1
|
Li XH, Guo D, Chen LQ, Chang ZH, Shi JX, Hu N, Chen C, Zhang XW, Bao SQ, Chen MM, Ming D. Low-intensity ultrasound ameliorates brain organoid integration and rescues microcephaly deficits. Brain 2024; 147:3817-3833. [PMID: 38739753 DOI: 10.1093/brain/awae150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
Human brain organoids represent a remarkable platform for modelling neurological disorders and a promising brain repair approach. However, the effects of physical stimulation on their development and integration remain unclear. Here, we report that low-intensity ultrasound significantly increases neural progenitor cell proliferation and neuronal maturation in cortical organoids. Histological assays and single-cell gene expression analyses revealed that low-intensity ultrasound improves the neural development in cortical organoids. Following organoid grafts transplantation into the injured somatosensory cortices of adult mice, longitudinal electrophysiological recordings and histological assays revealed that ultrasound-treated organoid grafts undergo advanced maturation. They also exhibit enhanced pain-related gamma-band activity and more disseminated projections into the host brain than the untreated groups. Finally, low-intensity ultrasound ameliorates neuropathological deficits in a microcephaly brain organoid model. Hence, low-intensity ultrasound stimulation advances the development and integration of brain organoids, providing a strategy for treating neurodevelopmental disorders and repairing cortical damage.
Collapse
Affiliation(s)
- Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Di Guo
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Li-Qun Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Zhe-Han Chang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Jian-Xin Shi
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Nan Hu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Xiao-Wang Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Shuang-Qing Bao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Meng-Meng Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| |
Collapse
|
2
|
Beck CL, Kunze A. Parallelized Mechanical Stimulation of Neuronal Calcium Through Cell-Internal Nanomagnetic Forces Provokes Lasting Shifts in the Network Activity State. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2406678. [PMID: 39460486 DOI: 10.1002/smll.202406678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Neurons differentiate mechanical stimuli force and rate to elicit unique functional responses, driving the need for further tools to generate various mechanical stimuli. Here, cell-internal nanomagnetic forces (iNMF) are introduced by manipulating internalized magnetic nanoparticles with an external magnetic field across cortical neuron networks in vitro. Under iNMF, cortical neurons exhibit calcium (Ca2+) influx, leading to modulation of activity observed through Ca2+ event rates. Inhibiting particle uptake or altering nanoparticle exposure time reduced the neuronal response to nanomagnetic forces, exposing the requirement of nanoparticle uptake to induce the Ca2+ response. In highly active cortical networks, iNMF robustly modulates synchronous network activity, which is lasting and repeatable. Using pharmacological blockers, it is shown that iNMF activates mechanosensitive ion channels to induce the Ca2+ influx. Then, in contrast to transient mechanically evoked neuronal activity, iNMF activates Ca2+-activated potassium (KCa) channels to stabilize the neuronal membrane potential and induce network activity shifts. The findings reveal the potential of magnetic nanoparticle-mediated mechanical stimulation to modulate neuronal circuit dynamics, providing insights into the biophysics of neuronal computation.
Collapse
Affiliation(s)
- Connor L Beck
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, MT, 59717, USA
| | - Anja Kunze
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, MT, 59717, USA
- Montana Nanotechnology Facility, Montana State University, Bozeman, MT, 59717, USA
- Optical Technology Center, Montana State University, Bozeman, MT, 59717, USA
| |
Collapse
|
3
|
Murphy KR, Farrell JS, Bendig J, Mitra A, Luff C, Stelzer IA, Yamaguchi H, Angelakos CC, Choi M, Bian W, DiIanni T, Pujol EM, Matosevich N, Airan R, Gaudillière B, Konofagou EE, Butts-Pauly K, Soltesz I, de Lecea L. Optimized ultrasound neuromodulation for non-invasive control of behavior and physiology. Neuron 2024; 112:3252-3266.e5. [PMID: 39079529 DOI: 10.1016/j.neuron.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/09/2024] [Accepted: 07/02/2024] [Indexed: 08/09/2024]
Abstract
Focused ultrasound can non-invasively modulate neural activity, but whether effective stimulation parameters generalize across brain regions and cell types remains unknown. We used focused ultrasound coupled with fiber photometry to identify optimal neuromodulation parameters for four different arousal centers of the brain in an effort to yield overt changes in behavior. Applying coordinate descent, we found that optimal parameters for excitation or inhibition are highly distinct, the effects of which are generally conserved across brain regions and cell types. Optimized stimulations induced clear, target-specific behavioral effects, whereas non-optimized protocols of equivalent energy resulted in substantially less or no change in behavior. These outcomes were independent of auditory confounds and, contrary to expectation, accompanied by a cyclooxygenase-dependent and prolonged reduction in local blood flow and temperature with brain-region-specific scaling. These findings demonstrate that carefully tuned and targeted ultrasound can exhibit powerful effects on complex behavior and physiology.
Collapse
Affiliation(s)
- Keith R Murphy
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Jordan S Farrell
- Department of Neurosurgery, Stanford University, Stanford, CA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Center, Harvard Medical School, Boston, MA, USA
| | - Jonas Bendig
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Anish Mitra
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Charlotte Luff
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Ina A Stelzer
- Department of Anesthesia, Stanford University, Stanford, CA, USA
| | - Hiroshi Yamaguchi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Department of Neuroscience, Nagoya University, Nagoya, Japan
| | | | - Mihyun Choi
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Wenjie Bian
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Tommaso DiIanni
- Department of Radiology, Stanford University, Stanford, CA, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Esther Martinez Pujol
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Noa Matosevich
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Raag Airan
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Brice Gaudillière
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Kim Butts-Pauly
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
4
|
Sánchez-Carranza O, Chakrabarti S, Kühnemund J, Schwaller F, Bégay V, García-Contreras JA, Wang L, Lewin GR. Piezo2 voltage-block regulates mechanical pain sensitivity. Brain 2024; 147:3487-3500. [PMID: 38984717 PMCID: PMC11449130 DOI: 10.1093/brain/awae227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/19/2024] [Accepted: 06/13/2024] [Indexed: 07/11/2024] Open
Abstract
PIEZO2 is a trimeric mechanically-gated ion channel expressed by most sensory neurons in the dorsal root ganglia. Mechanosensitive PIEZO2 channels are also genetically required for normal touch sensation in both mice and humans. We previously showed that PIEZO2 channels are also strongly modulated by membrane voltage. Specifically, it is only at very positive voltages that all channels are available for opening by mechanical force. Conversely, most PIEZO2 channels are blocked at normal negative resting membrane potentials. The physiological function of this unusual biophysical property of PIEZO2 channels, however, remained unknown. We characterized the biophysical properties of three PIEZO2 ion channel mutations at an evolutionarily conserved arginine (R2756). Using genome engineering in mice we generated Piezo2R2756H/R2756H and Piezo2R2756K/R2756K knock-in mice to characterize the physiological consequences of altering PIEZO2 voltage sensitivity in vivo. We measured endogenous mechanosensitive currents in sensory neurons isolated from the dorsal root ganglia and characterized mechanoreceptor and nociceptor function using electrophysiology. Mice were also assessed behaviourally and morphologically. Mutations at the conserved Arginine (R2756) dramatically changed the biophysical properties of the channel relieving voltage block and lowering mechanical thresholds for channel activation. Piezo2R2756H/R2756H and Piezo2R2756K/R2756K knock-in mice that were homozygous for gain-of-function mutations were viable and were tested for sensory changes. Surprisingly, mechanosensitive currents in nociceptors, neurons that detect noxious mechanical stimuli, were substantially sensitized in Piezo2 knock-in mice, but mechanosensitive currents in most mechanoreceptors that underlie touch sensation were only mildly affected by the same mutations. Single-unit electrophysiological recordings from sensory neurons innervating the glabrous skin revealed that rapidly-adapting mechanoreceptors that innervate Meissner's corpuscles exhibited slightly decreased mechanical thresholds in Piezo2 knock-in mice. Consistent with measurements of mechanically activated currents in isolated sensory neurons essentially all cutaneous nociceptors, both fast conducting Aδ-mechanonociceptors and unmyelinated C-fibre nociceptors were substantially more sensitive to mechanical stimuli and indeed acquired receptor properties similar to ultrasensitive touch receptors in Piezo2 knock-in mice. Mechanical stimuli also induced enhanced ongoing activity in cutaneous nociceptors in Piezo2 knock-in mice and hyper-sensitive PIEZO2 channels were sufficient alone to drive ongoing activity, even in isolated nociceptive neurons. Consistently, Piezo2 knock-in mice showed substantial behavioural hypersensitivity to noxious mechanical stimuli. Our data indicate that ongoing activity and sensitization of nociceptors, phenomena commonly found in human chronic pain syndromes, can be driven by relieving the voltage-block of PIEZO2 ion channels. Indeed, membrane depolarization caused by multiple noxious stimuli may sensitize nociceptors by relieving voltage-block of PIEZO2 channels.
