1
|
Azimzadeh JB, Quiñones PM, Oghalai JS, Ricci AJ. Infrared light stimulates the cochlea through a mechanical displacement detected and amplified by hair cells. Proc Natl Acad Sci U S A 2025; 122:e2422076122. [PMID: 40273108 DOI: 10.1073/pnas.2422076122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/01/2025] [Indexed: 04/26/2025] Open
Abstract
Although cochlear implants (CI) are the standard of care for profound sensorineural hearing loss they are technically constrained by the tendency of electrical current to spread within the fluid-filled chambers of the cochlea. This limits the resolution of individual electrodes and patients' perceptions of complex sounds. Infrared irradiation has been proposed as an alternative to electrical stimulation because it can elicit auditory responses while being spatially constrained, theoretically promising higher-fidelity hearing for the deaf. However, conflicting reports locate the site of infrared excitation at spiral ganglia neurons or hair cells. We use a combination of genetic, pharmacological, optical, and electrophysiological tools to determine the site of action of infrared irradiation. Infrared-evoked cochlear potentials are composed of two peaks: one driven by hair cells (the microphonic) and a second driven by spiral ganglion neurons (the neural response). Manipulations that prevented hair cell synaptic activity abolished the neural component, while manipulations blocking hair cell mechanotransduction abolished all responses, suggesting a mechanical component to the infrared response. Optical coherence tomography (OCT) confirmed that infrared irradiation creates a mechanical stimulus that is both amplified and detected by hair cells. Because infrared irradiation does not stimulate spiral ganglion neurons directly, it is unlikely to replace the electrical CI.
Collapse
Affiliation(s)
- Julien B Azimzadeh
- Department of Otolaryngology, Stanford University School of Medicine, Stanford CA 94304
| | - Patricia M Quiñones
- Department of Otolaryngology, University of Southern California, Los Angeles, CA 90033
| | - John S Oghalai
- Department of Otolaryngology, University of Southern California, Los Angeles, CA 90033
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90033
| | - Anthony J Ricci
- Department of Otolaryngology, Stanford University School of Medicine, Stanford CA 94304
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA d: 94304
| |
Collapse
|
2
|
Golovynska I, Golovynskyi S, Stepanov YV, Qu J, Zhang R, Qu J. Near-infrared light therapy normalizes amyloid load, neuronal lipid membrane order, rafts and cholesterol level in Alzheimer's disease. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 262:113086. [PMID: 39724841 DOI: 10.1016/j.jphotobiol.2024.113086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Cholesterol dysregulation, disorder of neuronal membrane lipid packing, and lipid rafts lead to the synthesis and accumulation of toxic amyloid-β (Aβ), contributing to the development of Alzheimer's disease (AD). Our study shows that near-infrared (NIR) transcranial photobiomodulation therapy (tPBMT) can reduce Aβ load and restore the properties of neuronal plasma membrane, including Aβ production, bilayer order, rafts, lipid content, and Ca2+ channels during AD. Mice in the experiments were exposed to 808-nm LED for 1 h daily over 3 months. In the APOE transgenic model with cholesterol dysregulation, the cholesterol levels increased by 22 times, causing healthy neurons to produce toxic Aβ three times faster, increasing its load by five times. Consequently, Aβ disrupts the membrane bilayer and prompts the formation of lipid rafts and pores. NIR-tPBMT can nearly half attenuate Aβ load, restore membrane lipid order and rigidity, reduce the number of lipid rafts, modulate cholesterol synthesis, normalize Ca2+ influx by activated endocytosis, and reduce neuronal death. The Ca2+ influx induced by light does not cause excitotoxicity but modulates Ca2+/calmodulin signaling involved in AD mechanisms and cell viability. The transcriptome analysis of the brain cortex and hippocampus shows that light can downregulate Ca2+/calmodulin-dependent AD-risk genes BACE, PSEN, and APP, and normalize cholesterol homeostasis via the HMGCR, DHCR7, and INSIG1 genes. Additionally, light enhances neuron resistance to the endoplasmic reticulum stress via activating transcription factors of the unfolded protein response. Thus, red/NIR light could be promising in combating AD, restoring synaptic plasticity in degenerating neurons and reducing Aβ load.
Collapse
Affiliation(s)
- Iuliia Golovynska
- Center for Biomedical Photonics, College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, PR China.
| | - Sergii Golovynskyi
- Center for Biomedical Photonics, College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, PR China
| | - Yurii V Stepanov
- Laboratory of Molecular and Cellular Mechanisms of Metastasis, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv 03022, Ukraine
| | - Jinghan Qu
- Center for Biomedical Photonics, College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, PR China
| | - Renlong Zhang
- Center for Biomedical Photonics, College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, PR China
| | - Junle Qu
- Center for Biomedical Photonics, College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, PR China.
| |
Collapse
|
3
|
Koskimäki S, Tojkander S. TRPV4-A Multifunctional Cellular Sensor Protein with Therapeutic Potential. SENSORS (BASEL, SWITZERLAND) 2024; 24:6923. [PMID: 39517820 PMCID: PMC11548305 DOI: 10.3390/s24216923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/02/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Abstract
Transient receptor potential vanilloid (TRPV) channel proteins belong to the superfamily of TRP proteins that form cationic channels in the animal cell membranes. These proteins have various subtype-specific functions, serving, for example, as sensors for pain, pressure, pH, and mechanical extracellular stimuli. The sensing of extracellular cues by TRPV4 triggers Ca2+-influx through the channel, subsequently coordinating numerous intracellular signaling cascades in a spatio-temporal manner. As TRPV channels play such a wide role in various cellular and physiological functions, loss or impaired TRPV protein activity naturally contributes to many pathophysiological processes. This review concentrates on the known functions of TRPV4 sensor proteins and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Sanna Koskimäki
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | | |
Collapse
|
4
|
Dumas N, Pecchi E, O'Connor R, Bos R, Moreau D. Infrared neuroglial modulation of spinal locomotor networks. Sci Rep 2024; 14:22282. [PMID: 39333287 PMCID: PMC11437012 DOI: 10.1038/s41598-024-73577-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
Infrared neural stimulation (INS) emerges as a promising tool for stimulating the nervous system by its high spatial precision and absence of the use of exogenous agents into the tissue, which led to the first successful proof of concept in human brain. While neural networks have been the focal point of INS research, this technique is also non cell type specific as it triggers activity in non electrically excitable cells. Despite increasing interest, there remains to demonstrate well defined simultaneous astrocytic and neuronal signals in response to INS. Using calcium imaging, we show that INS has the capacity to initiate calcium signaling in both astrocytes and neurons simultaneously from the rostral lumbar spinal cord, each exhibiting distinct temporal and amplitude characteristics. Importantly, the mechanism underlying infrared-induced neuronal and astrocytic calcium signaling differ, with neuronal activity relying on sodium channels, whereas induced astrocytic signaling is predominantly influenced by extracellular calcium and TRPV4 channels. Furthermore, our findings demonstrate the frequency shift of neuronal calcium oscillations through infrared stimulation. By deepening our understanding in INS fundamentals, this technique holds great promise for advancing neuroscience, deepening our understanding of pathologies, and potentially paving the way for future clinical applications.
