensitivity after nerve injury.SIGNIFICANCE STATEMENT In recent decades, the expansion of technologies to experimentally manipulate the rodent genome has contributed significantly to the field of neuroscience. While introduction of enzymatic or fluorescent reporter proteins to label neuronal populations is now commonplace, often potential toxicity effects are not fully considered. We show a role of Kv1.6 in acute and neuropathic pain states through analysis of two mouse models lacking Kv1.6 potassium channels: one with additional expression of LacZ and one without. We show that LacZ reporter enzymes induce unintended defects in sensory neurons, with an impact on behavioral data outcomes. To summarize we highlight the importance of Kv1.6 in recovery of normal sensory function following nerve injury, and careful interpretation of data from LacZ reporter models.
Collapse
|
22
|
Middleton SJ, Perez-Sanchez J, Dawes JM. The structure of sensory afferent compartments in health and disease. J Anat 2021; 241:1186-1210. [PMID: 34528255 PMCID: PMC9558153 DOI: 10.1111/joa.13544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/12/2021] [Accepted: 08/30/2021] [Indexed: 12/20/2022] Open
Abstract
Primary sensory neurons are a heterogeneous population of cells able to respond to both innocuous and noxious stimuli. Like most neurons they are highly compartmentalised, allowing them to detect, convey and transfer sensory information. These compartments include specialised sensory endings in the skin, the nodes of Ranvier in myelinated axons, the cell soma and their central terminals in the spinal cord. In this review, we will highlight the importance of these compartments to primary afferent function, describe how these structures are compromised following nerve damage and how this relates to neuropathic pain.
Collapse
Affiliation(s)
- Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - John M Dawes
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Caprini D, Schwartz S, Lanza E, Milanetti E, Lucente V, Ferrarese G, Chiodo L, Nicoletti M, Folli V. A Shearless Microfluidic Device Detects a Role in Mechanosensitivity for AWC ON Neuron in Caenorhabditis elegans. Adv Biol (Weinh) 2021; 5:e2100927. [PMID: 34423577 DOI: 10.1002/adbi.202100927] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/03/2021] [Indexed: 11/08/2022]
Abstract
AWC olfactory neurons are fundamental for chemotaxis toward volatile attractants in Caenorhabditis elegans. Here, it is shown that AWCON responds not only to chemicals but also to mechanical stimuli caused by fluid flow changes in a microfluidic device. The dynamics of calcium events are correlated with the stimulus amplitude. It is further shown that the mechanosensitivity of AWCON neurons has an intrinsic nature rather than a synaptic origin, and the calcium transient response is mediated by TAX-4 cGMP-gated cation channel, suggesting the involvement of one or more "odorant" receptors in AWCON mechano-transduction. In many cases, the responses show plateau properties resembling bistable calcium dynamics where neurons can switch from one stable state to the other. To investigate the unprecedentedly observed mechanosensitivity of AWCON neurons, a novel microfluidic device is designed to minimize the fluid shear flow in the arena hosting the nematodes. Animals in this device show reduced neuronal activation of AWCON neurons. The results observed indicate that the tangential component of the mechanical stress is the main contributor to the mechanosensitivity of AWCON . Furthermore, the microfluidic platform, integrating shearless perfusion and calcium imaging, provides a novel and more controlled solution for in vivo analysis both in micro-organisms and cultured cells.
Collapse
Affiliation(s)
- Davide Caprini
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy
| | - Silvia Schwartz
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy
| | - Enrico Lanza
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy
| | - Edoardo Milanetti
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy.,Department of Physics, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Valeria Lucente
- CREST OPTICS S.p.A., Via di Torre Rossa 66, Rome, 00165, Italy
| | - Giuseppe Ferrarese
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy.,Department of Engineering, Campus Bio-Medico University, Via Álvaro del Portillo 21, Rome, 00128, Italy
| | - Letizia Chiodo
- Department of Engineering, Campus Bio-Medico University, Via Álvaro del Portillo 21, Rome, 00128, Italy
| | - Martina Nicoletti
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy.,Department of Engineering, Campus Bio-Medico University, Via Álvaro del Portillo 21, Rome, 00128, Italy
| | - Viola Folli
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, Rome, 00161, Italy
| |
Collapse
|
24
|
Neural excitability increases with axonal resistance between soma and axon initial segment. Proc Natl Acad Sci U S A 2021; 118:2102217118. [PMID: 34389672 DOI: 10.1073/pnas.2102217118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The position of the axon initial segment (AIS) is thought to play a critical role in neuronal excitability. Previous experimental studies have found that a distal shift in AIS position correlates with a reduction in excitability. Yet theoretical work has suggested the opposite, because of increased electrical isolation. A distal shift in AIS position corresponds to an elevation of axial resistance R a We therefore examined how changes in R a at the axon hillock impact the voltage threshold (Vth) of the somatic action potential in L5 pyramidal neurons. Increasing R a by mechanically pinching the axon between the soma and the AIS was found to lower Vth by ∼6 mV. Conversely, decreasing R a by substituting internal ions with higher mobility elevated Vth All R a -dependent changes in Vth could be reproduced in a Hodgkin-Huxley compartmental model. We conclude that in L5 pyramidal neurons, excitability increases with axial resistance and therefore with a distal shift of the AIS.
Collapse
|
25
|
MacDonald DI, Luiz AP, Iseppon F, Millet Q, Emery EC, Wood JN. Silent cold-sensing neurons contribute to cold allodynia in neuropathic pain. Brain 2021; 144:1711-1726. [PMID: 33693512 PMCID: PMC8320254 DOI: 10.1093/brain/awab086] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/29/2020] [Accepted: 12/17/2020] [Indexed: 11/25/2022] Open
Abstract
Patients with neuropathic pain often experience innocuous cooling as excruciating pain. The cell and molecular basis of this cold allodynia is little understood. We used in vivo calcium imaging of sensory ganglia to investigate how the activity of peripheral cold-sensing neurons was altered in three mouse models of neuropathic pain: oxaliplatin-induced neuropathy, partial sciatic nerve ligation, and ciguatera poisoning. In control mice, cold-sensing neurons were few in number and small in size. In neuropathic animals with cold allodynia, a set of normally silent large diameter neurons became sensitive to cooling. Many of these silent cold-sensing neurons responded to noxious mechanical stimuli and expressed the nociceptor markers Nav1.8 and CGRPα. Ablating neurons expressing Nav1.8 resulted in diminished cold allodynia. The silent cold-sensing neurons could also be activated by cooling in control mice through blockade of Kv1 voltage-gated potassium channels. Thus, silent cold-sensing neurons are unmasked in diverse neuropathic pain states and cold allodynia results from peripheral sensitization caused by altered nociceptor excitability.