Collapse
Affiliation(s)
- Oscar Sánchez-Carranza
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Sampurna Chakrabarti
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Johannes Kühnemund
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Fred Schwaller
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Valérie Bégay
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Jonathan Alexis García-Contreras
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Lin Wang
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
| | - Gary R Lewin
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 10409, Germany
- Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- German Center for Mental Health (DZPG), partner site Berlin, 10117 Berlin, Germany
| |
Collapse
|
5
|
Xu T, Zhang Y, Li D, Lai C, Wang S, Zhang S. Mechanosensitive Ion Channels Piezo1 and Piezo2 Mediate Motor Responses In Vivo During Transcranial Focused Ultrasound Stimulation of the Rodent Cerebral Motor Cortex. IEEE Trans Biomed Eng 2024; 71:2900-2910. [PMID: 38748529 DOI: 10.1109/tbme.2024.3401136] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
OBJECTIVE Transcranial focused ultrasound (tFUS) neuromodulation offers a noninvasive, safe, deep brain stimulation with high precision, presenting potential in understanding neural circuits and treating brain disorders. This in vivo study investigated the mechanism of tFUS in activating the opening of the mechanosensitive ion channels Piezo1 and Piezo2 in the mouse motor cortex to induce motor responses. METHODS Piezo1 and Piezo2 were knocked down separately in the mouse motor cortex, followed by EMG and motor cortex immunofluorescence comparisons before and after knockdown under tFUS stimulation. RESULTS The results demonstrated that the stimulation-induced motor response success rates in Piezo knockdown mice were lower compared to the control group (Piezo1 knockdown: 57.63% ± 14.62%, Piezo2 knockdown: 73.71% ± 13.10%, Control mice: 85.69% ± 10.23%). Both Piezo1 and Piezo2 knockdowns showed prolonged motor response times (Piezo1 knockdown: 0.62 ± 0.19 s, Piezo2 knockdown: 0.60 ± 0.13 s, Control mice: 0.44 ± 0.12 s) compared to controls. Additionally, Piezo knockdown animals subjected to tFUS showed reduced immunofluorescent c-Fos expression in the target area when measured in terms of cells per unit area compared to the control group. CONCLUSION This in vivo study confirms the pivotal role of Piezo channels in tFUS-induced neuromodulation, highlighting their influence on motor response efficacy and timing. SIGNIFICANCE This study provides insights into the mechanistic underpinnings of noninvasive brain stimulation techniques and opens avenues for developing targeted therapies for neural disorders.
Collapse
|
6
|
Garcia-Sanchez J, Lin D, Liu WW. Mechanosensitive ion channels in glaucoma pathophysiology. Vision Res 2024; 223:108473. [PMID: 39180975 PMCID: PMC11398070 DOI: 10.1016/j.visres.2024.108473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/27/2024]
Abstract
Force sensing is a fundamental ability that allows cells and organisms to interact with their physical environment. The eye is constantly subjected to mechanical forces such as blinking and eye movements. Furthermore, elevated intraocular pressure (IOP) can cause mechanical strain at the optic nerve head, resulting in retinal ganglion cell death (RGC) in glaucoma. How mechanical stimuli are sensed and affect cellular physiology in the eye is unclear. Recent studies have shown that mechanosensitive ion channels are expressed in many ocular tissues relevant to glaucoma and may influence IOP regulation and RGC survival. Furthermore, variants in mechanosensitive ion channel genes may be associated with risk for primary open angle glaucoma. These findings suggest that mechanosensitive channels may be important mechanosensors mediating cellular responses to pressure signals in the eye. In this review, we focus on mechanosensitive ion channels from three major channel families-PIEZO, two-pore potassium and transient receptor potential channels. We review the key properties of these channels, their effects on cell function and physiology, and discuss their possible roles in glaucoma pathophysiology.
Collapse
Affiliation(s)
- Julian Garcia-Sanchez
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Danting Lin
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wendy W Liu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
7
|
Zou J, Chen H, Chen X, Lin Z, Yang Q, Tie C, Wang H, Niu L, Guo Y, Zheng H. Noninvasive closed-loop acoustic brain-computer interface for seizure control. Theranostics 2024; 14:5965-5981. [PMID: 39346532 PMCID: PMC11426232 DOI: 10.7150/thno.99820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/03/2024] [Indexed: 10/01/2024] Open
Abstract
Rationale: The brain-computer interface (BCI) is core tasks in comprehensively understanding the brain, and is one of the most significant challenges in neuroscience. The development of novel non-invasive neuromodulation technique will drive major innovations and breakthroughs in the field of BCI. Methods: We develop a new noninvasive closed-loop acoustic brain-computer interface (aBCI) for decoding the seizure onset based on the electroencephalography and triggering ultrasound stimulation of the vagus nerve to terminate seizures. Firstly, we create the aBCI system and decode the onset of seizure via a multi-level threshold model based on the analysis of wireless-collected electroencephalogram (EEG) signals recorded from above the hippocampus. Then, the different acoustic parameters induced acoustic radiation force were used to stimulate the vagus nerve in a rat model of epilepsy-induced by pentylenetetrazole. Finally, the results of epileptic EEG signal triggering ultrasound stimulation of the vagus nerve to control seizures. In addition, the mechanism of aBCI control seizures were investigated by real-time quantitative polymerase chain reaction (RT-qPCR). Results: In a rat model of epilepsy, the aBCI system selectively actives mechanosensitive neurons in the nodose ganglion while suppressing neuronal excitability in the hippocampus and amygdala, and stops seizures rapidly upon ultrasound stimulation of the vagus nerve. Physical transection or chemical blockade of the vagus nerve pathway abolish the antiepileptic effects of aBCI. In addition, aBCI shows significant antiepileptic effects compared to conventional vagus nerve electrical stimulation in an acute experiment. Conclusions: Closed-loop aBCI provides a novel, safe and effective tool for on-demand stimulation to treat abnormal neuronal discharges, opening the door to next generation non-invasive BCI.
Collapse
Affiliation(s)
- Junjie Zou
- 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 510282, China
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Houminji Chen
- 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 510282, China
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaoyan Chen
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhengrong Lin
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qihang Yang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Changjun Tie
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hong Wang
- The Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lili Niu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, 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 510282, China
| | - Hairong Zheng
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
8
|
Tang J, Feng M, Wang D, Zhang L, Yang K. Recent advancement of sonogenetics: A promising noninvasive cellular manipulation by ultrasound. Genes Dis 2024; 11:101112. [PMID: 38947740 PMCID: PMC11214298 DOI: 10.1016/j.gendis.2023.101112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 07/02/2024] Open
Abstract
Recent advancements in biomedical research have underscored the importance of noninvasive cellular manipulation techniques. Sonogenetics, a method that uses genetic engineering to produce ultrasound-sensitive proteins in target cells, is gaining prominence along with optogenetics, electrogenetics, and magnetogenetics. Upon stimulation with ultrasound, these proteins trigger a cascade of cellular activities and functions. Unlike traditional ultrasound modalities, sonogenetics offers enhanced spatial selectivity, improving precision and safety in disease treatment. This technology broadens the scope of non-surgical interventions across a wide range of clinical research and therapeutic applications, including neuromodulation, oncologic treatments, stem cell therapy, and beyond. Although current literature predominantly emphasizes ultrasonic neuromodulation, this review offers a comprehensive exploration of sonogenetics. We discuss ultrasound properties, the specific ultrasound-sensitive proteins employed in sonogenetics, and the technique's potential in managing conditions such as neurological disorders, cancer, and ophthalmic diseases, and in stem cell therapies. Our objective is to stimulate fresh perspectives for further research in this promising field.