Collapse
Affiliation(s)
- Nathan Dumas
- Mines Saint-Etienne, Centre CMP, Département BEL, 13541, Gardanne, France
| | - Emilie Pecchi
- Institut de Neurosciences de la Timone, CNRS UMR 7289 et Aix- Marseille Université, 13005, Marseille, France
| | - Rodney O'Connor
- Mines Saint-Etienne, Centre CMP, Département BEL, 13541, Gardanne, France
| | - Rémi Bos
- Institut de Neurosciences de la Timone, CNRS UMR 7289 et Aix- Marseille Université, 13005, Marseille, France
| | - David Moreau
- Mines Saint-Etienne, Centre CMP, Département BEL, 13541, Gardanne, France.
| |
Collapse
|
5
|
Conner GE. NADPH Alters DUOX1 Calcium Responsiveness. Redox Biol 2024; 75:103251. [PMID: 38936256 PMCID: PMC11259916 DOI: 10.1016/j.redox.2024.103251] [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: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024] Open
Abstract
Hydrogen peroxide is a key element in redox signaling and in setting cellular redox tone. DUOX1 and DUOX2, that directly synthesize hydrogen peroxide, are the most abundant NADPH oxidase transcripts in most epithelia. DUOX1 and DUOX2 hydrogen peroxide synthesis is regulated by intracellular calcium transients and thus cells can respond to signals and initiate responses by increasing cellular hydrogen peroxide synthesis. Nevertheless, many details of their enzymatic regulation are still unexplored. DUOX1 and DUOXA1 were expressed in HEK293T cells and activity was studied in homogenates and membrane fractions. When DUOX1 homogenates or membranes were pre-incubated in NADPH and started with addition of Ca2+, to mimic intracellular activation, progress curves were distinctly different from those pre-incubated in Ca2+ and started with NADPH. The Ca2+ EC50 for DUOX1's initial rate when pre-incubated in Ca2+, was three orders of magnitude lower (EC50 ∼ 10-6 M) than with preincubation in NADPH (EC50 ∼ 10-3 M). In addition, activity was several fold lower with Ca2+ start. Identical results were obtained using homogenates and membrane fractions. The data suggested that DUOX1 Ca2+ binding in expected physiological signaling conditions only slowly leads to maximal hydrogen peroxide synthesis and that full hydrogen peroxide synthesis activity in vivo only can occur when encountering extremely high concentration Ca2+ signals. Thus, a complex interplay of intracellular NADPH and Ca2+ concentrations regulate DUOX1 over a wide extent and may limit DUOX1 activity to a restricted range and spatial distribution.
Collapse
Affiliation(s)
- Gregory E Conner
- Department of Cell Biology, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, Miami Fl, 33136, USA.
| |
Collapse
|
6
|
Sander MY, Zhu X. Infrared neuromodulation-a review. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2024; 87:066701. [PMID: 38701769 DOI: 10.1088/1361-6633/ad4729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 05/03/2024] [Indexed: 05/05/2024]
Abstract
Infrared (IR) neuromodulation (INM) is an emerging light-based neuromodulation approach that can reversibly control neuronal and muscular activities through the transient and localized deposition of pulsed IR light without requiring any chemical or genetic pre-treatment of the target cells. Though the efficacy and short-term safety of INM have been widely demonstrated in both peripheral and central nervous systems, the investigations of the detailed cellular and biological processes and the underlying biophysical mechanisms are still ongoing. In this review, we discuss the current research progress in the INM field with a focus on the more recently discovered IR nerve inhibition. Major biophysical mechanisms associated with IR nerve stimulation are summarized. As the INM effects are primarily attributed to the spatiotemporal thermal transients induced by water and tissue absorption of pulsed IR light, temperature monitoring techniques and simulation models adopted in INM studies are discussed. Potential translational applications, current limitations, and challenges of the field are elucidated to provide guidance for future INM research and advancement.
Collapse
Affiliation(s)
- Michelle Y Sander
- Department of Electrical and Computer Engineering, Boston University, 8 Saint Mary's Street, Boston, MA 02215, United States of America
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, United States of America
- Division of Materials Science and Engineering, Boston University, 15 Saint Mary's Street, Brookline, MA 02446, United States of America
- Photonics Center, Boston University, 8 Saint Mary's Street, Boston, MA 02215, United States of America
- Neurophotonics Center, Boston University, 24 Cummington Mall, Boston, MA 02215, United States of America
| | - Xuedong Zhu
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, United States of America
- Photonics Center, Boston University, 8 Saint Mary's Street, Boston, MA 02215, United States of America
- Neurophotonics Center, Boston University, 24 Cummington Mall, Boston, MA 02215, United States of America
| |
Collapse
|
7
|
Sato H, Sugimoto F, Furukawa R, Tateno T. Modulatory Effects on Laminar Neural Activity Induced by Near-Infrared Light Stimulation with a Continuous Waveform to the Mouse Inferior Colliculus In Vivo. eNeuro 2024; 11:ENEURO.0521-23.2024. [PMID: 38627064 DOI: 10.1523/eneuro.0521-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 05/03/2024] Open
Abstract
Infrared neural stimulation (INS) is a promising area of interest for the clinical application of a neuromodulation method. This is in part because of its low invasiveness, whereby INS modulates the activity of the neural tissue mainly through temperature changes. Additionally, INS may provide localized brain stimulation with less tissue damage. The inferior colliculus (IC) is a crucial auditory relay nucleus and a potential target for clinical application of INS to treat auditory diseases and develop artificial hearing devices. Here, using continuous INS with low to high-power density, we demonstrate the laminar modulation of neural activity in the mouse IC in the presence and absence of sound. We investigated stimulation parameters of INS to effectively modulate the neural activity in a facilitatory or inhibitory manner. A mathematical model of INS-driven brain tissue was first simulated, temperature distributions were numerically estimated, and stimulus parameters were selected from the simulation results. Subsequently, INS was administered to the IC of anesthetized mice, and the modulation effect on the neural activity was measured using an electrophysiological approach. We found that the modulatory effect of INS on the spontaneous neural activity was bidirectional between facilitatory and inhibitory effects. The modulatory effect on sound-evoked responses produced only an inhibitory effect to all examined stimulus intensities. Thus, this study provides important physiological evidence on the response properties of IC neurons to INS. Overall, INS can be used for the development of new therapies for neurological disorders and functional support devices for auditory central processing.
Collapse
Affiliation(s)
- Hiromu Sato
- Bioengineering and Bioinformatics, Graduate School of Information Science and Technology, Hokkaido University, Sapporo 060-0814, Japan
| | - Futoshi Sugimoto
- Bioengineering and Bioinformatics, Graduate School of Information Science and Technology, Hokkaido University, Sapporo 060-0814, Japan
| | - Ryo Furukawa
- Bioengineering and Bioinformatics, Graduate School of Information Science and Technology, Hokkaido University, Sapporo 060-0814, Japan
| | - Takashi Tateno
- Division of Bioengineering and Bioinformatics, Faculty of Information Science and Technology, Hokkaido University, Sapporo 060-0814, Japan
| |
Collapse
|
8
|
Garrido-Peña A, Sanchez-Martin P, Reyes-Sanchez M, Levi R, Rodriguez FB, Castilla J, Tornero J, Varona P. Modulation of neuronal dynamics by sustained and activity-dependent continuous-wave near-infrared laser stimulation. NEUROPHOTONICS 2024; 11:024308. [PMID: 38764942 PMCID: PMC11100521 DOI: 10.1117/1.nph.11.2.024308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/21/2024]
Abstract
Significance Near-infrared laser illumination is a non-invasive alternative/complement to classical stimulation methods in neuroscience but the mechanisms underlying its action on neuronal dynamics remain unclear. Most studies deal with high-frequency pulsed protocols and stationary characterizations disregarding the dynamic modulatory effect of sustained and activity-dependent stimulation. The understanding of such modulation and its widespread dissemination can help to develop specific interventions for research applications and treatments for neural disorders. Aim We quantified the effect of continuous-wave near-infrared (CW-NIR) laser illumination on single neuron dynamics using sustained stimulation and an open-source activity-dependent protocol to identify the biophysical mechanisms underlying this modulation and its time course. Approach We characterized the effect by simultaneously performing long intracellular recordings of membrane potential while delivering sustained and closed-loop CW-NIR laser stimulation. We used waveform metrics and conductance-based models to assess the role of specific biophysical candidates on the modulation. Results We show that CW-NIR sustained illumination asymmetrically accelerates action potential dynamics and the spiking rate on single neurons, while closed-loop stimulation unveils its action at different phases of the neuron dynamics. Our model study points out the action of CW-NIR on specific ionic-channels and the key role of temperature on channel properties to explain the modulatory effect. Conclusions Both sustained and activity-dependent CW-NIR stimulation effectively modulate neuronal dynamics by a combination of biophysical mechanisms. Our open-source protocols can help to disseminate this non-invasive optical stimulation in novel research and clinical applications.