Collapse
Affiliation(s)
- Donald Iain MacDonald
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Ana P Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Queensta Millet
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Edward C Emery
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| |
Collapse
|
26
|
Joshi V, Strege PR, Farrugia G, Beyder A. Mechanotransduction in gastrointestinal smooth muscle cells: role of mechanosensitive ion channels. Am J Physiol Gastrointest Liver Physiol 2021; 320:G897-G906. [PMID: 33729004 PMCID: PMC8202201 DOI: 10.1152/ajpgi.00481.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mechanosensation, the ability to properly sense mechanical stimuli and transduce them into physiologic responses, is an essential determinant of gastrointestinal (GI) function. Abnormalities in this process result in highly prevalent GI functional and motility disorders. In the GI tract, several cell types sense mechanical forces and transduce them into electrical signals, which elicit specific cellular responses. Some mechanosensitive cells like sensory neurons act as specialized mechanosensitive cells that detect forces and transduce signals into tissue-level physiological reactions. Nonspecialized mechanosensitive cells like smooth muscle cells (SMCs) adjust their function in response to forces. Mechanosensitive cells use various mechanoreceptors and mechanotransducers. Mechanoreceptors detect and convert force into electrical and biochemical signals, and mechanotransducers amplify and direct mechanoreceptor responses. Mechanoreceptors and mechanotransducers include ion channels, specialized cytoskeletal proteins, cell junction molecules, and G protein-coupled receptors. SMCs are particularly important due to their role as final effectors for motor function. Myogenic reflex-the ability of smooth muscle to contract in response to stretch rapidly-is a critical smooth muscle function. Such rapid mechanotransduction responses rely on mechano-gated and mechanosensitive ion channels, which alter their ion pores' opening in response to force, allowing fast electrical and Ca2+ responses. Although GI SMCs express a variety of such ion channels, their identities remain unknown. Recent advancements in electrophysiological, genetic, in vivo imaging, and multi-omic technologies broaden our understanding of how SMC mechano-gated and mechanosensitive ion channels regulate GI functions. This review discusses GI SMC mechanosensitivity's current developments with a particular emphasis on mechano-gated and mechanosensitive ion channels.
Collapse
Affiliation(s)
- Vikram Joshi
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota
| | - Peter R. Strege
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota
| | - Gianrico Farrugia
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota,2Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Arthur Beyder
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota,2Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
27
|
Lin Z, Bian T, Zhou W, Wang Y, Huang X, Zou J, Zhou H, Niu L, Tang J, Meng L. Modulation of Neuronal Excitability by Low- Intensity Ultrasound in Two Principal Neurons of Rat Anteroventral Cochlear Nucleus. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:1752-1761. [PMID: 33460373 DOI: 10.1109/tuffc.2021.3052203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Ultrasonic neuromodulation has proved to be a promising new approach for direct neuromodulation or potential noninvasive deep brain stimulation technology for treating various neurological disorders. Previous studies have demonstrated that ultrasonic waves can noninvasively diffuse through the intact skull and thus precisely target specific brain regions with high spatial resolution. However, its neuromodulatory effects over different cell types of target nuclei have not been fully elucidated. In the present study, we investigated the neuronal excitability resulted from ultrasound stimulation on the two major neurons of anteroventral cochlear nucleus (AVCN) in vitro. Our results demonstrated that bushy cells (BCs) were well maintaining one action potential (AP) in response to the pairing of a sequence of depolarizing current pulses and 60-s continuous low-intensity ultrasound (LIUS), and meanwhile, stellate cells (SCs) significantly increased the firing rate. The ultrasonic waves with an acoustic pressure of 0.13 MPa were elicited by an on-chip ultrasonic stimulation system compatible with patch-clamp recording. Furthermore, LIUS significantly improved the neuronal excitability in both BCs and SCs based on their intrinsic excitability. Modulation of membrane properties among cell types was due to the LIUS-induced increase in the total inward sodium currents ( INa ) and outward potassium currents ( IKv ). LIUS significantly, at a similar rate, increased the amplitude of total inward sodium currents in both cell types. Meanwhile, LIUS induces a higher rate of the outward potassium currents in the BCs compared with SCs. Therefore, this study could provide new evidence for safe use of ultrasonic neuromodulation and its potential therapy for many auditory diseases, such as the central auditory processing disorder.
Collapse
|
28
|
Murano H, Kaneko T, Zaw SYM, Sone PP, Zaw ZCT, Okada Y, Sunakawa M, Katsube KI, Okiji T. Pulp inflammation induces Kv1.1 K + channel down-regulation in rat thalamus. Oral Dis 2021; 28:1674-1681. [PMID: 33811796 DOI: 10.1111/odi.13866] [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: 06/29/2020] [Revised: 03/07/2021] [Accepted: 03/16/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Signals from inflamed tooth pulp activate thalamic neurons to evoke central sensitization. We aimed to gain insights into the mechanisms mediating the early phase of pulpal inflammation-induced thalamic neural and glial activation. MATERIALS AND METHODS Pulpal inflammation was induced via the application of mustard oil (MO) to the upper first molar of Wistar rats with local anesthesia (LA) or saline injection. After 0.5, 1, 2, and 24 hr, contralateral thalami were subjected to microarrays, a real-time polymerase chain reaction and immunohistochemistry to identify differentially expressed genes and assess potassium voltage-gated channel subfamily A member 1 (Kv1.1)-expressing axons and glial fibrillary acidic protein (GFAP)-expressing astrocytes. RESULTS The Kv1.1 gene (Kcna1) was down-regulated and the density of Kv1.1-expressing axons decreased in non-anesthetized rats, but not in anesthetized rats 1 hr after the MO treatment. The density of GFAP-expressing astrocytes increased in both groups until 24 hr after the MO treatment, with a greater increase being observed in the saline-injection group than in the LA group. CONCLUSIONS MO induced the transient down-regulation of Kcna1, transiently reduced the density of Kv1.1-expressing axons, and increased astrocytes in thalami within 1 hr of pulpal application. These results suggest central sensitization represented by neuronal hyperexcitability and astrocyte activation.