Collapse
Affiliation(s)
- Jin Tang
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Mingxuan Feng
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Dong Wang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Liang Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ke Yang
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| |
Collapse
|
9
|
He Z, Liu Q, Yang R, Zhou Y, Liu X, Deng H, Cong H, Liu Y, Liao L. Low-Intensity Ultrasound Tibial Nerve Stimulation Suppresses Bladder Activity in Rats. Neuromodulation 2024:S1094-7159(24)00133-8. [PMID: 39078346 DOI: 10.1016/j.neurom.2024.06.005] [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: 02/19/2024] [Revised: 05/21/2024] [Accepted: 06/15/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND AND OBJECTIVE Noninvasive neuromodulation, particularly through low-intensity ultrasound, holds promise in the fields of neuroscience and neuro-engineering. Ultrasound can stimulate the central nervous system to treat neurologic disorders of the brain and activate peripheral nerve activity. The aim of this study is to investigate the inhibitory effect of low-intensity ultrasonic tibial nerve stimulation on both the physiological state and the overactive bladder (OAB) model in rats. MATERIALS AND METHODS A total of 28 female Sprague-Dawley rats were used in this study. Continuous transurethral instillation of 0.9% normal saline into the bladder was initially performed to stimulate physiological bladder activity. Subsequently, a solution containing 0.3% acetic acid dissolved in saline was instilled to induce rat models of OAB. The study comprised two phases: initial observation of bladder response to low-intensity ultrasound (1 MHz, 1 W/cm2, 50% duty cycle) in seven rats; subsequent exploration of ultrasound frequency (3 MHz) and intensity (2 W/cm2 and 3 W/cm2) effects in 21 rats. The intercontraction intervals (ICIs) were the primary outcome measure. Histologic analysis of tibial nerves and surrounding muscle tissues determined safe ultrasound parameters. RESULTS Low-intensity ultrasound tibial nerve stimulation significantly inhibited normal and OAB activity. Ultrasound stimulation at 1 MHz, 1 W/cm2, with a 50% duty cycle significantly prolonged the ICI in both normal (p < 0.0001) and OAB rats (p < 0.01), as did transitioning to a 3 MHz frequency (p = 0.001 for normal rats; p < 0.01 for OAB rats). Similarly, at an intensity of 2 W/cm2 and 1 MHz frequency with a 50% duty cycle, ultrasound stimulation significantly prolonged the ICI in both normal (p < 0.01) and OAB rats (p < 0.005). Furthermore, switching to a 3 W/cm2 ultrasound intensity also significantly extended the ICI in both normal (p < 0.05) and OAB rats (p = 0.01). However, after different ultrasound intensities and frequencies, there was no statistical difference in ICI ratios (preultrasound stimulation vs postultrasound stimulation/preultrasound stimulation ∗ 100%) in all rats (p > 0.05). Low-intensity ultrasound tibial nerve stimulation did not influence baseline pressure, threshold pressure, or maximum pressure. In addition, a latency period in bladder reflex inhibition was induced by low-intensity ultrasound tibial nerve stimulation in some rats. Histologic analysis indicated no evident nerve or muscle tissue damage or abnormalities. CONCLUSIONS This study confirmed the potential of transcutaneous ultrasound tibial nerve stimulation to improve bladder function. According to the findings, the ultrasonic intensities ranging from 1 to 3 W/cm2 and frequencies of 1 MHz and 3 MHz are both feasible and safe treatment parameters. This study portended the promise of low-intensity ultrasound tibial nerve stimulation as a treatment for OAB and provides a basis and reference for future clinical applications.
Collapse
Affiliation(s)
- Zitian He
- Department of Rehabilitation, Yuying Children's Hospital, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Urology, China Rehabilitation Research Center, Beijing, China; The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qinggang Liu
- Department of Urology, China Rehabilitation Research Center, Beijing, China; Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Ruiyao Yang
- Department of Urology, China Rehabilitation Research Center, Beijing, China
| | - Yongheng Zhou
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Urology, China Rehabilitation Research Center, Beijing, China; University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Xin Liu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Urology, China Rehabilitation Research Center, Beijing, China; University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Han Deng
- Department of Urology, China Rehabilitation Research Center, Beijing, China; School of Rehabilitation, Capital Medical University, Beijing, China
| | - Huiling Cong
- Department of Urology, China Rehabilitation Research Center, Beijing, China; School of Rehabilitation, Capital Medical University, Beijing, China
| | - Yixi Liu
- Department of Urology, China Rehabilitation Research Center, Beijing, China; School of Rehabilitation, Capital Medical University, Beijing, China
| | - Limin Liao
- Department of Rehabilitation, Yuying Children's Hospital, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Urology, China Rehabilitation Research Center, Beijing, China; The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China; Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; University of Health and Rehabilitation Sciences, Qingdao, Shandong, China; School of Rehabilitation, Capital Medical University, Beijing, China.
| |
Collapse
|
10
|
Horii K, Ogawa B, Nagase N, Morimoto I, Abe C, Ogawa T, Choi S, Nin F. The cochlear hook region detects harmonics beyond the canonical hearing range. PNAS NEXUS 2024; 3:pgae280. [PMID: 39055687 PMCID: PMC11272074 DOI: 10.1093/pnasnexus/pgae280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Ultrasound, or sound at frequencies exceeding the conventional range of human hearing, is not only audible to mice, microbats, and dolphins, but also creates an auditory sensation when delivered through bone conduction in humans. Although ultrasound is utilized for brain activation and in hearing aids, the physiological mechanism of ultrasonic hearing remains unknown. In guinea pigs, we found that ultrasound above the hearing range delivered through ossicles of the middle ear evokes an auditory brainstem response and a mechano-electrical transduction current through hair cells, as shown by the local field potential called the cochlear microphonic potential (CM). The CM synchronizes with ultrasound, and like the response to audible sounds is actively and nonlinearly amplified. In vivo optical nano-vibration analysis revealed that the sensory epithelium in the hook region, the basal extreme of the cochlear turns, resonates in response both to ultrasound within the hearing range and to harmonics beyond the hearing range. The results indicate that hair cells can respond to stimulation at the optimal frequency and its harmonics, and the hook region detects ultrasound stimuli with frequencies more than two octaves higher than the upper limit of the ordinary hearing range.
Collapse
Affiliation(s)
- Kazuhiro Horii
- Division of Biological Principles, Department of Physiology and Biophysics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Bakushi Ogawa
- Division of Biological Principles, Department of Physiology and Biophysics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
- Division of Sensorimotor Medicine, Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Noriko Nagase
- Division of Biological Principles, Department of Physiology and Biophysics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
- Division of Sensorimotor Medicine, Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Iori Morimoto
- Division of Biological Principles, Department of Physiology and Biophysics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Chikara Abe
- Division of Biological Principles, Department of Physiology and Biophysics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Takenori Ogawa
- Division of Sensorimotor Medicine, Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Samuel Choi
- Graduate School of Science and Technology, Niigata University, 8050 Ikarashi, Nishi-ku, Niigata, 950-2181, Japan
| | - Fumiaki Nin
- Division of Biological Principles, Department of Physiology and Biophysics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| |
Collapse
|
11
|
Pellow C, Pichardo S, Pike GB. A systematic review of preclinical and clinical transcranial ultrasound neuromodulation and opportunities for functional connectomics. Brain Stimul 2024; 17:734-751. [PMID: 38880207 DOI: 10.1016/j.brs.2024.06.005] [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: 03/01/2024] [Revised: 05/21/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Low-intensity transcranial ultrasound has surged forward as a non-invasive and disruptive tool for neuromodulation with applications in basic neuroscience research and the treatment of neurological and psychiatric conditions. OBJECTIVE To provide a comprehensive overview and update of preclinical and clinical transcranial low intensity ultrasound for neuromodulation and emphasize the emerging role of functional brain mapping to guide, better understand, and predict responses. METHODS A systematic review was conducted by searching the Web of Science and Scopus databases for studies on transcranial ultrasound neuromodulation, both in humans and animals. RESULTS 187 relevant studies were identified and reviewed, including 116 preclinical and 71 clinical reports with subjects belonging to diverse cohorts. Milestones of ultrasound neuromodulation are described within an overview of the broader landscape. General neural readouts and outcome measures are discussed, potential confounds are noted, and the emerging use of functional magnetic resonance imaging is highlighted. CONCLUSION Ultrasound neuromodulation has emerged as a powerful tool to study and treat a range of conditions and its combination with various neural readouts has significantly advanced this platform. In particular, the use of functional magnetic resonance imaging has yielded exciting inferences into ultrasound neuromodulation and has the potential to advance our understanding of brain function, neuromodulatory mechanisms, and ultimately clinical outcomes. It is anticipated that these preclinical and clinical trials are the first of many; that transcranial low intensity focused ultrasound, particularly in combination with functional magnetic resonance imaging, has the potential to enhance treatment for a spectrum of neurological conditions.
Collapse
Affiliation(s)
- Carly Pellow
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada.
| | - Samuel Pichardo
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada; Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| | - G Bruce Pike
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada; Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| |
Collapse
|
12
|
Olaitan GO, Ganesana M, Strohman A, Lynch WJ, Legon W, Jill Venton B. Focused Ultrasound Modulates Dopamine in a Mesolimbic Reward Circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580202. [PMID: 38979318 PMCID: PMC11230179 DOI: 10.1101/2024.02.13.580202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Dopamine is a neurotransmitter that plays a significant role in reward and motivation. Dysfunction in the mesolimbic dopamine pathway has been linked to a variety of psychiatric disorders, including addiction. Low-intensity focused ultrasound (LIFU) has demonstrated effects on brain activity, but how LIFU affects dopamine neurotransmission is not known. Here, we applied three different intensities (6.5, 13, and 26 W/cm 2 I sppa ) of 2-minute LIFU to the prelimbic region (PLC) and measured dopamine in the nucleus accumbens (NAc) core using fast-scan cyclic voltammetry. Two minutes of LIFU sonication at 13 W/cm 2 to the PLC significantly reduced dopamine release by ∼ 50% for up to 2 hours. However, double the intensity (26 W/cm 2 ) resulted in less inhibition (∼30%), and half the intensity (6.5 W/cm 2 ) did not result in any inhibition of dopamine. Anatomical controls applying LIFU to the primary somatosensory cortex did not change NAc core dopamine, and applying LIFU to the PLC did not affect dopamine release in the caudate or NAc shell. Histological evaluations showed no evidence of cell damage or death. Modeling of temperature rise demonstrates a maximum temperature change of 0.5°C with 13 W/cm 2 , suggesting that modulation is not due to thermal mechanisms. These studies show that LIFU at a moderate intensity provides a noninvasive, high spatial resolution means to modulate specific mesolimbic circuits that could be used in future studies to target and repair pathways that are dysfunctional in addiction and other psychiatric diseases.