Collapse
Affiliation(s)
- Alicia Garrido-Peña
- Grupo de Neurocomputación Biológica, Departamento de Ingeniería Informática, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Madrid, Spain
| | - Pablo Sanchez-Martin
- Grupo de Neurocomputación Biológica, Departamento de Ingeniería Informática, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Reyes-Sanchez
- Grupo de Neurocomputación Biológica, Departamento de Ingeniería Informática, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rafael Levi
- Grupo de Neurocomputación Biológica, Departamento de Ingeniería Informática, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Madrid, Spain
| | - Francisco B. Rodriguez
- Grupo de Neurocomputación Biológica, Departamento de Ingeniería Informática, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Madrid, Spain
| | - Javier Castilla
- Hospital los Madroños, Center for Clinical Neuroscience, Brunete, Spain
| | - Jesus Tornero
- Hospital los Madroños, Center for Clinical Neuroscience, Brunete, Spain
| | - Pablo Varona
- Grupo de Neurocomputación Biológica, Departamento de Ingeniería Informática, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
9
|
Ma Q, Wu J, Li H, Ma X, Yin R, Bai L, Tang H, Liu N. The role of TRPV4 in programmed cell deaths. Mol Biol Rep 2024; 51:248. [PMID: 38300413 DOI: 10.1007/s11033-023-09199-2] [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: 10/01/2023] [Accepted: 12/30/2023] [Indexed: 02/02/2024]
Abstract
Programmed cell death is a major life activity of both normal development and disease. Necroptosis is early recognized as a caspase-independent form of programmed cell death followed obviously inflammation. Apoptosis is a gradually recognized mode of cell death that is characterized by a special morphological changes and unique caspase-dependent biological process. Ferroptosis, pyroptosis and autophagy are recently identified non-apoptotic regulated cell death that each has its own characteristics. The transient receptor potential vanilloid 4 (TRPV4) is a kind of nonselective calcium-permeable cation channel, which is received more and more attention in biology studies. It is widely expressed in human tissues and mainly located on the membrane of cells. Several researchers have identified that the influx Ca2+ from TRPV4 acts as a key role in the loss of cells by apoptosis, ferroptosis, necroptosis, pyroptosis and autophagy via mediating endoplasmic reticulum (ER) stress, oxidative stress and inflammation. This effect is bad for the normal function of organs on the one hand, on the other hand, it is benefit for anticancer activities. In this review, we will summarize the current discovery on the role and impact of TRPV4 in these programmed cell death pathological mechanisms to provide a new prospect of gene therapeutic target of related diseases.
Collapse
Affiliation(s)
- Qingjie Ma
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Jilin Wu
- Department of Anesthesiology, Kunming Children's Hospital, Kunming, 650034, China
| | - Huixian Li
- Department of Anesthesiology, The People's Hospital of Wenshan Zhuang and Miao Minority Autonomous Prefecture, Wenshan, 663099, China
| | - Xiaoshu Ma
- The Second Clinical Medical College of Binzhou Medical College, Binzhou, 256699, China
| | - Renwan Yin
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Liping Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Heng Tang
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Na Liu
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China.
| |
Collapse
|
10
|
Song L, Wang H, Peng R. Advances in the Regulation of Neural Function by Infrared Light. Int J Mol Sci 2024; 25:928. [PMID: 38256001 PMCID: PMC10815576 DOI: 10.3390/ijms25020928] [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: 12/19/2023] [Revised: 01/02/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
In recent years, with the rapid development of optical technology, infrared light has been increasingly used in biomedical fields. Research has shown that infrared light could play roles in light stimulation and biological regulation. Infrared light has been used to regulate neural function due to its high spatial resolution, safety and neural sensitivity and has been considered a useful method to replace traditional neural regulation approaches. Infrared neuromodulation methods have been used for neural activation, central nervous system disorder treatment and cognitive enhancement. Research on the regulation of neural function by infrared light stimulation began only recently, and the underlying mechanism remains unclear. This article reviews the characteristics of infrared light, the advantages and disadvantages of infrared neuromodulation, its effects on improving individual health, and its mechanism. This article aims to provide a reference for future research on the use of infrared neural regulation to treat neuropsychological disorders.
Collapse
Affiliation(s)
| | - Hui Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ruiyun Peng
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
11
|
Almasri RM, Ladouceur F, Mawad D, Esrafilzadeh D, Firth J, Lehmann T, Poole-Warren LA, Lovell NH, Al Abed A. Emerging trends in the development of flexible optrode arrays for electrophysiology. APL Bioeng 2023; 7:031503. [PMID: 37692375 PMCID: PMC10491464 DOI: 10.1063/5.0153753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023] Open
Abstract
Optical-electrode (optrode) arrays use light to modulate excitable biological tissues and/or transduce bioelectrical signals into the optical domain. Light offers several advantages over electrical wiring, including the ability to encode multiple data channels within a single beam. This approach is at the forefront of innovation aimed at increasing spatial resolution and channel count in multichannel electrophysiology systems. This review presents an overview of devices and material systems that utilize light for electrophysiology recording and stimulation. The work focuses on the current and emerging methods and their applications, and provides a detailed discussion of the design and fabrication of flexible arrayed devices. Optrode arrays feature components non-existent in conventional multi-electrode arrays, such as waveguides, optical circuitry, light-emitting diodes, and optoelectronic and light-sensitive functional materials, packaged in planar, penetrating, or endoscopic forms. Often these are combined with dielectric and conductive structures and, less frequently, with multi-functional sensors. While creating flexible optrode arrays is feasible and necessary to minimize tissue-device mechanical mismatch, key factors must be considered for regulatory approval and clinical use. These include the biocompatibility of optical and photonic components. Additionally, material selection should match the operating wavelength of the specific electrophysiology application, minimizing light scattering and optical losses under physiologically induced stresses and strains. Flexible and soft variants of traditionally rigid photonic circuitry for passive optical multiplexing should be developed to advance the field. We evaluate fabrication techniques against these requirements. We foresee a future whereby established telecommunications techniques are engineered into flexible optrode arrays to enable unprecedented large-scale high-resolution electrophysiology systems.