Collapse
Affiliation(s)
- Hiroki Murano
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tomoatsu Kaneko
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Su Yee Myo Zaw
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Phyo Pyai Sone
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Zar Chi Thein Zaw
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yamato Okada
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Mitsuhiro Sunakawa
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | | | - Takashi Okiji
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
29
|
Lacagnina MJ, Heijnen CJ, Watkins LR, Grace PM. Autoimmune regulation of chronic pain. Pain Rep 2021; 6:e905. [PMID: 33981931 PMCID: PMC8108590 DOI: 10.1097/pr9.0000000000000905] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/19/2020] [Accepted: 01/19/2021] [Indexed: 01/24/2023] Open
Abstract
Chronic pain is an unpleasant and debilitating condition that is often poorly managed by existing therapeutics. Reciprocal interactions between the nervous system and the immune system have been recognized as playing an essential role in the initiation and maintenance of pain. In this review, we discuss how neuroimmune signaling can contribute to peripheral and central sensitization and promote chronic pain through various autoimmune mechanisms. These pathogenic autoimmune mechanisms involve the production and release of autoreactive antibodies from B cells. Autoantibodies-ie, antibodies that recognize self-antigens-have been identified as potential molecules that can modulate the function of nociceptive neurons and thereby induce persistent pain. Autoantibodies can influence neuronal excitability by activating the complement pathway; by directly signaling at sensory neurons expressing Fc gamma receptors, the receptors for the Fc fragment of immunoglobulin G immune complexes; or by binding and disrupting ion channels expressed by nociceptors. Using examples primarily from rheumatoid arthritis, complex regional pain syndrome, and channelopathies from potassium channel complex autoimmunity, we suggest that autoantibody signaling at the central nervous system has therapeutic implications for designing novel disease-modifying treatments for chronic pain.
Collapse
Affiliation(s)
- Michael J. Lacagnina
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cobi J. Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linda R. Watkins
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Peter M. Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
30
|
Picton LD, Bertuzzi M, Pallucchi I, Fontanel P, Dahlberg E, Björnfors ER, Iacoviello F, Shearing PR, El Manira A. A spinal organ of proprioception for integrated motor action feedback. Neuron 2021; 109:1188-1201.e7. [PMID: 33577748 DOI: 10.1016/j.neuron.2021.01.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/11/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Proprioception is essential for behavior and provides a sense of our body movements in physical space. Proprioceptor organs are thought to be only in the periphery. Whether the central nervous system can intrinsically sense its own movement remains unclear. Here we identify a segmental organ of proprioception in the adult zebrafish spinal cord, which is embedded by intraspinal mechanosensory neurons expressing Piezo2 channels. These cells are late-born, inhibitory, commissural neurons with unique molecular and physiological profiles reflecting a dual sensory and motor function. The central proprioceptive organ locally detects lateral body movements during locomotion and provides direct inhibitory feedback onto rhythm-generating interneurons responsible for the central motor program. This dynamically aligns central pattern generation with movement outcome for efficient locomotion. Our results demonstrate that a central proprioceptive organ monitors self-movement using hybrid neurons that merge sensory and motor entities into a unified network.
Collapse
Affiliation(s)
- Laurence D Picton
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Maria Bertuzzi
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Irene Pallucchi
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Pierre Fontanel
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Elin Dahlberg
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Francesco Iacoviello
- Electrochemical Innovation Lab, Department of Chemical Engineering, University College London, London, UK
| | - Paul R Shearing
- Electrochemical Innovation Lab, Department of Chemical Engineering, University College London, London, UK
| | | |
Collapse
|
31
|
Fang XZ, Zhou T, Xu JQ, Wang YX, Sun MM, He YJ, Pan SW, Xiong W, Peng ZK, Gao XH, Shang Y. Structure, kinetic properties and biological function of mechanosensitive Piezo channels. Cell Biosci 2021; 11:13. [PMID: 33422128 PMCID: PMC7796548 DOI: 10.1186/s13578-020-00522-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
Mechanotransduction couples mechanical stimulation with ion flux, which is critical for normal biological processes involved in neuronal cell development, pain sensation, and red blood cell volume regulation. Although they are key mechanotransducers, mechanosensitive ion channels in mammals have remained difficult to identify. In 2010, Coste and colleagues revealed a novel family of mechanically activated cation channels in eukaryotes, consisting of Piezo1 and Piezo2 channels. These have been proposed as the long-sought-after mechanosensitive cation channels in mammals. Piezo1 and Piezo2 exhibit a unique propeller-shaped architecture and have been implicated in mechanotransduction in various critical processes, including touch sensation, balance, and cardiovascular regulation. Furthermore, several mutations in Piezo channels have been shown to cause multiple hereditary human disorders, such as autosomal recessive congenital lymphatic dysplasia. Notably, mutations that cause dehydrated hereditary xerocytosis alter the rate of Piezo channel inactivation, indicating the critical role of their kinetics in normal physiology. Given the importance of Piezo channels in understanding the mechanotransduction process, this review focuses on their structural details, kinetic properties and potential function as mechanosensors. We also briefly review the hereditary diseases caused by mutations in Piezo genes, which is key for understanding the function of these proteins.