Collapse
|
13
|
Guo C, Cun Y, Xia B, Chen S, Zhang C, Chen Y, Shan E, Zhang P, Tai X. An analysis of stimulation methods used in rehabilitation equipment for children with cerebral palsy. Front Neurol 2024; 15:1371332. [PMID: 38966084 PMCID: PMC11223519 DOI: 10.3389/fneur.2024.1371332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/31/2024] [Indexed: 07/06/2024] Open
Abstract
Objective This paper summarizes the research progress into stimulation methods used in rehabilitation equipment for pediatric cerebral palsy (CP) for the past 20 years from 2003 to 2023. We also provide ideas for innovative research and development of artificial intelligence-based rehabilitation equipment. Methods Through a certain search strategy, Keywords are searched in the China National Knowledge Network Database (CNKI), the Wanfang Database knowledge service platform, the Chongqing VIP information service, PubMed, Web of Science, Cochrane, ScienceDirect, Medline, Embase, and IEEE database. A total of 3,049 relevant articles were retrieved, and 49 articles were included that mentioned research and development of rehabilitation equipment. We excluded articles that were not specific to children with CP, were duplicated or irrelevant literature, were missing data, the full article was not available, the article did not describe the method of stimulation used with the rehabilitation equipment on children with CP, were not Chinese and English, and were the types of reviews and commentaries. Results Physical stimulation is the main stimulation method of rehabilitation equipment for children with CP. Force stimulation is the main mode of physical stimulation, and there are 17 articles that have verified the clinical efficacy of force stimulation-based equipment. Conclusion Research on the stimulation mode of pediatric cerebral palsy rehabilitation equipment is likely to focus on simulating the force of the Chinese medicine called "tuina manipulation." When this method is combined with artificial intelligence and personalized direction we believe this will lay the foundation for future development of a novel therapy for children with CP.
Collapse
Affiliation(s)
- Cunxiao Guo
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture and Tuina Rehabilitation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yongdan Cun
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture and Tuina Rehabilitation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan College of Business Management, Kunming, China
| | - Bo Xia
- Master of Science in Computer Science, Sofia University, Palo Alto, CA, United States
| | - Suyu Chen
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture and Tuina Rehabilitation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Can Zhang
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture and Tuina Rehabilitation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yiping Chen
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture and Tuina Rehabilitation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Exian Shan
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture and Tuina Rehabilitation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Pengyue Zhang
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture and Tuina Rehabilitation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Xiantao Tai
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture and Tuina Rehabilitation, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
14
|
Lee SA, Kamimura HAS, Smith M, Konofagou EE. Functional Cerebral Neurovascular Mapping During Focused Ultrasound Peripheral Neuromodulation of Neuropathic Pain. IEEE Trans Biomed Eng 2024; 71:1770-1779. [PMID: 38198257 PMCID: PMC11105977 DOI: 10.1109/tbme.2024.3352025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
BACKGROUND Nociceptive pain is required for healthy function, yet, neuropathic pain (disease or injury) can be severely debilitating. Though a wide-array of treatment options are available, they are often systemic and/or invasive. As a promising neuromodulation treatment, Focused ultrasound (FUS) is a noninvasive and highly spatially-targeted technique shown to stimulate neural activity, yet, effects on pain signaling are currently unknown. OBJECTIVE Develop and validate a method for studying FUS nerve stimulation modulation of pain-evoked neural responses in vivo. METHODS We developed a high-resolution functional ultrasound (fUS) method capable of mapping cortical responses in healthy and neuropathic pain mice in response to FUS neuromodulation treatment. RESULTS FUS-evoked hemodynamic responses are correlated with the intensity of peripheral neuromodulation. We confirm functional connectivity is altered in neuropathic mice and demonstrate that FUS can modulate neuropathic pain-evoked hemodynamics. CONCLUSIONS The findings presented herein provides evidence for an FUS-based nerve pain method and validates the fUS technique developed for monitoring pain-evoked hemodynamics. SIGNIFICANCE We anticipate that the findings presented herein describe a noninvasive and flexible nerve modulation technique for pain mitigation, furthering evidence for clinical translation.
Collapse
|
15
|
Sorum B, Docter T, Panico V, Rietmeijer RA, Brohawn SG. Tension activation of mechanosensitive two-pore domain K+ channels TRAAK, TREK-1, and TREK-2. Nat Commun 2024; 15:3142. [PMID: 38605031 PMCID: PMC11009253 DOI: 10.1038/s41467-024-47208-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
TRAAK, TREK-1, and TREK-2 are mechanosensitive two-pore domain K+ (K2P) channels that contribute to action potential propagation, sensory transduction, and muscle contraction. While structural and functional studies have led to models that explain their mechanosensitivity, we lack a quantitative understanding of channel activation by membrane tension. Here, we define the tension response of mechanosensitive K2Ps using patch-clamp recording and imaging. All are low-threshold mechanosensitive channels (T10%/50% 0.6-2.7 / 4.4-6.4 mN/m) with distinct response profiles. TRAAK is most sensitive, TREK-1 intermediate, and TREK-2 least sensitive. TRAAK and TREK-1 are activated broadly over a range encompassing nearly all physiologically relevant tensions. TREK-2, in contrast, activates over a narrower range like mechanosensitive channels Piezo1, MscS, and MscL. We further show that low-frequency, low-intensity focused ultrasound increases membrane tension to activate TRAAK and MscS. This work provides insight into tension gating of mechanosensitive K2Ps relevant to understanding their physiological roles and potential applications for ultrasonic neuromodulation.
Collapse
Affiliation(s)
- Ben Sorum
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, 94720, USA
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, 08103, USA
| | - Trevor Docter
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, 94720, USA
| | - Vincent Panico
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, 94720, USA
| | - Robert A Rietmeijer
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, 94720, USA
| | - Stephen G Brohawn
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA.
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA.
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
16
|
Prieto ML, Maduke M. Towards an ion-channel-centric approach to ultrasound neuromodulation. Curr Opin Behav Sci 2024; 56:101355. [PMID: 38505510 PMCID: PMC10947167 DOI: 10.1016/j.cobeha.2024.101355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Ultrasound neuromodulation is a promising technology that could revolutionize study and treatment of brain conditions ranging from mood disorders to Alzheimer's disease and stroke. An understanding of how ultrasound directly modulates specific ion channels could provide a roadmap for targeting specific neurological circuits and achieving desired neurophysiological outcomes. Although experimental challenges make it difficult to unambiguously identify which ion channels are sensitive to ultrasound in vivo, recent progress indicates that there are likely several different ion channels involved, including members of the K2P, Piezo, and TRP channel families. A recent result linking TRPM2 channels in the hypothalamus to induction of torpor by ultrasound in rodents demonstrates the feasibility of targeting a specific ion channel in a specific population of neurons.
Collapse
Affiliation(s)
- Martin Loynaz Prieto
- Department of Molecular and Cellular Physiology, Stanford University, 279 Campus Drive West, B151 Beckman Center, Stanford, CA 94305
| | - Merritt Maduke
- Department of Molecular and Cellular Physiology, Stanford University, 279 Campus Drive West, B155 Beckman Center, Stanford, CA 94305
| |
Collapse
|
17
|
Chakrabarti S, Klich JD, Khallaf MA, Hulme AJ, Sánchez-Carranza O, Baran ZM, Rossi A, Huang ATL, Pohl T, Fleischer R, Fürst C, Hammes A, Bégay V, Hörnberg H, Finol-Urdaneta RK, Poole K, Dottori M, Lewin GR. Touch sensation requires the mechanically gated ion channel ELKIN1. Science 2024; 383:992-998. [PMID: 38422143 DOI: 10.1126/science.adl0495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/26/2024] [Indexed: 03/02/2024]
Abstract
Touch perception is enabled by mechanically activated ion channels, the opening of which excites cutaneous sensory endings to initiate sensation. In this study, we identify ELKIN1 as an ion channel likely gated by mechanical force, necessary for normal touch sensitivity in mice. Touch insensitivity in Elkin1-/- mice was caused by a loss of mechanically activated currents (MA currents) in around half of all sensory neurons activated by light touch (low-threshold mechanoreceptors). Reintroduction of Elkin1 into sensory neurons from Elkin1-/- mice restored MA currents. Additionally, small interfering RNA-mediated knockdown of ELKIN1 from induced human sensory neurons substantially reduced indentation-induced MA currents, supporting a conserved role for ELKIN1 in human touch. Our data identify ELKIN1 as a core component of touch transduction in mice and potentially in humans.