Collapse
Affiliation(s)
- Reem M. Almasri
- Graduate School of Biomedical Engineering, UNSW, Sydney, NSW 2052, Australia
| | | | - Damia Mawad
- School of Materials Science and Engineering, UNSW, Sydney, NSW 2052, Australia
| | - Dorna Esrafilzadeh
- Graduate School of Biomedical Engineering, UNSW, Sydney, NSW 2052, Australia
| | - Josiah Firth
- Australian National Fabrication Facility, UNSW, Sydney, NSW 2052, Australia
| | - Torsten Lehmann
- School of Electrical Engineering and Telecommunications, UNSW, Sydney, NSW 2052, Australia
| | | | | | - Amr Al Abed
- Graduate School of Biomedical Engineering, UNSW, Sydney, NSW 2052, Australia
| |
Collapse
|
12
|
Tian L, Zeng M, Tian G, Xu J. In-vitro quantitative measurement and analysis of the photosensitivity of cells to a weak pulse laser. BIOMEDICAL OPTICS EXPRESS 2023; 14:3584-3596. [PMID: 37497496 PMCID: PMC10368051 DOI: 10.1364/boe.494620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/05/2023] [Accepted: 06/13/2023] [Indexed: 07/28/2023]
Abstract
Light can trigger electrical activity in certain types of cells, and is considered to be a better means of biological regulation than electrical stimulation in the future. Due to the specificity and selectivity of natural cells' photoresponse to optical signals, constructing an applicable method to explore which kinds of cells have photosensitivity and which bands of light could induce its photoresponse most effectively, is of great significance for lights' medical applications. This paper firstly proposed a universal and operable system and corresponding method to quantitatively measure and analyze photosensitivity of cells in vitro to weak pulse laser, which is constructed with Ca2+ imaging module, adjustable laser lights module and laser positioning module. With the measurement system and method, the photosensitive effects of the natural spiral ganglion cells (SGCs) of mice are tested systemantically. Then a new photoresponse band of light (453 nm, 300 µs) is found for SGCs, and its minimum threshold is measured as 5.3 mJ/cm2. The results verify that the proposed method is applicable to screen the cells with photosensitive response, as well as to measure and analyze the working optical parameters, thus is beneficial for the optical biophysics and photobiology.
Collapse
Affiliation(s)
- Lan Tian
- School of Microelectronics, Shandong University, Jinan 250100, Shandong, China
| | - Ming Zeng
- School of Microelectronics, Shandong University, Jinan 250100, Shandong, China
| | - Geng Tian
- School of Life Sciences, Shandong University, Qingdao 266237, Shandong, China
| | - Jingjing Xu
- School of Microelectronics, Shandong University, Jinan 250100, Shandong, China
| |
Collapse
|
13
|
Barrett JN, Barrett EF, Rajguru SM. Mitochondrial responses to intracellular Ca 2+ release following infrared stimulation. J Neurophysiol 2023; 129:700-716. [PMID: 36752512 PMCID: PMC10026987 DOI: 10.1152/jn.00293.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Many studies of Ca2+ effects on mitochondrial respiration in intact cells have used electrical and/or chemical stimulation to elevate intracellular [Ca2+], and have reported increases in [NADH] and increased ADP/ATP ratios as dominant controllers of respiration. This study tested a different form of stimulation: brief temperature increases produced by pulses of infrared light (IR, 1,863 nm, 8-10°C for ∼5 s). Fluorescence imaging techniques applied to single PC-12 cells in low µM extracellular [Ca2+] revealed IR stimulation-induced increases in both cytosolic (fluo5F) and mitochondrial (rhod2) [Ca2+]. IR stimulation increased O2 consumption (porphyrin fluorescence), and produced an alkaline shift in mitochondrial matrix pH (Snarf1), indicating activation of the electron transport chain (ETC). The increase in O2 consumption persisted in oligomycin, and began during a decrease in NADH, suggesting that the initial increase in ETC activity was not driven by increased ATP synthase activity or an increased fuel supply to ETC complex I. Imaging with two potentiometric dyes [tetramethyl rhodamine methyl ester (TMRM) and R123] indicated a depolarizing shift in ΔΨm that persisted in high [K+] medium. High-resolution fluorescence imaging disclosed large, reversible mitochondrial depolarizations that were inhibited by cyclosporin A (CSA), consistent with the opening of transient mitochondrial permeability transition pores. IR stimulation also produced a Ca2+-dependent increase in superoxide production (MitoSox) that was not inhibited by CSA, indicating that the increase in superoxide did not require transition pore opening. Thus, the intracellular Ca2+ release that follows pulses of infrared light offers new insights into Ca2+-dependent processes controlling respiration and reactive oxygen species in intact cells.NEW & NOTEWORTHY Pulses of infrared light (IR) provide a novel method for rapidly transferring Ca2+ from the endoplasmic reticulum to mitochondria in intact cells. In PC12 cells the resulting ETC activation was not driven by increased ATP synthase activity or NADH. IR stimulation produced a Ca2+-dependent, reversible depolarization of ΔΨm that was partially blocked by cyclosporin A, and a Ca2+-dependent increase in superoxide that did not require transition pore opening.
Collapse
Affiliation(s)
- John N Barrett
- Department of Physiology and Biophysics, University of Miami, Florida, United States
- Neuroscience Program, University of Miami, Florida, United States
| | - Ellen F Barrett
- Department of Physiology and Biophysics, University of Miami, Florida, United States
- Neuroscience Program, University of Miami, Florida, United States
| | - Suhrud M Rajguru
- Department of Biomedical Engineering, University of Miami, Florida, United States
- Department of Otolaryngology, University of Miami, Florida, United States
| |
Collapse
|
14
|
Pahlavan B, Buitrago N, Santamaria F. Macromolecular rate theory explains the temperature dependence of membrane conductance kinetics. Biophys J 2023; 122:522-532. [PMID: 36567527 PMCID: PMC9941726 DOI: 10.1016/j.bpj.2022.12.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 10/19/2022] [Accepted: 12/23/2022] [Indexed: 12/25/2022] Open
Abstract
The factor Q10 is used in neuroscience to adjust reaction rates of voltage-activated membrane conductances to different temperatures and is widely assumed to be constant. By performing an analysis of published data of the reaction rates of sodium, potassium, and calcium membrane conductances, we demonstrate that 1) Q10 is temperature dependent, 2) this relationship is similar across conductances, and 3) there is a strong effect at low temperatures (<15°C). We show that macromolecular rate theory (MMRT) explains this temperature dependency. MMRT predicts the existence of optimal temperatures at which reaction rates decrease as temperature increases, a phenomenon that we also found in the published data sets. We tested the consequences of using MMRT-adjusted reaction rates in the Hodgkin-Huxley model of the squid's giant axon. The MMRT-adjusted model reproduces the temperature dependence of the rising and falling times of the action potential. Furthermore, the model also reproduces these properties for different squid species that live in different climates. In a second example, we compare spiking patterns of biophysical models based on human pyramidal neurons from the Allen Cell Types database at room and physiological temperatures. The original models, calibrated at 34°C, failed to generate realistic spikes at room temperature in more than half of the tested models, while the MMRT produces realistic spiking in all conditions. In another example, we show that using the MMRT correction in hippocampal pyramidal cell models results in 100% differences in voltage responses. Finally, we show that the shape of the Q10 function results in systematic errors in predicting reaction rates. We propose that the optimal temperature could be a thermodynamical barrier to avoid over excitation in neurons. While this study is centered on membrane conductances, our results have important consequences for all biochemical reactions involved in cell signaling.