Collapse
Affiliation(s)
- Xiang-Zhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Zhou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji-Qian Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Xin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Miao-Miao Sun
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Jun He
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shang-Wen Pan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Xiong
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe-Kang Peng
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Hui Gao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
32
|
Li S, Yan Z. Mechanotransduction Ion Channels in Hearing and Touch. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:371-385. [DOI: 10.1007/978-981-16-4254-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
33
|
Mechanical stimulation of the scalp improves the extra- and intracranial blood circulation in humans and mice. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2020. [DOI: 10.1016/j.jtcms.2020.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
34
|
Analgesic Effects of Topical Amitriptyline in Patients With Chemotherapy-Induced Peripheral Neuropathy: Mechanistic Insights From Studies in Mice. THE JOURNAL OF PAIN 2020; 22:440-453. [PMID: 33227509 DOI: 10.1016/j.jpain.2020.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/15/2020] [Accepted: 11/03/2020] [Indexed: 12/25/2022]
Abstract
Oral amitriptyline hydrochloride (amitriptyline) is ineffective against some forms of chronic pain and is often associated with dose-limiting adverse events. We evaluated the potential effectiveness of high-dose topical amitriptyline in a preliminary case series of chemotherapy-induced peripheral neuropathy patients and investigated whether local or systemic adverse events associated with the use of amitriptyline were present in these patients. We also investigated the mechanism of action of topically administered amitriptyline in mice. Our case series suggested that topical 10% amitriptyline treatment was associated with pain relief in chemotherapy-induced peripheral neuropathy patients, without the side effects associated with systemic absorption. Topical amitriptyline significantly increased mechanical withdrawal thresholds when applied to the hind paw of mice, and inhibited the firing responses of C-, Aβ- and Aδ-type peripheral nerve fibers in ex vivo skin-saphenous nerve preparations. Whole-cell patch-clamp recordings on cultured sensory neurons revealed that amitriptyline was a potent inhibitor of the main voltage-gated sodium channels (Nav1.7, Nav1.8, and Nav1.9) found in nociceptors. Calcium imaging showed that amitriptyline activated the transient receptor potential cation channel, TRPA1. Our case series indicated that high-dose 10% topical amitriptyline could alleviate neuropathic pain without adverse local or systemic effects. This analgesic action appeared to be mediated through local inhibition of voltage-gated sodium channels. PERSPECTIVE: Our preliminary case series suggested that topical amitriptyline could provide effective pain relief for chemotherapy-induced peripheral neuropathy patients without any systemic or local adverse events. Investigation of the mechanism of this analgesic action in mice revealed that this activity was mediated through local inhibition of nociceptor Nav channels.
Collapse
|
35
|
Smith PA. K + Channels in Primary Afferents and Their Role in Nerve Injury-Induced Pain. Front Cell Neurosci 2020; 14:566418. [PMID: 33093824 PMCID: PMC7528628 DOI: 10.3389/fncel.2020.566418] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Sensory abnormalities generated by nerve injury, peripheral neuropathy or disease are often expressed as neuropathic pain. This type of pain is frequently resistant to therapeutic intervention and may be intractable. Numerous studies have revealed the importance of enduring increases in primary afferent excitability and persistent spontaneous activity in the onset and maintenance of peripherally induced neuropathic pain. Some of this activity results from modulation, increased activity and /or expression of voltage-gated Na+ channels and hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. K+ channels expressed in dorsal root ganglia (DRG) include delayed rectifiers (Kv1.1, 1.2), A-channels (Kv1.4, 3.3, 3.4, 4.1, 4.2, and 4.3), KCNQ or M-channels (Kv7.2, 7.3, 7.4, and 7.5), ATP-sensitive channels (KIR6.2), Ca2+-activated K+ channels (KCa1.1, 2.1, 2.2, 2.3, and 3.1), Na+-activated K+ channels (KCa4.1 and 4.2) and two pore domain leak channels (K2p; TWIK related channels). Function of all K+ channel types is reduced via a multiplicity of processes leading to altered expression and/or post-translational modification. This also increases excitability of DRG cell bodies and nociceptive free nerve endings, alters axonal conduction and increases neurotransmitter release from primary afferent terminals in the spinal dorsal horn. Correlation of these cellular changes with behavioral studies provides almost indisputable evidence for K+ channel dysfunction in the onset and maintenance of neuropathic pain. This idea is underlined by the observation that selective impairment of just one subtype of DRG K+ channel can produce signs of pain in vivo. Whilst it is established that various mediators, including cytokines and growth factors bring about injury-induced changes in DRG function and excitability, evidence presently available points to a seminal role for interleukin 1β (IL-1β) in control of K+ channel function. Despite the current state of knowledge, attempts to target K+ channels for therapeutic pain management have met with limited success. This situation may change with the advent of personalized medicine. Identification of specific sensory abnormalities and genetic profiling of individual patients may predict therapeutic benefit of K+ channel activators.
Collapse
Affiliation(s)
- Peter A. Smith
- Department of Pharmacology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
36
|
Michel N, Narayanan P, Shomroni O, Schmidt M. Maturational Changes in Mouse Cutaneous Touch and Piezo2-Mediated Mechanotransduction. Cell Rep 2020; 32:107912. [PMID: 32697985 DOI: 10.1016/j.celrep.2020.107912] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/22/2020] [Accepted: 06/25/2020] [Indexed: 01/28/2023] Open
Abstract
The age of studied animals has a profound impact on experimental outcomes in animal-based research. In mice, age influences molecular, morphological, physiological, and behavioral parameters, particularly during rapid postnatal growth and maturation until adulthood (at 12 weeks of age). Despite this knowledge, most biomedical studies use a wide-spanning age range from 4 to 12 weeks, raising concerns about reproducibility and potential masking of relevant age differences. Here, using mouse behavior and electrophysiology in cultured dorsal root ganglia (DRG), we reveal a decline in behavioral cutaneous touch sensitivity and Piezo2-mediated mechanotransduction in vitro during mouse maturation but not thereafter. In addition, we identify distinct transcript changes in individual Piezo2-expressing mechanosensitive DRG neurons by combining electrophysiology with single-cell RNA sequencing (patch-seq). Taken together, our study emphasizes the need for accurate age matching and uncovers hitherto unknown maturational plasticity in cutaneous touch at the level of behavior, mechanotransduction, and transcripts.