Collapse
Affiliation(s)
- Sampurna Chakrabarti
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Jasmin D Klich
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Mohammed A Khallaf
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
- Department of Zoology and Entomology, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Amy J Hulme
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Oscar Sánchez-Carranza
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Zuzanna M Baran
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
- Molecular and Cellular Basis of Behavior, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Alice Rossi
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Angela Tzu-Lun Huang
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Tobias Pohl
- Molecular and Cellular Basis of Behavior, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Raluca Fleischer
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Carina Fürst
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
- Molecular Pathways in Cortical Development, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Annette Hammes
- Molecular Pathways in Cortical Development, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Valérie Bégay
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Hanna Hörnberg
- Molecular and Cellular Basis of Behavior, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
- NeuroCure Cluster of Excellence, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Rocio K Finol-Urdaneta
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Kate Poole
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Mirella Dottori
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Gary R Lewin
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
- Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- German Center for Mental Health (DZPG), partner site Berlin, 10117 Berlin, Germany
| |
Collapse
|
18
|
Xu R, Treeby BE, Martin E. Safety Review of Therapeutic Ultrasound for Spinal Cord Neuromodulation and Blood-Spinal Cord Barrier Opening. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:317-331. [PMID: 38182491 DOI: 10.1016/j.ultrasmedbio.2023.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 01/07/2024]
Abstract
New focused ultrasound spinal cord applications have emerged, particularly those improving therapeutic agent delivery to the spinal cord via blood-spinal cord barrier opening and the neuromodulation of spinal cord tracts. One hurdle in the development of these applications is safety. It may be possible to use safety trends from seminal and subsequent works in focused ultrasound to guide the development of safety guidelines for spinal cord applications. We collated data from decades of pre-clinical studies and illustrate a clear relationship between damage, time-averaged spatial peak intensity and exposure duration. This relationship suggests a thermal mechanism underlies ultrasound-induced spinal cord damage. We developed minimum and mean thresholds for damage from these pre-clinical studies. When these thresholds were plotted against the parameters used in recent pre-clinical ultrasonic spinal cord neuromodulation studies, the majority of the neuromodulation studies were near or above the minimum threshold. This suggests that a thermal neuromodulatory effect may exist for ultrasonic spinal cord neuromodulation, and that the thermal dose must be carefully controlled to avoid damage to the spinal cord. By contrast, the intensity-exposure duration threshold had no predictive value when applied to blood-spinal cord barrier opening studies that employed injected contrast agents. Most blood-spinal cord barrier opening studies observed slight to severe damage, except for small animal studies that employed an active feedback control method to limit pressures based on measured bubble oscillation behavior. The development of new focused ultrasound spinal cord applications perhaps reflects the recent success in the development of focused ultrasound brain applications, and recent work has begun on the translation of these technologies from brain to spinal cord. However, a great deal of work remains to be done, particularly with respect to developing and accepting safety standards for these applications.
Collapse
Affiliation(s)
- Rui Xu
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK.
| | - Bradley E Treeby
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Eleanor Martin
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| |
Collapse
|
19
|
Coste B, Delmas P. PIEZO Ion Channels in Cardiovascular Functions and Diseases. Circ Res 2024; 134:572-591. [PMID: 38422173 DOI: 10.1161/circresaha.123.322798] [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] [Indexed: 03/02/2024]
Abstract
The cardiovascular system provides blood supply throughout the body and as such is perpetually applying mechanical forces to cells and tissues. Thus, this system is primed with mechanosensory structures that respond and adapt to changes in mechanical stimuli. Since their discovery in 2010, PIEZO ion channels have dominated the field of mechanobiology. These have been proposed as the long-sought-after mechanosensitive excitatory channels involved in touch and proprioception in mammals. However, more and more pieces of evidence point to the importance of PIEZO channels in cardiovascular activities and disease development. PIEZO channel-related cardiac functions include transducing hemodynamic forces in endothelial and vascular cells, red blood cell homeostasis, platelet aggregation, and arterial blood pressure regulation, among others. PIEZO channels contribute to pathological conditions including cardiac hypertrophy and pulmonary hypertension and congenital syndromes such as generalized lymphatic dysplasia and xerocytosis. In this review, we highlight recent advances in understanding the role of PIEZO channels in cardiovascular functions and diseases. Achievements in this quickly expanding field should open a new road for efficient control of PIEZO-related diseases in cardiovascular functions.
Collapse
Affiliation(s)
- Bertrand Coste
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| | - Patrick Delmas
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| |
Collapse
|
20
|
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
|
21
|
Wang G, Zhou Y, Yu C, Yang Q, Chen L, Ling S, Chen P, Xing J, Wu H, Zhao Q. Intravital photoacoustic brain stimulation with high-precision. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:S11520. [PMID: 38333219 PMCID: PMC10851606 DOI: 10.1117/1.jbo.29.s1.s11520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 02/10/2024]
Abstract
Significance Neural regulation at high precision vitally contributes to propelling fundamental understanding in the field of neuroscience and providing innovative clinical treatment options. Recently, photoacoustic brain stimulation has emerged as a cutting-edge method for precise neuromodulation and shows great potential for clinical application. Aim The goal of this perspective is to outline the advancements in photoacoustic brain stimulation in recent years. And, we also provide an outlook delineating several prospective paths through which this burgeoning approach may be substantively refined for augmented capability and wider implementations. Approach First, the mechanisms of photoacoustic generation as well as the potential mechanisms of photoacoustic brain stimulation are provided and discussed. Then, the state-of-the-art achievements corresponding to this technology are reviewed. Finally, future directions for photoacoustic technology in neuromodulation are provided. Results Intensive research endeavors have prompted substantial advancements in photoacoustic brain stimulation, illuminating the unique advantages of this modality for noninvasive and high-precision neuromodulation via a nongenetic way. It is envisaged that further technology optimization and randomized prospective clinical trials will enable a wide acceptance of photoacoustic brain stimulation in clinical practice. Conclusions The innovative practice of photoacoustic technology serves as a multifaceted neuromodulation approach, possessing noninvasive, high-accuracy, and nongenetic characteristics. It has a great potential that could considerably enhance not only the fundamental underpinnings of neuroscience research but also its practical implementations in a clinical setting.
Collapse
Affiliation(s)
- Guangxing Wang
- Xiamen University, School of Public Health, Center for Molecular Imaging and Translational Medicine, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen, China
- Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen, China
| | - Yuying Zhou
- Xiamen University, School of Public Health, Center for Molecular Imaging and Translational Medicine, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen, China
- Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen, China
| | - Chunhui Yu
- Xiamen University, School of Public Health, Center for Molecular Imaging and Translational Medicine, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen, China
- Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen, China
| | - Qiong Yang
- Xiamen University, School of Public Health, Center for Molecular Imaging and Translational Medicine, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen, China
- Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen, China
| | - Lin Chen
- Xiamen University, School of Public Health, Center for Molecular Imaging and Translational Medicine, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen, China
- Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen, China
| | - Shuting Ling
- Xiamen University, School of Public Health, Center for Molecular Imaging and Translational Medicine, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen, China
- Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen, China
| | - Pengyu Chen
- Xiamen University, School of Public Health, Center for Molecular Imaging and Translational Medicine, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen, China
- Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen, China
| | - Jiwei Xing
- Xiamen University, School of Public Health, Center for Molecular Imaging and Translational Medicine, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen, China
- Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen, China
| | - Huiling Wu
- Xiamen University, School of Public Health, Center for Molecular Imaging and Translational Medicine, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen, China
- Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen, China
| | - Qingliang Zhao
- Xiamen University, School of Public Health, Center for Molecular Imaging and Translational Medicine, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen, China
- Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, China
| |
Collapse
|
22
|
Grewal S, Gonçalves de Andrade E, Kofoed RH, Matthews PM, Aubert I, Tremblay MÈ, Morse SV. Using focused ultrasound to modulate microglial structure and function. Front Cell Neurosci 2023; 17:1290628. [PMID: 38164436 PMCID: PMC10757935 DOI: 10.3389/fncel.2023.1290628] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/31/2023] [Indexed: 01/03/2024] Open
Abstract
Transcranial focused ultrasound (FUS) has the unique ability to target regions of the brain with high spatial precision, in a minimally invasive manner. Neuromodulation studies have shown that FUS can excite or inhibit neuronal activity, demonstrating its tremendous potential to improve the outcome of neurological diseases. Recent evidence has also shed light on the emerging promise that FUS has, with and without the use of intravenously injected microbubbles, in modulating the blood-brain barrier and the immune cells of the brain. As the resident immune cells of the central nervous system, microglia are at the forefront of the brain's maintenance and immune defense. Notably, microglia are highly dynamic and continuously survey the brain parenchyma by extending and retracting their processes. This surveillance activity aids microglia in performing key physiological functions required for brain activity and plasticity. In response to stressors, microglia rapidly alter their cellular and molecular profile to help facilitate a return to homeostasis. While the underlying mechanisms by which both FUS and FUS + microbubbles modify microglial structure and function remain largely unknown, several studies in adult mice have reported changes in the expression of the microglia/macrophage marker ionized calcium binding adaptor molecule 1, and in their phagocytosis, notably of protein aggregates, such as amyloid beta. In this review, we discuss the demonstrated and putative biological effects of FUS and FUS + microbubbles in modulating microglial activities, with an emphasis on the key cellular and molecular changes observed in vitro and in vivo across models of brain health and disease. Understanding how this innovative technology can modulate microglia paves the way for future therapeutic strategies aimed to promote beneficial physiological microglial roles, and prevent or treat maladaptive responses.