Collapse
Affiliation(s)
- Bahram Pahlavan
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas
| | - Nicolas Buitrago
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas
| | - Fidel Santamaria
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas.
| |
Collapse
|
15
|
Heat-hypersensitive mutants of ryanodine receptor type 1 revealed by microscopic heating. Proc Natl Acad Sci U S A 2022; 119:e2201286119. [PMID: 35925888 PMCID: PMC9371657 DOI: 10.1073/pnas.2201286119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Malignant hyperthermia (MH) is a life-threatening disorder caused largely by mutations in ryanodine receptor type 1 (RyR1) Ca2+-release channels. Enhanced Ca2+ release through the mutant channels induces excessive heat development upon exposure to volatile anesthetics. However, the mechanism by which Ca2+ release is accelerated at an elevated temperature is yet to be identified. Fluorescence Ca2+ imaging with rapid heating by an infrared laser beam provides direct evidence that heat induces Ca2+ release through the RyR1 channel. And the mutant channels are more heat sensitive than the wild-type channels, thereby causing an increase in the cytosolic Ca2+ concentration in mutant cells. It is likely that the heat-induced Ca2+ release participates as an enhancer in the cellular mechanism of MH. Thermoregulation is an important aspect of human homeostasis, and high temperatures pose serious stresses for the body. Malignant hyperthermia (MH) is a life-threatening disorder in which body temperature can rise to a lethal level. Here we employ an optically controlled local heat-pulse method to manipulate the temperature in cells with a precision of less than 1 °C and find that the mutants of ryanodine receptor type 1 (RyR1), a key Ca2+ release channel underlying MH, are heat hypersensitive compared with the wild type (WT). We show that the local heat pulses induce an intracellular Ca2+ burst in human embryonic kidney 293 cells overexpressing WT RyR1 and some RyR1 mutants related to MH. Fluorescence Ca2+ imaging using the endoplasmic reticulum–targeted fluorescent probes demonstrates that the Ca2+ burst originates from heat-induced Ca2+ release (HICR) through RyR1-mutant channels because of the channels’ heat hypersensitivity. Furthermore, the variation in the heat hypersensitivity of four RyR1 mutants highlights the complexity of MH. HICR likewise occurs in skeletal muscles of MH model mice. We propose that HICR contributes an additional positive feedback to accelerate thermogenesis in patients with MH.
Collapse
|
16
|
Adams WR, Gautam R, Locke A, Masson LE, Borrachero-Conejo AI, Dollinger B, Throckmorton GA, Duvall C, Jansen ED, Mahadevan-Jansen A. Visualizing Lipid Dynamics Role in Infrared Neural Stimulation using Stimulated Raman Scattering. Biophys J 2022; 121:1525-1540. [PMID: 35276133 PMCID: PMC9072573 DOI: 10.1016/j.bpj.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/14/2021] [Accepted: 03/04/2022] [Indexed: 11/02/2022] Open
Abstract
Infrared neural stimulation, or INS, uses pulsed infrared light to yield label-free neural stimulation with broad experimental and translational utility. Despite its robust demonstration, INS's mechanistic and biophysical underpinnings have been the subject of debate for more than a decade. The role of lipid membrane thermodynamics appears to play an important role in how fast IR-mediated heating nonspecifically drives action potential generation. Direct observation of lipid membrane dynamics during INS remains to be shown in a live neural model system. We used hyperspectral stimulated Raman scattering (hsSRS) microscopy to study biochemical signatures of high-speed vibrational dynamics underlying INS in a live neural cell culture model. Findings suggest that lipid bilayer structural changes are occurring during INS in vitro in NG108-15 neuroglioma cells. Lipid-specific signatures of cell SRS spectra varied with stimulation energy and radiant exposure. Spectroscopic observations agree with high-speed ratiometric fluorescence imaging of a conventional lipophilic membrane structure reporter, di-4-ANNEPS. Overall, the presented findings support the hypothesis that INS causes changes in the lipid membrane of neural cells by changing lipid membrane packing order. Furthermore, this work highlights the potential of hsSRS as a method to study biophysical and biochemical dynamics safely in live cells.
Collapse
Affiliation(s)
- Wilson R Adams
- Dept. of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Rekha Gautam
- Dept. of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Andrea Locke
- Dept. of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Laura E Masson
- Dept. of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | | | - Bryan Dollinger
- Dept. of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | | | - Craig Duvall
- Dept. of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - E Duco Jansen
- Dept. of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Dept. of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anita Mahadevan-Jansen
- Dept. of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Dept. of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
17
|
Jiang W, Wang Z, Xiao S, Zeng D, Wu Z, Peng C, Chen F. Pulsed infrared stimulation evoked electrical potential in mouse vestibular system. Neurosci Lett 2022; 775:136510. [DOI: 10.1016/j.neulet.2022.136510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/17/2022] [Accepted: 02/01/2022] [Indexed: 10/19/2022]
|
18
|
Liu N, Bai L, Lu Z, Gu R, Zhao D, Yan F, Bai J. TRPV4 contributes to ER stress and inflammation: implications for Parkinson’s disease. J Neuroinflammation 2022; 19:26. [PMID: 35093118 PMCID: PMC8800324 DOI: 10.1186/s12974-022-02382-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Background Parkinson’s disease (PD) is a progressive neurodegenerative disorder. Its molecular mechanism is still unclear, and pharmacological treatments are unsatisfactory. Transient receptor potential vanilloid 4 (TRPV4) is a nonselective Ca2+ channel. It has recently emerged as a critical risk factor in the pathophysiology of neuronal injuries and cerebral diseases. Our previous study reported that TRPV4 contributed to endoplasmic reticulum (ER) stress in the MPP+-induced cell model of PD. In the present study, we detected the role and the mechanism of TRPV4 in 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice. Methods Intracerebral injection of an adeno-associated virus (AAV) into the substantia nigra (SN) of mice was used to knockdown or upregulate the expression of TRPV4 and intraperitoneal injection of MPTP. Rotarod and pole tests were used to evaluate the locomotor ability of mice. We used immunohistochemistry, Nissl staining and Western blot to detect the alterations in the number of tyrosine hydroxylase (TH)-positive neurons, Nissl-positive neurons, the levels of ER stress-associated molecules and proinflammatory cytokines in the SN. Results The SN was transfected with AAV for 3 weeks and expressed the target protein with green fluorescence. Knockdown of TRPV4 via injection of a constructed AAV-TRPV4 shRNAi into the SN alleviated the movement deficits of PD mice. Upregulation of TRPV4 via injection of a constructed AAV-TRPV4 aggravated the above movement disorders. The expression of TRPV4 was upregulated in the SN of MPTP-treated mice. Injection of AAV-TRPV4 shRNAi into the SN rescued the number of TH-positive and Nissl-positive neurons in the SN decreased by MPTP, while injection of AAV-TRPV4 induced the opposite effect. Moreover, MPTP-decreased Sarco/endoplasmic reticulum Ca2+-ATPase 2 (SERCA2) and pro-cysteinyl aspartate specific proteinase-12 (procaspase-12), MPTP-increased Glucose-regulated protein 78 (GRP78), Glucose-regulated protein 94 (GRP94) and C/EBP homologous protein (CHOP) were inhibited by AAV-TRPV4 shRNAi infection, and enhanced by AAV-TRPV4. In the same way, MPTP-decreased procaspase-1, MPTP-increased Interleukin-18 (IL-18), Cyclooxgenase-2 (COX-2) and 5-Lipoxygenase (5-LOX) were inhibited by AAV-TRPV4 shRNAi, or further exacerbated by AAV-TRPV4. Conclusions These results suggest that TRPV4 mediates ER stress and inflammation pathways, contributing to the loss of dopamine (DA) neurons in the SN and movement deficits in PD mice. Moreover, this study provides a new perspective on molecular targets and gene therapies for the treatment of PD in the future.
Collapse
|
19
|
Zhu X, Lin JW, Turnali A, Sander MY. Single infrared light pulses induce excitatory and inhibitory neuromodulation. BIOMEDICAL OPTICS EXPRESS 2022; 13:374-388. [PMID: 35154878 PMCID: PMC8803021 DOI: 10.1364/boe.444577] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
The excitatory and inhibitory effects of single and brief infrared (IR) light pulses (2 µm) with millisecond durations and various power levels are investigated with a custom-built fiber amplification system. Intracellular recordings from motor axons of the crayfish opener neuromuscular junction are performed ex vivo. Single IR light pulses induce a membrane depolarization during the light pulses, which is followed by a hyperpolarization that can last up to 100 ms. The depolarization amplitude is dependent on the optical pulse duration, total energy deposition and membrane potential, but is insensitive to tetrodotoxin. The hyperpolarization reverses its polarity near the potassium equilibrium potential and is barium-sensitive. The membrane depolarization activates an action potential (AP) when the axon is near firing threshold, while the hyperpolarization reversibly inhibits rhythmically firing APs. In summary, we demonstrate for the first time that single and brief IR light pulses can evoke initial depolarization followed by hyperpolarization on individual motor axons. The corresponding mechanisms and functional outcomes of the dual effects are investigated.