Collapse
Affiliation(s)
- Niklas Michel
- Max-Planck Institute of Experimental Medicine and University of Goettingen, Somatosensory Signaling and Systems Biology Group, 37075 Goettingen, Germany
| | - Pratibha Narayanan
- Max-Planck Institute of Experimental Medicine and University of Goettingen, Somatosensory Signaling and Systems Biology Group, 37075 Goettingen, Germany
| | - Orr Shomroni
- NGS Integrative Genomics, Department of Human Genetics at the University Medical Center Goettingen (UMG), 37075 Goettingen, Germany
| | - Manuela Schmidt
- Max-Planck Institute of Experimental Medicine and University of Goettingen, Somatosensory Signaling and Systems Biology Group, 37075 Goettingen, Germany.
| |
Collapse
|
37
|
Díaz-Rodríguez SM, López-López D, Herrero-Turrión MJ, Gómez-Nieto R, Canal-Alonso A, Lopéz DE. Inferior Colliculus Transcriptome After Status Epilepticus in the Genetically Audiogenic Seizure-Prone Hamster GASH/Sal. Front Neurosci 2020; 14:508. [PMID: 32528245 PMCID: PMC7264424 DOI: 10.3389/fnins.2020.00508] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/22/2020] [Indexed: 01/31/2023] Open
Abstract
The Genetic Audiogenic Seizure Hamster from Salamanca (GASH/Sal), an animal model of reflex epilepsy, exhibits generalized tonic–clonic seizures in response to loud sound with the epileptogenic focus localized in the inferior colliculus (IC). Ictal events in seizure-prone strains cause gene deregulation in the epileptogenic focus, which can provide insights into the epileptogenic mechanisms. Thus, the present study aimed to determine the expression profile of key genes in the IC of the GASH/Sal after the status epilepticus. For such purpose, we used RNA-Seq to perform a comparative study between the IC transcriptome of GASH/Sal and that of control hamsters both subjected to loud sound stimulation. After filtering for normalization and gene selection, a total of 36 genes were declared differentially expressed from the RNA-seq analysis in the IC. A set of differentially expressed genes were validated by RT-qPCR showing significant differentially expression between GASH/Sal hamsters and Syrian control hamsters. The confirmed differentially expressed genes were classified on ontological categories associated with epileptogenic events similar to those produced by generalized tonic seizures in humans. Subsequently, based on the result of metabolomics, we found the interleukin-4 and 13-signaling, and nucleoside transport as presumably altered routes in the GASH/Sal model. This research suggests that seizures in GASH/Sal hamsters are generated by multiple molecular substrates, which activate biological processes, molecular processes, cellular components and metabolic pathways associated with epileptogenic events similar to those produced by tonic seizures in humans. Therefore, our study supports the use of the GASH/Sal as a valuable animal model for epilepsy research, toward establishing correlations with human epilepsy and searching new biomarkers of epileptogenesis.
Collapse
Affiliation(s)
- Sandra M Díaz-Rodríguez
- Institute of Neurosciences of Castilla y León, University of Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, University of Salamanca, Salamanca, Spain.,Department of Cellular Biology and Pathology, University of Salamanca, Salamanca, Spain
| | - Daniel López-López
- Institute of Neurosciences of Castilla y León, University of Salamanca, Salamanca, Spain
| | - Manuel J Herrero-Turrión
- Institute of Neurosciences of Castilla y León, University of Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, University of Salamanca, Salamanca, Spain.,Neurological Tissue Bank INCYL (BTN-INCYL), Salamanca, Spain
| | - Ricardo Gómez-Nieto
- Institute of Neurosciences of Castilla y León, University of Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, University of Salamanca, Salamanca, Spain.,Department of Cellular Biology and Pathology, University of Salamanca, Salamanca, Spain
| | - Angel Canal-Alonso
- Institute of Biomedical Research of Salamanca, University of Salamanca, Salamanca, Spain.,BISITE Research Group, University of Salamanca, Salamanca, Spain
| | - Dolores E Lopéz
- Institute of Neurosciences of Castilla y León, University of Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, University of Salamanca, Salamanca, Spain.,Department of Cellular Biology and Pathology, University of Salamanca, Salamanca, Spain
| |
Collapse
|
38
|
D’Adamo MC, Liantonio A, Rolland JF, Pessia M, Imbrici P. Kv1.1 Channelopathies: Pathophysiological Mechanisms and Therapeutic Approaches. Int J Mol Sci 2020; 21:ijms21082935. [PMID: 32331416 PMCID: PMC7215777 DOI: 10.3390/ijms21082935] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/19/2020] [Accepted: 04/20/2020] [Indexed: 12/27/2022] Open
Abstract
Kv1.1 belongs to the Shaker subfamily of voltage-gated potassium channels and acts as a critical regulator of neuronal excitability in the central and peripheral nervous systems. KCNA1 is the only gene that has been associated with episodic ataxia type 1 (EA1), an autosomal dominant disorder characterized by ataxia and myokymia and for which different and variable phenotypes have now been reported. The iterative characterization of channel defects at the molecular, network, and organismal levels contributed to elucidating the functional consequences of KCNA1 mutations and to demonstrate that ataxic attacks and neuromyotonia result from cerebellum and motor nerve alterations. Dysfunctions of the Kv1.1 channel have been also associated with epilepsy and kcna1 knock-out mouse is considered a model of sudden unexpected death in epilepsy. The tissue-specific association of Kv1.1 with other Kv1 members, auxiliary and interacting subunits amplifies Kv1.1 physiological roles and expands the pathogenesis of Kv1.1-associated diseases. In line with the current knowledge, Kv1.1 has been proposed as a novel and promising target for the treatment of brain disorders characterized by hyperexcitability, in the attempt to overcome limited response and side effects of available therapies. This review recounts past and current studies clarifying the roles of Kv1.1 in and beyond the nervous system and its contribution to EA1 and seizure susceptibility as well as its wide pharmacological potential.
Collapse
Affiliation(s)
- Maria Cristina D’Adamo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida MDS-2080, Malta; (M.C.D.); (M.P.)
| | - Antonella Liantonio
- Department of Pharmacy–Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | | | - Mauro Pessia
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida MDS-2080, Malta; (M.C.D.); (M.P.)
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain Po Box 17666, UAE
| | - Paola Imbrici
- Department of Pharmacy–Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy;
- Correspondence:
| |
Collapse
|
39
|
Sharif-Naeini R. Role of mechanosensitive ion channels in the sensation of pain. J Neural Transm (Vienna) 2020; 127:407-414. [PMID: 32249335 DOI: 10.1007/s00702-020-02182-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/24/2020] [Indexed: 01/27/2023]
Abstract
Our ability to sense mechanical cues from our environment depend on the capacity of molecular sensor capable of converting mechanical energy into biochemical or electrical signals. This process, termed mechanotransduction, relies on the activity of mechanosensitive ion channels (MSCs) that are expressed in most tissues, including cells of the inner and outer ear, sensory and sympathetic neurons, and vascular cells. However, the precise role these channels play in the physiology of the cells and organs, where they are expressed is not completely understood. In this review, we will explore some of the recent findings on the role of MSCs to our sense of mechanical pain.