Collapse
Affiliation(s)
- Sarina Grewal
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Elisa Gonçalves de Andrade
- Neuroscience Graduate Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Rikke Hahn Kofoed
- Department of Neurosurgery, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Center for Experimental Neuroscience-CENSE, Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Paul M. Matthews
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Isabelle Aubert
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Sophie V. Morse
- Department of Bioengineering, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
23
|
Cornelssen C, Finlinson E, Rolston JD, Wilcox KS. Ultrasonic therapies for seizures and drug-resistant epilepsy. Front Neurol 2023; 14:1301956. [PMID: 38162441 PMCID: PMC10756913 DOI: 10.3389/fneur.2023.1301956] [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: 09/25/2023] [Accepted: 11/09/2023] [Indexed: 01/03/2024] Open
Abstract
Ultrasonic therapy is an increasingly promising approach for the treatment of seizures and drug-resistant epilepsy (DRE). Therapeutic focused ultrasound (FUS) uses thermal or nonthermal energy to either ablate neural tissue or modulate neural activity through high- or low-intensity FUS (HIFU, LIFU), respectively. Both HIFU and LIFU approaches have been investigated for reducing seizure activity in DRE, and additional FUS applications include disrupting the blood-brain barrier in the presence of microbubbles for targeted-drug delivery to the seizure foci. Here, we review the preclinical and clinical studies that have used FUS to treat seizures. Additionally, we review effective FUS parameters and consider limitations and future directions of FUS with respect to the treatment of DRE. While detailed studies to optimize FUS applications are ongoing, FUS has established itself as a potential noninvasive alternative for the treatment of DRE and other neurological disorders.
Collapse
Affiliation(s)
- Carena Cornelssen
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States
| | - Eli Finlinson
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States
| | - John D. Rolston
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Karen S. Wilcox
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
24
|
Ashe J, Graf J, Madhavan R, Wallace K, Cotero V, Abate S, Pandey RK, Herzog R, Porindla SN, Shoudy D, Fan Y, Kao TJ, Puleo C. Investigation of liver-targeted peripheral focused ultrasound stimulation (pFUS) and its effect on glucose homeostasis and insulin resistance in type 2 diabetes mellitus: a proof of concept, phase 1 trial. QJM 2023; 116:667-685. [PMID: 37243693 DOI: 10.1093/qjmed/hcad098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 04/26/2023] [Indexed: 05/29/2023] Open
Abstract
BACKGROUND Mechanical waves produced by ultrasound pulses have been shown to activate mechanosensitive ion channels and modulate peripheral nerves. However, while peripheral ultrasound neuromodulation has been demonstrated in vitro and in pre-clinical models, there have been few reports of clinical tests. AIM We modified a diagnostic imaging system for ultrasound neuromodulation in human subjects. We report the first safety and feasibility outcomes in subjects with type 2 diabetes (T2D) mellitus and discuss these outcomes in relation to previous pre-clinical results. DESIGN The study was performed as an open label feasibility study to assess the effects of hepatic ultrasound (targeted to the porta hepatis) on glucometabolic parameters in subjects with T2D. Stimulation (peripheral focused ultrasound stimulation treatment) was performed for 3 days (i.e. 15 min per day), preceded by a baseline examination and followed by a 2-week observation period. METHODS Multiple metabolic assays were employed including measures of fasting glucose and insulin, insulin resistance and glucose metabolism. The safety and tolerability were also assessed by monitoring adverse events, changes in vital signs, electrocardiogram parameters and clinical laboratory measures. RESULTS AND CONCLUSION We report post-pFUS trends in several outcomes that were consistent with previous pre-clinical findings. Fasting insulin was lowered, resulting in a reduction of HOMA-IR scores (P-value 0.01; corrected Wilcoxon signed-rank test). Additional safety and exploratory markers demonstrated no device-related adverse impact of pFUS. Our findings demonstrate that pFUS represents a promising new treatment modality that could be used as a non-pharmaceutical adjunct or even alternative to current drug treatments in diabetes.
Collapse
Affiliation(s)
- J Ashe
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - J Graf
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - R Madhavan
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - K Wallace
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - V Cotero
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - S Abate
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - R K Pandey
- General Electric (GE) Research, Bengaluru, India
| | - R Herzog
- Yale Endocrinology & Metabolism, Yale School of Medicine, New Haven, CT, USA
| | - S N Porindla
- General Electric (GE) Research, Bengaluru, India
| | - D Shoudy
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Y Fan
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - T-J Kao
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - C Puleo
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| |
Collapse
|
25
|
Zheng H, Niu L, Qiu W, Liang D, Long X, Li G, Liu Z, Meng L. The Emergence of Functional Ultrasound for Noninvasive Brain-Computer Interface. RESEARCH (WASHINGTON, D.C.) 2023; 6:0200. [PMID: 37588619 PMCID: PMC10427153 DOI: 10.34133/research.0200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 07/04/2023] [Indexed: 08/18/2023]
Abstract
A noninvasive brain-computer interface is a central task in the comprehensive analysis and understanding of the brain and is an important challenge in international brain-science research. Current implanted brain-computer interfaces are cranial and invasive, which considerably limits their applications. The development of new noninvasive reading and writing technologies will advance substantial innovations and breakthroughs in the field of brain-computer interfaces. Here, we review the theory and development of the ultrasound brain functional imaging and its applications. Furthermore, we introduce latest advancements in ultrasound brain modulation and its applications in rodents, primates, and human; its mechanism and closed-loop ultrasound neuromodulation based on electroencephalograph are also presented. Finally, high-frequency acoustic noninvasive brain-computer interface is prospected based on ultrasound super-resolution imaging and acoustic tweezers.
Collapse
Affiliation(s)
- Hairong Zheng
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology,
Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lili Niu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology,
Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Weibao Qiu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology,
Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Dong Liang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology,
Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiaojing Long
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology,
Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Guanglin Li
- Shenzhen Institute of Advanced Integration Technology, Chinese Academy of Sciences and The Chinese University of Hong Kong, Shenzhen, 518055, China
| | - Zhiyuan Liu
- Shenzhen Institute of Advanced Integration Technology, Chinese Academy of Sciences and The Chinese University of Hong Kong, Shenzhen, 518055, China
| | - Long Meng
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology,
Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
26
|
Mandeville R, Sanchez B, Johnston B, Bazarek S, Thum JA, Birmingham A, See RHB, Leochico CFD, Kumar V, Dowlatshahi AS, Brown J, Stashuk D, Rutkove SB. A scoping review of current and emerging techniques for evaluation of peripheral nerve health, degeneration, and regeneration: part 1, neurophysiology. J Neural Eng 2023; 20:041001. [PMID: 37279730 DOI: 10.1088/1741-2552/acdbeb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/06/2023] [Indexed: 06/08/2023]
Abstract
Peripheral neuroregeneration research and therapeutic options are expanding exponentially. With this expansion comes an increasing need to reliably evaluate and quantify nerve health. Valid and responsive measures that can serve as biomarkers of the nerve status are essential for both clinical and research purposes for diagnosis, longitudinal follow-up, and monitoring the impact of any intervention. Furthermore, such biomarkers can elucidate regeneration mechanisms and open new avenues for research. Without these measures, clinical decision-making falls short, and research becomes more costly, time-consuming, and sometimes infeasible. As a companion to Part 2, which is focused on non-invasive imaging, Part 1 of this two-part scoping review systematically identifies and critically examines many current and emerging neurophysiological techniques that have the potential to evaluate peripheral nerve health, particularly from the perspective of regenerative therapies and research.