Collapse
Affiliation(s)
- Xuedong Zhu
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, USA
- Neurophotonics Center, Boston University, 24 Cummington Mall, Boston, MA 02215, USA
- Photonics Center, Boston University, 8 Saint Mary’s Street, Boston, MA 02215, USA
| | - Jen-Wei Lin
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Ahmet Turnali
- Department of Electrical and Computer Engineering, Boston University, 8 Saint Mary’s Street, Boston, MA 02215, USA
- Photonics Center, Boston University, 8 Saint Mary’s Street, Boston, MA 02215, USA
| | - Michelle Y. Sander
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, USA
- Neurophotonics Center, Boston University, 24 Cummington Mall, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, Boston University, 8 Saint Mary’s Street, Boston, MA 02215, USA
- Photonics Center, Boston University, 8 Saint Mary’s Street, Boston, MA 02215, USA
- Division of Materials Science and Engineering, Boston University, 15 Saint Mary’s Street, Brookline, MA 02446, USA
| |
Collapse
|
20
|
Rice D, Martinelli GP, Jiang W, Holstein GR, Rajguru SM. Pulsed Infrared Stimulation of Vertical Semicircular Canals Evokes Cardiovascular Changes in the Rat. Front Neurol 2021; 12:680044. [PMID: 34122320 PMCID: PMC8193737 DOI: 10.3389/fneur.2021.680044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 04/20/2021] [Indexed: 11/28/2022] Open
Abstract
A variety of stimuli activating vestibular end organs, including sinusoidal galvanic vestibular stimulation, whole body rotation and tilt, and head flexion have been shown to evoke significant changes in blood pressure (BP) and heart rate (HR). While a role for the vertical semicircular canals in altering autonomic activity has been hypothesized, studies to-date attribute the evoked BP and HR responses to the otolith organs. The present study determined whether unilateral activation of the posterior (PC) or anterior (AC) semicircular canal is sufficient to elicit changes in BP and/or HR. The study employed frequency-modulated pulsed infrared radiation (IR: 1,863 nm) directed via optical fibers to PC or AC of adult male Long-Evans rats. BP and HR changes were detected using a small-animal single pressure telemetry device implanted in the femoral artery. Eye movements evoked during IR of the vestibular endorgans were used to confirm the stimulation site. We found that sinusoidal IR delivered to either PC or AC elicited a rapid decrease in BP and HR followed by a stimulation frequency-matched modulation. The magnitude of the initial decrements in HR and BP did not correlate with the energy of the suprathreshold stimulus. This response pattern was consistent across multiple trials within an experimental session, replicable, and in most animals showed no evidence of habituation or an additive effect. Frequency modulated electrical current delivered to the PC and IR stimulation of the AC, caused decrements in HR and BP that resembled those evoked by IR of the PC. Frequency domain heart rate variability assessment revealed that, in most subjects, IR stimulation increased the low frequency (LF) component and decreased the high frequency (HF) component, resulting in an increase in the LF/HF ratio. This ratio estimates the relative contributions of sympathetic nervous system (SNS) and parasympathetic nervous system (PNS) activities. An injection of atropine, a muscarinic cholinergic receptor antagonist, diminished the IR evoked changes in HR, while the non-selective beta blocker propranolol eliminated changes in both HR and BP. This study provides direct evidence that activation of a single vertical semicircular canal is sufficient to activate and modulate central pathways that control HR and BP.
Collapse
Affiliation(s)
- Darrian Rice
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States
| | - Giorgio P Martinelli
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Weitao Jiang
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States
| | - Gay R Holstein
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Suhrud M Rajguru
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States.,Department of Otolaryngology, University of Miami, Miami, FL, United States
| |
Collapse
|
21
|
Kaszas A, Szalay G, Slézia A, Bojdán A, Vanzetta I, Hangya B, Rózsa B, O'Connor R, Moreau D. Two-photon GCaMP6f imaging of infrared neural stimulation evoked calcium signals in mouse cortical neurons in vivo. Sci Rep 2021; 11:9775. [PMID: 33963220 PMCID: PMC8105372 DOI: 10.1038/s41598-021-89163-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
Infrared neural stimulation is a promising tool for stimulating the brain because it can be used to excite with high spatial precision without the need of delivering or inserting any exogenous agent into the tissue. Very few studies have explored its use in the brain, as most investigations have focused on sensory or motor nerve stimulation. Using intravital calcium imaging with the genetically encoded calcium indicator GCaMP6f, here we show that the application of infrared neural stimulation induces intracellular calcium signals in Layer 2/3 neurons in mouse cortex in vivo. The number of neurons exhibiting infrared-induced calcium response as well as the amplitude of those signals are shown to be both increasing with the energy density applied. By studying as well the spatial extent of the stimulation, we show that reproducibility of the stimulation is achieved mainly in the central part of the infrared beam path. Stimulating in vivo at such a degree of precision and without any exogenous chromophores enables multiple applications, from mapping the brain's connectome to applications in systems neuroscience and the development of new therapeutic tools for investigating the pathological brain.
Collapse
Affiliation(s)
- Attila Kaszas
- Mines Saint-Etienne, Centre CMP, Département BEL, F - 13541, Gardanne, France
- Institut de Neurosciences de la Timone, CNRS UMR 7289 & Aix-Marseille Université, 13005, Marseille, France
| | - Gergely Szalay
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, 1083, Hungary
| | - Andrea Slézia
- Mines Saint-Etienne, Centre CMP, Département BEL, F - 13541, Gardanne, France
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, Budapest, 1083, Hungary
| | - Alexandra Bojdán
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, 1083, Hungary
| | - Ivo Vanzetta
- Institut de Neurosciences de la Timone, CNRS UMR 7289 & Aix-Marseille Université, 13005, Marseille, France
| | - Balázs Hangya
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, Budapest, 1083, Hungary
| | - Balázs Rózsa
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest, 1083, Hungary
- Two-Photon Laboratory, Faculty of Information Technology, Pázmány Péter Catholic University, Budapest, 1083, Hungary
| | - Rodney O'Connor
- Mines Saint-Etienne, Centre CMP, Département BEL, F - 13541, Gardanne, France
- Institut de Neurosciences de la Timone, CNRS UMR 7289 & Aix-Marseille Université, 13005, Marseille, France
| | - David Moreau
- Mines Saint-Etienne, Centre CMP, Département BEL, F - 13541, Gardanne, France.
| |
Collapse
|
22
|
Littlefield PD, Richter C. Near-infrared stimulation of the auditory nerve: A decade of progress toward an optical cochlear implant. Laryngoscope Investig Otolaryngol 2021; 6:310-319. [PMID: 33869763 PMCID: PMC8035937 DOI: 10.1002/lio2.541] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 12/14/2020] [Accepted: 02/12/2021] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES We provide an appraisal of recent research on stimulation of the auditory system with light. In particular, we discuss direct infrared stimulation and ongoing controversies regarding the feasibility of this modality. We also discuss advancements and barriers to the development of an optical cochlear implant. METHODS This is a review article that covers relevant animal studies. RESULTS The auditory system has been stimulated with infrared light, and in a much more spatially selective manner than with electrical stimulation. However, there are experiments from other labs that have not been able to reproduce these results. This has resulted in an ongoing controversy regarding the feasibility of infrared stimulation, and the reasons for these experimental differences still require explanation. The neural response characteristics also appear to be much different than with electrical stimulation. The electrical stimulation paradigms used for modern cochlear implants do not apply well to optical stimulation and new coding strategies are under development. Stimulation with infrared light brings the risk of heat accumulation in the tissue at high pulse repetition rates, so optimal pulse shapes and combined optical/electrical stimulation are being investigated to mitigate this. Optogenetics is another promising technique, which makes neurons more sensitive to light stimulation by inserting light sensitive ion channels via viral vectors. Challenges of optogenetics include the expression of light sensitive channels in sufficient density in the target neurons, and the risk of damaging neurons by the expression of a foreign protein. CONCLUSION Optical stimulation of the nervous system is a promising new field, and there has been progress toward the development of a cochlear implant that takes advantage of the benefits of optical stimulation. There are barriers, and controversies, but so far none that seem intractable. LEVEL OF EVIDENCE NA (animal studies and basic research).