Collapse
Affiliation(s)
- Reza Sharif-Naeini
- Department of Physiology and Cell Information Systems, McGill University, Montreal, QC, H3G-0B1, Canada.
| |
Collapse
|
40
|
Cohen E, Kumar R, Zinger T, Priel A, Treinin M. GTL-1, a Calcium Activated TRPM Channel, Enhances Nociception. Front Pharmacol 2020; 10:1567. [PMID: 32009965 PMCID: PMC6978279 DOI: 10.3389/fphar.2019.01567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/03/2019] [Indexed: 12/23/2022] Open
Abstract
C. elegans PVD neurons are conserved for morphology, function and molecular determinants with mammalian polymodal nociceptors. Functions of polymodal nociceptors require activities of multiple ion channels and receptors including members of the TRP family. GTL-1, a member of the TRPM subclass of TRP channels, was previously shown to amplify PVD-mediated responses to optogenetic stimuli. Here we characterize effects of GTL-1 on PVD-mediated behavioral responses to noxious stimuli. We show that GTL-1 is required within PVD for the immediate and enduring response to thermal (cold) stimuli. But, find no significant reduction in percent animals responding to single or to repeated noxious mechanical stimuli. Nevertheless, PVD specific knockdown of gtl-1expression reduces the magnitude of responses to noxious mechanical stimuli. To understand GTL-1's mechanism of action we expressed it in HEK293 cells. Our results show GTL-1-dependent currents induced by activation of a Gαq-coupled Designer Receptor Exclusively Activated by Designer Drugs (DREADD). In addition, using excised patches we show that GTL-1 can be activated by internal calcium. Our results are consistent with indirect, calcium dependent, activation of GTL-1 by noxious stimuli. This mechanism explains the GTL-1-dependent amplification of responses to multiple stimuli optogenetic and sensory in PVD.
Collapse
Affiliation(s)
- Emiliano Cohen
- Department of Medical Neurobiology, Hadassah-Medical School, Jerusalem, Israel
| | - Rakesh Kumar
- Faculty of Medicine, School of Pharmacy, The Institute for Drug Research, Hebrew University, Jerusalem, Israel
| | - Tal Zinger
- Department of Medical Neurobiology, Hadassah-Medical School, Jerusalem, Israel
| | - Avi Priel
- Faculty of Medicine, School of Pharmacy, The Institute for Drug Research, Hebrew University, Jerusalem, Israel
| | - Millet Treinin
- Department of Medical Neurobiology, Hadassah-Medical School, Jerusalem, Israel
| |
Collapse
|
41
|
Strege PR, Mercado-Perez A, Mazzone A, Saito YA, Bernard CE, Farrugia G, Beyder A. SCN5A mutation G615E results in Na V1.5 voltage-gated sodium channels with normal voltage-dependent function yet loss of mechanosensitivity. Channels (Austin) 2019; 13:287-298. [PMID: 31262209 PMCID: PMC6629189 DOI: 10.1080/19336950.2019.1632670] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 06/06/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022] Open
Abstract
SCN5A is expressed in cardiomyocytes and gastrointestinal (GI) smooth muscle cells (SMCs) as the voltage-gated mechanosensitive sodium channel NaV1.5. The influx of Na+ through NaV1.5 produces a fast depolarization in membrane potential, indispensable for electrical excitability in cardiomyocytes and important for electrical slow waves in GI smooth muscle. As such, abnormal NaV1.5 voltage gating or mechanosensitivity may result in channelopathies. SCN5A mutation G615E - found separately in cases of acquired long-QT syndrome, sudden cardiac death, and irritable bowel syndrome - has a relatively minor effect on NaV1.5 voltage gating. The aim of this study was to test whether G615E impacts mechanosensitivity. Mechanosensitivity of wild-type (WT) or G615E-NaV1.5 in HEK-293 cells was examined by shear stress on voltage- or current-clamped whole cells or pressure on macroscopic patches. Unlike WT, voltage-clamped G615E-NaV1.5 showed a loss in shear- and pressure-sensitivity of peak current yet a normal leftward shift in the voltage-dependence of activation. In current-clamp, shear stress led to a significant increase in firing spike frequency with a decrease in firing threshold for WT but not G615E-NaV1.5. Our results show that the G615E mutation leads to functionally abnormal NaV1.5 channels, which cause disruptions in mechanosensitivity and mechano-electrical feedback and suggest a potential contribution to smooth muscle pathophysiology.
Collapse
Affiliation(s)
- Peter R. Strege
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Arnaldo Mercado-Perez
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
- Medical Scientist Training Program (MSTP), Mayo Clinic, Rochester, MN, USA
| | - Amelia Mazzone
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Yuri A. Saito
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Cheryl E. Bernard
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Gianrico Farrugia
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Arthur Beyder
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
42
|
Anderson EO, Schneider ER, Matson JD, Gracheva EO, Bagriantsev SN. TMEM150C/Tentonin3 Is a Regulator of Mechano-gated Ion Channels. Cell Rep 2019; 23:701-708. [PMID: 29669276 PMCID: PMC5929159 DOI: 10.1016/j.celrep.2018.03.094] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/21/2018] [Accepted: 03/20/2018] [Indexed: 11/24/2022] Open
Abstract
Neuronal mechano-sensitivity relies on mechano-gated ion channels, but pathways regulating their activity remain poorly understood. TMEM150C was proposed to mediate mechano-activated current in proprioceptive neurons. Here, we studied functional interaction of TMEM150C with mechano-gated ion channels from different classes (Piezo2, Piezo1, and the potassium channel TREK-1) using two independent methods of mechanical stimulation. We found that TMEM150C significantly prolongs the duration of the mechano-current produced by all three channels, decreases apparent activation threshold in Piezo2, and induces persistent current in Piezo1. We also show that TMEM150C is co-expressed with Piezo2 in trigeminal neurons, expanding its role beyond proprioceptors. Finally, we cloned TMEM150C from the trigeminal neurons of the tactile-foraging domestic duck and showed that it functions similarly to the mouse ortholog, demonstrating evolutionary conservation among vertebrates. Our studies reveal TMEM150C as a general regulator of mechano-gated ion channels from different classes.