Collapse
Affiliation(s)
- Ross Mandeville
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215, United States of America
| | - Benjamin Sanchez
- Department Electrical and Computer Engineering, University of Utah, Salt Lake City, UT 84112, United States of America
| | - Benjamin Johnston
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA 02115, United States of America
| | - Stanley Bazarek
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA 02115, United States of America
| | - Jasmine A Thum
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Austin Birmingham
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Reiner Henson B See
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Carl Froilan D Leochico
- Department of Physical Medicine and Rehabilitation, St. Luke's Medical Center, Global City, Taguig, The Philippines
- Department of Rehabilitation Medicine, Philippine General Hospital, University of the Philippines Manila, Manila, The Philippines
| | - Viksit Kumar
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Arriyan S Dowlatshahi
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215, United States of America
| | - Justin Brown
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Daniel Stashuk
- Department of Systems Design Engineering, University of Waterloo, Ontario N2L 3G1, Canada
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215, United States of America
| |
Collapse
|
27
|
Yang Y, Yuan J, Field RL, Ye D, Hu Z, Xu K, Xu L, Gong Y, Yue Y, Kravitz AV, Bruchas MR, Cui J, Brestoff JR, Chen H. Induction of a torpor-like hypothermic and hypometabolic state in rodents by ultrasound. Nat Metab 2023; 5:789-803. [PMID: 37231250 PMCID: PMC10229429 DOI: 10.1038/s42255-023-00804-z,] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 04/11/2023] [Indexed: 08/22/2023]
Abstract
Torpor is an energy-conserving state in which animals dramatically decrease their metabolic rate and body temperature to survive harsh environmental conditions. Here, we report the noninvasive, precise and safe induction of a torpor-like hypothermic and hypometabolic state in rodents by remote transcranial ultrasound stimulation at the hypothalamus preoptic area (POA). We achieve a long-lasting (>24 h) torpor-like state in mice via closed-loop feedback control of ultrasound stimulation with automated detection of body temperature. Ultrasound-induced hypothermia and hypometabolism (UIH) is triggered by activation of POA neurons, involves the dorsomedial hypothalamus as a downstream brain region and subsequent inhibition of thermogenic brown adipose tissue. Single-nucleus RNA-sequencing of POA neurons reveals TRPM2 as an ultrasound-sensitive ion channel, the knockdown of which suppresses UIH. We also demonstrate that UIH is feasible in a non-torpid animal, the rat. Our findings establish UIH as a promising technology for the noninvasive and safe induction of a torpor-like state.
Collapse
Affiliation(s)
- Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Jinyun Yuan
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Rachael L Field
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Zhongtao Hu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Kevin Xu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Lu Xu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Yan Gong
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Yimei Yue
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Alexxai V Kravitz
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael R Bruchas
- Departments of Anesthesiology and Pain Medicine, Pharmacology, and Bioengineering, Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Jianmin Cui
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Jonathan R Brestoff
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA.
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA.
- Division of Neurotechnology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
28
|
Yang Y, Yuan J, Field RL, Ye D, Hu Z, Xu K, Xu L, Gong Y, Yue Y, Kravitz AV, Bruchas MR, Cui J, Brestoff JR, Chen H. Induction of a torpor-like hypothermic and hypometabolic state in rodents by ultrasound. Nat Metab 2023; 5:789-803. [PMID: 37231250 PMCID: PMC10229429 DOI: 10.1038/s42255-023-00804-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 04/11/2023] [Indexed: 05/27/2023]
Abstract
Torpor is an energy-conserving state in which animals dramatically decrease their metabolic rate and body temperature to survive harsh environmental conditions. Here, we report the noninvasive, precise and safe induction of a torpor-like hypothermic and hypometabolic state in rodents by remote transcranial ultrasound stimulation at the hypothalamus preoptic area (POA). We achieve a long-lasting (>24 h) torpor-like state in mice via closed-loop feedback control of ultrasound stimulation with automated detection of body temperature. Ultrasound-induced hypothermia and hypometabolism (UIH) is triggered by activation of POA neurons, involves the dorsomedial hypothalamus as a downstream brain region and subsequent inhibition of thermogenic brown adipose tissue. Single-nucleus RNA-sequencing of POA neurons reveals TRPM2 as an ultrasound-sensitive ion channel, the knockdown of which suppresses UIH. We also demonstrate that UIH is feasible in a non-torpid animal, the rat. Our findings establish UIH as a promising technology for the noninvasive and safe induction of a torpor-like state.
Collapse
Affiliation(s)
- Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Jinyun Yuan
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Rachael L Field
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Zhongtao Hu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Kevin Xu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Lu Xu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Yan Gong
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Yimei Yue
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Alexxai V Kravitz
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael R Bruchas
- Departments of Anesthesiology and Pain Medicine, Pharmacology, and Bioengineering, Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Jianmin Cui
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Jonathan R Brestoff
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA.
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA.
- Division of Neurotechnology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
29
|
Liu T, Fu Y, Shi J, He S, Chen D, Li W, Chen Y, Zhang L, Lv Q, Yang Y, Jin Q, Wang J, Xie M. Noninvasive ultrasound stimulation to treat myocarditis through splenic neuro-immune regulation. J Neuroinflammation 2023; 20:94. [PMID: 37069636 PMCID: PMC10108488 DOI: 10.1186/s12974-023-02773-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 04/05/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND The cholinergic anti-inflammatory pathway (CAP) has been widely studied to modulate the immune response. Current stimulating strategies are invasive or imprecise. Noninvasive low-intensity pulsed ultrasound (LIPUS) has become increasingly appreciated for targeted neuronal modulation. However, its mechanisms and physiological role on myocarditis remain poorly defined. METHODS The mouse model of experimental autoimmune myocarditis was established. Low-intensity pulsed ultrasound was targeted at the spleen to stimulate the spleen nerve. Under different ultrasound parameters, histological tests and molecular biology were performed to observe inflammatory lesions and changes in immune cell subsets in the spleen and heart. In addition, we evaluated the dependence of the spleen nerve and cholinergic anti-inflammatory pathway of low-intensity pulsed ultrasound in treating autoimmune myocarditis in mice through different control groups. RESULTS The echocardiography and flow cytometry of splenic or heart infiltrating immune cells revealed that splenic ultrasound could alleviate the immune response, regulate the proportion and function of CD4+ Treg and macrophages by activating cholinergic anti-inflammatory pathway, and finally reduce heart inflammatory injury and improve cardiac remodeling, which is as effective as an acetylcholine receptor agonists GTS-21. Transcriptome sequencing showed significant differential expressed genes due to ultrasound modulation. CONCLUSIONS It is worth noting that the ultrasound therapeutic efficacy depends greatly on acoustic pressure and exposure duration, and the effective targeting organ was the spleen but not the heart. This study provides novel insight into the therapeutic potentials of LIPUS, which are essential for its future application.
Collapse
Affiliation(s)
- Tianshu Liu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yanan Fu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jiawei Shi
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Shukun He
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Dandan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Wenqu Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yali Yang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Jing Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
30
|
Huerta TS, Haider B, Adamovich-Zeitlin R, Chen AC, Chaudhry S, Zanos TP, Chavan SS, Tracey KJ, Chang EH. Calcium imaging and analysis of the jugular-nodose ganglia enables identification of distinct vagal sensory neuron subsets. J Neural Eng 2023; 20:10.1088/1741-2552/acbe1e. [PMID: 36920156 PMCID: PMC10790314 DOI: 10.1088/1741-2552/acbe1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 02/22/2023] [Indexed: 03/16/2023]
Abstract
Objective.Sensory nerves of the peripheral nervous system (PNS) transmit afferent signals from the body to the brain. These peripheral nerves are composed of distinct subsets of fibers and associated cell bodies, which reside in peripheral ganglia distributed throughout the viscera and along the spinal cord. The vagus nerve (cranial nerve X) is a complex polymodal nerve that transmits a wide array of sensory information, including signals related to mechanical, chemical, and noxious stimuli. To understand how stimuli applied to the vagus nerve are encoded by vagal sensory neurons in the jugular-nodose ganglia, we developed a framework for micro-endoscopic calcium imaging and analysis.Approach.We developed novel methods forin vivoimaging of the intact jugular-nodose ganglion using a miniature microscope (Miniscope) in transgenic mice with the genetically-encoded calcium indicator GCaMP6f. We adapted the Python-based analysis package Calcium Imaging Analysis (CaImAn) to process the resulting one-photon fluorescence data into calcium transients for subsequent analysis. Random forest classification was then used to identify specific types of neuronal responders.Results.We demonstrate that recordings from the jugular-nodose ganglia can be accomplished through careful surgical dissection and ganglia stabilization. Using a customized acquisition and analysis pipeline, we show that subsets of vagal sensory neurons respond to different chemical stimuli applied to the vagus nerve. Successful classification of the responses with a random forest model indicates that certain calcium transient features, such as amplitude and duration, are important for encoding these stimuli by sensory neurons.Significance.This experimental approach presents a new framework for investigating how individual vagal sensory neurons encode various stimuli on the vagus nerve. Our surgical and analytical approach can be applied to other PNS ganglia in rodents and other small animal species to elucidate previously unexplored roles for peripheral neurons in a diverse set of physiological functions.