Collapse
Affiliation(s)
| | - Claus‐Peter Richter
- Department of OtolaryngologyNorthwestern UniversityChicagoIllinoisUSA
- Department of Communication Sciences and DisordersNorthwestern UniversityEvanstonIllinoisUSA
- Department of Biomedical EngineeringNorthwestern UniversityEvanstonIllinoisUSA
- The Hugh Knowles Center, Department of Communication Sciences and DisordersNorthwestern UniversityEvanstonIllinoisUSA
| |
Collapse
|
23
|
Brown WGA, Needham K, Begeng JM, Thompson AC, Nayagam BA, Kameneva T, Stoddart PR. Response of primary auditory neurons to stimulation with infrared light in vitro. J Neural Eng 2021; 18:046003. [PMID: 33724234 DOI: 10.1088/1741-2552/abe7b8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Infrared light can be used to modulate the activity of neuronal cells through thermally-evoked capacitive currents and thermosensitive ion channel modulation. The infrared power threshold for action potentials has previously been found to be far lower in the in vivo cochlea when compared with other neuronal targets, implicating spiral ganglion neurons (SGNs) as a potential target for infrared auditory prostheses. However, conflicting experimental evidence suggests that this low threshold may arise from an intermediary mechanism other than direct SGN stimulation, potentially involving residual hair cell activity. APPROACH Patch-clamp recordings from cultured SGNs were used to explicitly quantify the capacitive and ion channel currents in an environment devoid of hair cells. Neurons were irradiated by a 1870 nm laser with pulse durations of 0.2-5.0 ms and powers up to 1.5 W. A Hodgkin-Huxley-type model was established by first characterising the voltage dependent currents, and then incorporating laser-evoked currents separated into temperature-dependent and temperature-gradient-dependent components. This model was found to accurately simulate neuronal responses and allowed the results to be extrapolated to stimulation parameter spaces not accessible during this study. MAIN RESULTS The previously-reported low in vivo SGN stimulation threshold was not observed, and only subthreshold depolarisation was achieved, even at high light exposures. Extrapolating these results with our Hodgkin-Huxley-type model predicts an action potential threshold which does not deviate significantly from other neuronal types. SIGNIFICANCE This suggests that the low-threshold response that is commonly reported in vivo may arise from an alternative mechanism, and calls into question the potential usefulness of the effect for auditory prostheses. The step-wise approach to modelling optically-evoked currents described here may prove useful for analysing a wider range of cell types where capacitive currents and conductance modulation are dominant.
Collapse
Affiliation(s)
- William G A Brown
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, John Street, Hawthorn VIC 3122, Australia
| | | | | | | | | | | | | |
Collapse
|
24
|
Fekete Z, Horváth ÁC, Zátonyi A. Infrared neuromodulation:a neuroengineering perspective. J Neural Eng 2020; 17:051003. [PMID: 33055373 DOI: 10.1088/1741-2552/abb3b2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Infrared neuromodulation (INM) is a branch of photobiomodulation that offers direct or indirect control of cellular activity through elevation of temperature in a spatially confined region of the target tissue. Research on INM started about 15 ago and is gradually attracting the attention of the neuroscience community, as numerous experimental studies have provided firm evidence on the safe and reproducible excitation and inhibition of neuronal firing in both in vitro and in vivo conditions. However, its biophysical mechanism is not fully understood and several engineered interfaces have been created to investigate infrared stimulation in both the peripheral and central nervous system. In this review, recent applications and present knowledge on the effects of INM on cellular activity are summarized, and an overview of the technical approaches to deliver infrared light to cells and to interrogate the optically evoked response is provided. The micro- and nanoengineered interfaces used to investigate the influence of INM are described in detail.
Collapse
Affiliation(s)
- Z Fekete
- Research Group for Implantable Microsystems, Faculty of Information Technology & Bionics, Pázmány Péter Catholic University, Budapest 1083, Hungary. Author to whom any correspondence should be addressed
| | | | | |
Collapse
|
25
|
Zhou J. Two potential molecular signaling pathways of the UFL1 gene to induce the endoplasmic reticulum stress and apoptosis of the ovarian granulosa cell. Med Hypotheses 2020; 145:110328. [PMID: 33035966 DOI: 10.1016/j.mehy.2020.110328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 11/17/2022]
Abstract
Endoplasmic reticulum stress (ERS) is a crucial physiological and pathological process takes place in the endoplasmic reticulum that usually induced by various intracellular and extracellular factors. It causes multiple diseases, including breast cancer, hepatocellular carcinoma, and premature ovarian failure that mainly associates with the ovarian granulosa cells. To effectively alleviate and cure the ERS and following diseases, molecular signaling pathways that are responsible for inducing ERS must be deeply investigated. There are many intracellular pathways to initiate the ERS, among which, detailed molecular mechanism the UFM1-specific ligase 1 (UFL1) gene induced analogous ubiquitylation related pathway is still unclear. However, some researches have reported that the UFL1 gene is responsible for initiating the ERS in the ovarian granulosa cell and premature ovarian failure. In this article, a new, highly possible molecular signaling pathway is proposed and hoping to provide a unique aspect for the following researches about ERS, especially in the ovarian granulosa cell.
Collapse
Affiliation(s)
- Jingyang Zhou
- Class 182, Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, Jiangxi Province, People's Republic of China.
| |
Collapse
|
26
|
Liu N, Liu J, Wen X, Bai L, Shao R, Bai J. TRPV4 contributes to ER stress: Relation to apoptosis in the MPP +-induced cell model of Parkinson's disease. Life Sci 2020; 261:118461. [PMID: 32961227 DOI: 10.1016/j.lfs.2020.118461] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/04/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022]
Abstract
AIMS Parkinson's disease (PD) is a multifactorial neurodegenerative disorder. Its molecular mechanism is still unclear. Endoplasmic reticulum (ER) stress has been highlighted in PD. Transient receptor potential vanilloid 4 (TRPV4) is a kind of nonselective calcium cation channel. A defined role for TRPV4 in PD has not been reported. The purpose of the present research was to investigate the molecular mechanisms by which TRPV4 regulates ER stress induced by the 1-methyl-4-phenylpyridinium ion (MPP+) in PC12 cells. MAIN METHODS PC12 cells were pretreated with the TRPV4-specific antagonist HC067047 or transfected with TRPV4 siRNA followed by treatment with MPP+. Cell viability was measured by the CCK-8 Assay. The expression of TRPV4, sarco/endoplasmic reticulum Ca2+-ATPase 2 (SERCA2), glucose-regulated protein 78 (GRP78), glucose-regulated protein 94 (GRP94), C/EBP homologous protein (CHOP), procaspase-12, and tyrosine hydroxylase (TH) was detected by western blot and RT-PCR. KEY FINDINGS The expression of TRPV4 was upregulated, while cell viability was decreased by MPP+, which was reversed by HC067047. The ER stress common molecular signature SERCA2 was depressed by MPP+. Moreover, MPP+ induced upregulation of GRP78, GRP94, CHOP, and decrease in procaspase-12 and TH. HC067047 and TRPV4 siRNA reversed MPP+-induced ER stress and restored TH production. SIGNIFICANCE TRPV4 functions upstream of ER stress induced by MPP+ and holds promise as a prospective pharmacotherapy target for PD.