Collapse
Affiliation(s)
- Evan O Anderson
- Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Eve R Schneider
- Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jon D Matson
- Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Elena O Gracheva
- Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sviatoslav N Bagriantsev
- Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
43
|
Ciotu CI, Tsantoulas C, Meents J, Lampert A, McMahon SB, Ludwig A, Fischer MJM. Noncanonical Ion Channel Behaviour in Pain. Int J Mol Sci 2019; 20:E4572. [PMID: 31540178 PMCID: PMC6770626 DOI: 10.3390/ijms20184572] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 12/19/2022] Open
Abstract
Ion channels contribute fundamental properties to cell membranes. Although highly diverse in conductivity, structure, location, and function, many of them can be regulated by common mechanisms, such as voltage or (de-)phosphorylation. Primarily considering ion channels involved in the nociceptive system, this review covers more novel and less known features. Accordingly, we outline noncanonical operation of voltage-gated sodium, potassium, transient receptor potential (TRP), and hyperpolarization-activated cyclic nucleotide (HCN)-gated channels. Noncanonical features discussed include properties as a memory for prior voltage and chemical exposure, alternative ion conduction pathways, cluster formation, and silent subunits. Complementary to this main focus, the intention is also to transfer knowledge between fields, which become inevitably more separate due to their size.
Collapse
Affiliation(s)
- Cosmin I Ciotu
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Jannis Meents
- Institute of Physiology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Angelika Lampert
- Institute of Physiology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Stephen B McMahon
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UR, UK
| | - Andreas Ludwig
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Michael J M Fischer
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
44
|
Moehring F, Halder P, Seal RP, Stucky CL. Uncovering the Cells and Circuits of Touch in Normal and Pathological Settings. Neuron 2019; 100:349-360. [PMID: 30359601 DOI: 10.1016/j.neuron.2018.10.019] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 01/18/2023]
Abstract
The sense of touch is fundamental as it provides vital, moment-to-moment information about the nature of our physical environment. Primary sensory neurons provide the basis for this sensation in the periphery; however, recent work demonstrates that touch transduction mechanisms also occur upstream of the sensory neurons via non-neuronal cells such as Merkel cells and keratinocytes. Within the spinal cord, deep dorsal horn circuits transmit innocuous touch centrally and also transform touch into pain in the setting of injury. Here non-neuronal cells play a key role in the induction and maintenance of persistent mechanical pain. This review highlights recent advances in our understanding of mechanosensation, including a growing appreciation for the role of non-neuronal cells in both touch and pain.
Collapse
Affiliation(s)
- Francie Moehring
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Priyabrata Halder
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Center for the Neural Basis of Cognition, Pittsburgh, PA 15213, USA
| | - Rebecca P Seal
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Center for the Neural Basis of Cognition, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, Pittsburgh, PA 15213, USA
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
45
|
Lin Z, Huang X, Zhou W, Zhang W, Liu Y, Bian T, Niu L, Meng L, Guo Y. Ultrasound Stimulation Modulates Voltage-Gated Potassium Currents Associated With Action Potential Shape in Hippocampal CA1 Pyramidal Neurons. Front Pharmacol 2019; 10:544. [PMID: 31178727 PMCID: PMC6538798 DOI: 10.3389/fphar.2019.00544] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/30/2019] [Indexed: 11/28/2022] Open
Abstract
Potassium channels (K+) play an important role in the regulation of cellular signaling. Dysfunction of potassium channels is associated with several severe ion channels diseases, such as long QT syndrome, episodic ataxia and epilepsy. Ultrasound stimulation has proven to be an effective non-invasive tool for the modulation of ion channels and neural activity. In this study, we demonstrate that ultrasound stimulation enables to modulate the potassium currents and has an impact on the shape modulation of action potentials (AP) in the hippocampal pyramidal neurons using whole-cell patch-clamp recordings in vitro. The results show that outward potassium currents in neurons increase significantly, approximately 13%, in response to 30 s ultrasound stimulation. Simultaneously, the increasing outward potassium currents directly decrease the resting membrane potential (RMP) from −64.67 ± 1.10 mV to −67.51 ± 1.35 mV. Moreover, the threshold current and AP fall rate increase while the reduction of AP half-width and after-hyperpolarization peak time is detected. During ultrasound stimulation, reduction of the membrane input resistance of pyramidal neurons can be found and shorter membrane time constant is achieved. Additionally, we verify that the regulation of potassium currents and shape of action potential is mainly due to the mechanical effects induced by ultrasound. Therefore, ultrasound stimulation may offer an alternative tool to treat some ion channels diseases related to potassium channels.
Collapse
Affiliation(s)
- Zhengrong Lin
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaowei Huang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wei Zhou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wenjun Zhang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Key Laboratory of E&M, Ministry of Education and Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| | - Yingzhe Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Sino-Dutch Biomedical and Information Engineering School, Northeastern University, Shenyang, China
| | - Tianyuan Bian
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Sino-Dutch Biomedical and Information Engineering School, Northeastern University, Shenyang, China
| | - Lili Niu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Long Meng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yanwu Guo
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
46
|
Pethő Z, Najder K, Bulk E, Schwab A. Mechanosensitive ion channels push cancer progression. Cell Calcium 2019; 80:79-90. [PMID: 30991298 DOI: 10.1016/j.ceca.2019.03.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023]
Abstract
In many cases, the mechanical properties of a tumor are different from those of the host tissue. Mechanical cues regulate cancer development by affecting both tumor cells and their microenvironment, by altering cell migration, proliferation, extracellular matrix remodeling and metastatic spread. Cancer cells sense mechanical stimuli such as tissue stiffness, shear stress, tissue pressure of the extracellular space (outside-in mechanosensation). These mechanical cues are transduced into a cellular response (e. g. cell migration and proliferation; inside-in mechanotransduction) or to a response affecting the microenvironment (e. g. inducing a fibrosis or building up growth-induced pressure; inside-out mechanotransduction). These processes heavily rely on mechanosensitive membrane proteins, prominently ion channels. Mechanosensitive ion channels are involved in the Ca2+-signaling of the tumor and stroma cells, both directly, by mediating Ca2+ influx (e. g. Piezo and TRP channels), or indirectly, by maintaining the electrochemical gradient necessary for Ca2+ influx (e. g. K2P, KCa channels). This review aims to discuss the diverse roles of mechanosenstive ion channels in cancer progression, especially those involved in Ca2+-signaling, by pinpointing their functional relevance in tumor pathophysiology.