Collapse
Affiliation(s)
- Tomás S Huerta
- Laboratory for Biomedical Sciences, Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States of America
| | - Bilal Haider
- Laboratory for Biomedical Sciences, Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
| | - Richard Adamovich-Zeitlin
- Laboratory for Biomedical Sciences, Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States of America
| | - Adrian C Chen
- Laboratory for Biomedical Sciences, Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States of America
| | - Saher Chaudhry
- Laboratory for Biomedical Sciences, Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
| | - Theodoros P Zanos
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States of America
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States of America
| | - Sangeeta S Chavan
- Laboratory for Biomedical Sciences, Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States of America
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States of America
| | - Kevin J Tracey
- Laboratory for Biomedical Sciences, Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States of America
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States of America
| | - Eric H Chang
- Laboratory for Biomedical Sciences, Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States of America
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States of America
| |
Collapse
|
31
|
Young MN, Sindoni MJ, Lewis AH, Zauscher S, Grandl J. The energetics of rapid cellular mechanotransduction. Proc Natl Acad Sci U S A 2023; 120:e2215747120. [PMID: 36795747 PMCID: PMC9974467 DOI: 10.1073/pnas.2215747120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/18/2023] [Indexed: 02/17/2023] Open
Abstract
Cells throughout the human body detect mechanical forces. While it is known that the rapid (millisecond) detection of mechanical forces is mediated by force-gated ion channels, a detailed quantitative understanding of cells as sensors of mechanical energy is still lacking. Here, we combine atomic force microscopy with patch-clamp electrophysiology to determine the physical limits of cells expressing the force-gated ion channels (FGICs) Piezo1, Piezo2, TREK1, and TRAAK. We find that, depending on the ion channel expressed, cells can function either as proportional or nonlinear transducers of mechanical energy and detect mechanical energies as little as ~100 fJ, with a resolution of up to ~1 fJ. These specific energetic values depend on cell size, channel density, and cytoskeletal architecture. We also make the surprising discovery that cells can transduce forces either nearly instantaneously (<1 ms) or with a substantial time delay (~10 ms). Using a chimeric experimental approach and simulations, we show how such delays can emerge from channel-intrinsic properties and the slow diffusion of tension in the membrane. Overall, our experiments reveal the capabilities and limits of cellular mechanosensing and provide insights into molecular mechanisms that different cell types may employ to specialize for their distinct physiological roles.
Collapse
Affiliation(s)
- Michael N. Young
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
| | - Michael J. Sindoni
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
| | - Amanda H. Lewis
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
| | - Stefan Zauscher
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC27710
| | - Jörg Grandl
- Department of Neurobiology, Duke University Medical Center, Durham, NC27710
| |
Collapse
|
32
|
Sorum B, Docter T, Panico V, Rietmeijer RA, Brohawn SG. Pressure and ultrasound activate mechanosensitive TRAAK K + channels through increased membrane tension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523644. [PMID: 36712118 PMCID: PMC9882092 DOI: 10.1101/2023.01.11.523644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
TRAAK is a mechanosensitive two-pore domain K + (K2P) channel found in nodes of Ranvier within myelinated axons. It displays low leak activity at rest and is activated up to one hundred-fold by increased membrane tension. Structural and functional studies have led to physical models for channel gating and mechanosensitivity, but no quantitative analysis of channel activation by tension has been reported. Here, we use simultaneous patch-clamp recording and fluorescent imaging to determine the tension response characteristics of TRAAK. TRAAK shows high sensitivity and a broad response to tension spanning nearly the entire physiologically relevant tension range. This graded response profile distinguishes TRAAK from similarly low-threshold mechanosensitive channels Piezo1 and MscS, which activate in a step-like fashion over a narrow tension range. We further use patch imaging to show that ultrasonic activation of TRAAK and MscS is due to increased membrane tension. Together, these results provide mechanistic insight into TRAAK tension gating, a framework for exploring the role of mechanosensitive K + channels at nodes of Ranvier, and biophysical context for developing ultrasound as a mechanical stimulation technique for neuromodulation.
Collapse
Affiliation(s)
- Ben Sorum
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, California 94720, USA.,Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California 94720, USA.,California Institute for Quantitative Biology (QB3), University of California, Berkeley, CA 94720, USA
| | - Trevor Docter
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, California 94720, USA.,Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California 94720, USA.,California Institute for Quantitative Biology (QB3), University of California, Berkeley, CA 94720, USA
| | - Vincent Panico
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, California 94720, USA.,Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California 94720, USA.,California Institute for Quantitative Biology (QB3), University of California, Berkeley, CA 94720, USA
| | - Robert A. Rietmeijer
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, California 94720, USA.,Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California 94720, USA.,California Institute for Quantitative Biology (QB3), University of California, Berkeley, CA 94720, USA
| | - Stephen G. Brohawn
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, California 94720, USA.,Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California 94720, USA.,California Institute for Quantitative Biology (QB3), University of California, Berkeley, CA 94720, USA.,Correspondence:
| |
Collapse
|
33
|
Bao SC, Li F, Xiao Y, Niu L, Zheng H. Peripheral focused ultrasound stimulation and its applications: From therapeutics to human-computer interaction. Front Neurosci 2023; 17:1115946. [PMID: 37123351 PMCID: PMC10140332 DOI: 10.3389/fnins.2023.1115946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
Peripheral focused ultrasound stimulation (pFUS) has gained increasing attention in the past few decades, because it can be delivered to peripheral nerves, neural endings, or sub-organs. With different stimulation parameters, ultrasound stimulation could induce different modulation effects. Depending on the transmission medium, pFUS can be classified as body-coupled US stimulation, commonly used for therapeutics or neuromodulation, or as an air-coupled contactless US haptic system, which provides sensory inputs and allows distinct human-computer interaction paradigms. Despite growing interest in pFUS, the underlying working mechanisms remain only partially understood, and many applications are still in their infancy. This review focused on existing applications, working mechanisms, the latest progress, and future directions of pFUS. In terms of therapeutics, large-sample randomized clinical trials in humans are needed to translate these state of art techniques into treatments for specific diseases. The airborne US for human-computer interaction is still in its preliminary stage, but further efforts in task-oriented US applications might provide a promising interaction tool soon.
Collapse
Affiliation(s)
- Shi-Chun Bao
- National Innovation Center for Advanced Medical Devices, Shenzhen, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fei Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yang Xiao
- National Innovation Center for Advanced Medical Devices, Shenzhen, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lili Niu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- *Correspondence: Hairong Zheng,
| |
Collapse
|
34
|
Ojeda-Alonso J, Bégay V, Garcia-Contreras JA, Campos-Pérez AF, Purfürst B, Lewin GR. Lack of evidence for participation of TMEM150C in sensory mechanotransduction. J Gen Physiol 2022; 154:e202213098. [PMID: 36256908 PMCID: PMC9582506 DOI: 10.1085/jgp.202213098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022] Open
Abstract
The membrane protein TMEM150C has been proposed to form a mechanosensitive ion channel that is required for normal proprioceptor function. Here, we examined whether expression of TMEM150C in neuroblastoma cells lacking Piezo1 is associated with the appearance of mechanosensitive currents. Using three different modes of mechanical stimuli, indentation, membrane stretch, and substrate deflection, we could not evoke mechanosensitive currents in cells expressing TMEM150C. We next asked if TMEM150C is necessary for the normal mechanosensitivity of cutaneous sensory neurons. We used an available mouse model in which the Tmem150c locus was disrupted through the insertion of a LacZ cassette with a splice acceptor that should lead to transcript truncation. Analysis of these mice indicated that ablation of the Tmem150c gene was not complete in sensory neurons of the dorsal root ganglia (DRG). Using a CRISPR/Cas9 strategy, we made a second mouse model in which a large part of the Tmem150c gene was deleted and established that these Tmem150c-/- mice completely lack TMEM150C protein in the DRGs. We used an ex vivo skin nerve preparation to characterize the mechanosenstivity of mechanoreceptors and nociceptors in the glabrous skin of the Tmem150c-/- mice. We found no quantitative alterations in the physiological properties of any type of cutaneous sensory fiber in Tmem150c-/- mice. Since it has been claimed that TMEM150C is required for normal proprioceptor function, we made a quantitative analysis of locomotion in Tmem150c-/- mice. Here again, we found no indication that there was altered gait in Tmem150c-/- mice compared to wild-type controls. In summary, we conclude that existing mouse models that have been used to investigate TMEM150C function in vivo are problematic. Furthermore, we could find no evidence that TMEM150C forms a mechanosensitive channel or that it is necessary for the normal mechanosensitivity of cutaneous sensory neurons.
Collapse
Affiliation(s)
- Julia Ojeda-Alonso
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Valérie Bégay
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jonathan Alexis Garcia-Contreras
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Andrea Fernanda Campos-Pérez
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Bettina Purfürst
- Electron Microscopy Core Facility, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Gary R. Lewin
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
35
|
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
|