Collapse
Affiliation(s)
- Zoltán Pethő
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany.
| | - Karolina Najder
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Etmar Bulk
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Albrecht Schwab
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| |
Collapse
|
47
|
Zhang Z, Qiu W, Gong H, Li G, Jiang Q, Liang P, Zheng H, Zhang P. Low-intensity ultrasound suppresses low-Mg2+-induced epileptiform discharges in juvenile mouse hippocampal slices. J Neural Eng 2019; 16:036006. [DOI: 10.1088/1741-2552/ab0b9a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
48
|
Abstract
Voltage-gated potassium (Kv) channels are increasingly recognised as key regulators of nociceptive excitability. Kcns1 is one of the first potassium channels to be associated with neuronal hyperexcitability and mechanical sensitivity in the rat, as well as pain intensity and risk of developing chronic pain in humans. Here, we show that in mice, Kcns1 is predominantly expressed in the cell body and axons of myelinated sensory neurons positive for neurofilament-200, including Aδ-fiber nociceptors and low-threshold Aβ mechanoreceptors. In the spinal cord, Kcns1 was detected in laminae III to V of the dorsal horn where most sensory A fibers terminate, as well as large motoneurons of the ventral horn. To investigate Kcns1 function specifically in the periphery, we generated transgenic mice in which the gene is deleted in all sensory neurons but retained in the central nervous system. Kcns1 ablation resulted in a modest increase in basal mechanical pain, with no change in thermal pain processing. After neuropathic injury, Kcns1 KO mice exhibited exaggerated mechanical pain responses and hypersensitivity to both noxious and innocuous cold, consistent with increased A-fiber activity. Interestingly, Kcns1 deletion also improved locomotor performance in the rotarod test, indicative of augmented proprioceptive signalling. Our results suggest that restoring Kcns1 function in the periphery may be of some use in ameliorating mechanical and cold pain in chronic states.
Collapse
|
49
|
Startek JB, Boonen B, Talavera K, Meseguer V. TRP Channels as Sensors of Chemically-Induced Changes in Cell Membrane Mechanical Properties. Int J Mol Sci 2019; 20:E371. [PMID: 30654572 PMCID: PMC6359677 DOI: 10.3390/ijms20020371] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/11/2019] [Accepted: 01/12/2019] [Indexed: 12/19/2022] Open
Abstract
Transient Receptor Potential ion channels (TRPs) have been described as polymodal sensors, being responsible for transducing a wide variety of stimuli, and being involved in sensory functions such as chemosensation, thermosensation, mechanosensation, and photosensation. Mechanical and chemical stresses exerted on the membrane can be transduced by specialized proteins into meaningful intracellular biochemical signaling, resulting in physiological changes. Of particular interest are compounds that can change the local physical properties of the membrane, thereby affecting nearby proteins, such as TRP channels, which are highly sensitive to the membrane environment. In this review, we provide an overview of the current knowledge of TRP channel activation as a result of changes in the membrane properties induced by amphipathic structural lipidic components such as cholesterol and diacylglycerol, and by exogenous amphipathic bacterial endotoxins.
Collapse
Affiliation(s)
- Justyna B Startek
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain & Disease Research, Herestraat 49, Campus Gasthuisberg O&N1 bus 802, 3000 Leuven, Belgium.
| | - Brett Boonen
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain & Disease Research, Herestraat 49, Campus Gasthuisberg O&N1 bus 802, 3000 Leuven, Belgium.
| | - Karel Talavera
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven; VIB Center for Brain & Disease Research, Herestraat 49, Campus Gasthuisberg O&N1 bus 802, 3000 Leuven, Belgium.
| | - Victor Meseguer
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández y CSIC, E-03550 Alicante , Spain.
| |
Collapse
|
50
|
Feng XJ, Ma LX, Jiao C, Kuang HX, Zeng F, Zhou XY, Cheng XE, Zhu MY, Zhang DY, Jiang CY, Liu T. Nerve injury elevates functional Cav3.2 channels in superficial spinal dorsal horn. Mol Pain 2019; 15:1744806919836569. [PMID: 30803310 PMCID: PMC6458665 DOI: 10.1177/1744806919836569] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/02/2019] [Accepted: 02/12/2019] [Indexed: 01/23/2023] Open
Abstract
Cav3 channels play an important role in modulating chronic pain. However, less is known about the functional changes of Cav3 channels in superficial spinal dorsal horn in neuropathic pain states. Here, we examined the effect of partial sciatic nerve ligation (PSNL) on either expression or electrophysiological properties of Cav3 channels in superficial spinal dorsal horn. Our in vivo studies showed that the blockers of Cav3 channels robustly alleviated PSNL-induced mechanical allodynia and thermal hyperalgesia, which lasted at least 14 days following PSNL. Meanwhile, PSNL triggered an increase in both mRNA and protein levels of Cav3.2 but not Cav3.1 or Cav3.3 in rats. However, in Cav3.2 knockout mice, PSNL predominantly attenuated mechanical allodynia but not thermal hyperalgesia. In addition, the results of whole-cell patch-clamp recordings showed that both the overall proportion of Cav3 current-expressing neurons and the Cav3 current density in individual neurons were elevated in spinal lamina II neurons from PSNL rats, which could not be recapitulated in Cav3.2 knockout mice. Altogether, our findings reveal that the elevated functional Cav3.2 channels in superficial spinal dorsal horn may contribute to the mechanical allodynia in PSNL-induced neuropathic pain model.
Collapse
Affiliation(s)
- Xiao-Jin Feng
- Center for Experimental Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Long-Xian Ma
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Cui Jiao
- Department of Pediatrics, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hai-Xia Kuang
- Department of Pediatrics, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fei Zeng
- Department of Pain Clinic, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xue-Ying Zhou
- Department of Pediatrics, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao-E Cheng
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Meng-Ye Zhu
- Department of Pain Clinic, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Da-Ying Zhang
- Department of Pain Clinic, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chang-Yu Jiang
- Jisheng Han Academician Workstation for Pain Medicine, Nanshan Hospital, Shenzhen, China
| | - Tao Liu
- Center for Experimental Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Pediatrics, the First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|