Collapse
Affiliation(s)
- Na Liu
- Faculty of Life science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Department of Anesthesiology, The First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Jinyu Liu
- Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Xianbin Wen
- Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Liping Bai
- Faculty of Life science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Ruifei Shao
- Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming 650500, China.
| |
Collapse
|
27
|
Channels that Cooperate with TRPV4 in the Brain. J Mol Neurosci 2020; 70:1812-1820. [PMID: 32524421 DOI: 10.1007/s12031-020-01574-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/27/2020] [Indexed: 12/26/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a nonselective Ca2+-permeable cation channel that is a member of the TRP channel family. It is clear that TRPV4 channels are broadly expressed in the brain. As they are expressed on the plasma membrane, they interact with other channels and play a crucial role in nervous system activity. Under some pathological conditions, TRPV4 channels are upregulated and sensitized via cellular signaling pathways, and this can cause nervous system diseases. In this review, we focus on receptors that cooperate with TRPV4, including large-conductance Ca2+-activated K+(BKca) channels, N-methyl-D-aspartate receptors (NMDARs), α-amino-3-hydroxy-5-methyl-4-isoxazole-propionate receptors (AMPARs), inositol 1,4,5-trisphosphate receptors (IP3Rs), ryanodine receptors (RyRs), aquaporin 4 (AQP4), and other potential cooperative receptors in the brain. The data demonstrate how these channels work together to cause nervous system diseases under pathological conditions. The aim of this review was to discuss the receptors and signaling pathways related to TRPV4 based on recent data on the important physiological functions of TRPV4 channels to provide new clues for future studies and prospective therapeutic targets for related brain diseases.
Collapse
|
28
|
Borrachero-Conejo AI, Adams WR, Saracino E, Mola MG, Wang M, Posati T, Formaggio F, De Bellis M, Frigeri A, Caprini M, Hutchinson MR, Muccini M, Zamboni R, Nicchia GP, Mahadevan-Jansen A, Benfenati V. Stimulation of water and calcium dynamics in astrocytes with pulsed infrared light. FASEB J 2020; 34:6539-6553. [PMID: 32202681 DOI: 10.1096/fj.201903049r] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/25/2020] [Accepted: 03/06/2020] [Indexed: 06/14/2024]
Abstract
Astrocytes are non-neuronal cells that govern the homeostatic regulation of the brain through ions and water transport, and Ca2+ -mediated signaling. As they are tightly integrated into neural networks, label-free tools that can modulate cell function are needed to evaluate the role of astrocytes in brain physiology and dysfunction. Using live-cell fluorescence imaging, pharmacology, electrophysiology, and genetic manipulation, we show that pulsed infrared light can modulate astrocyte function through changes in intracellular Ca2+ and water dynamics, providing unique mechanistic insight into the effect of pulsed infrared laser light on astroglial cells. Water transport is activated and, IP3 R, TRPA1, TRPV4, and Aquaporin-4 are all involved in shaping the dynamics of infrared pulse-evoked intracellular calcium signal. These results demonstrate that astrocyte function can be modulated with infrared light. We expect that targeted control over calcium dynamics and water transport will help to study the crucial role of astrocytes in edema, ischemia, glioma progression, stroke, and epilepsy.
Collapse
Affiliation(s)
- Ana I Borrachero-Conejo
- Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche, Bologna, Italy
| | - Wilson R Adams
- Department Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Biophotonics Center, Vanderbilt University, Nashville, TN, USA
| | - Emanuela Saracino
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Ricerche, Bologna, Italy
| | - Maria Grazia Mola
- Department of Bioscience, Biotechnology and Biopharmaceutics and Centre of Excellence in Comparative Genomics, University of Bari Aldo Moro, Bari, Italy
| | - Manqing Wang
- Vanderbilt Biophotonics Center, Vanderbilt University, Nashville, TN, USA
- Bioengineering College, Chongqing University, Chongqing, China
| | - Tamara Posati
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Ricerche, Bologna, Italy
| | - Francesco Formaggio
- Dipartimento di Farmacia e Biotecnologie, University of Bologna, Bologna, Italy
| | - Manuela De Bellis
- Department of Bioscience, Biotechnology and Biopharmaceutics and Centre of Excellence in Comparative Genomics, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Frigeri
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, School of Medicine, University of Bari Aldo Moro, Bari, Italy
- Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University, New York, NY, USA
| | - Marco Caprini
- Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche, Bologna, Italy
- Dipartimento di Farmacia e Biotecnologie, University of Bologna, Bologna, Italy
| | - Mark R Hutchinson
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Michele Muccini
- Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche, Bologna, Italy
| | - Roberto Zamboni
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Ricerche, Bologna, Italy
| | - Grazia Paola Nicchia
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Ricerche, Bologna, Italy
- Department of Bioscience, Biotechnology and Biopharmaceutics and Centre of Excellence in Comparative Genomics, University of Bari Aldo Moro, Bari, Italy
- Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University, New York, NY, USA
| | - Anita Mahadevan-Jansen
- Department Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Biophotonics Center, Vanderbilt University, Nashville, TN, USA
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Valentina Benfenati
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Ricerche, Bologna, Italy
| |
Collapse
|
29
|
Role of the TRPV Channels in the Endoplasmic Reticulum Calcium Homeostasis. Cells 2020; 9:cells9020317. [PMID: 32013022 PMCID: PMC7072170 DOI: 10.3390/cells9020317] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/08/2020] [Accepted: 01/21/2020] [Indexed: 01/01/2023] Open
Abstract
It has been widely established that transient receptor potential vanilloid (TRPV) channels play a crucial role in calcium homeostasis in mammalian cells. Modulation of TRPV channels activity can modify their physiological function leading to some diseases and disorders like neurodegeneration, pain, cancer, skin disorders, etc. It should be noted that, despite TRPV channels importance, our knowledge of the TRPV channels functions in cells is mostly limited to their plasma membrane location. However, some TRPV channels were shown to be expressed in the endoplasmic reticulum where their modulation by activators and/or inhibitors was demonstrated to be crucial for intracellular signaling. In this review, we have intended to summarize the poorly studied roles and functions of these channels in the endoplasmic reticulum.
Collapse
|
30
|
Zhu X, Lin JW, Sander MY. Infrared inhibition and waveform modulation of action potentials in the crayfish motor axon. BIOMEDICAL OPTICS EXPRESS 2019; 10:6580-6594. [PMID: 31853418 PMCID: PMC6913409 DOI: 10.1364/boe.10.006580] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/12/2019] [Accepted: 11/19/2019] [Indexed: 05/02/2023]
Abstract
The infrared (IR) inhibition of axonal activities in the crayfish neuromuscular preparation is studied using 2 µm IR light pulses with varying durations. The intracellular neuronal activities are monitored with two-electrode current clamp, while the IR-induced temperature changes are measured by the open patch technique simultaneously. It is demonstrated that the IR pulses can reversibly shape or block locally initiated action potentials. Suppression of the AP amplitude and duration and decrease in axonal excitability by IR pulses are quantitatively analyzed. While the AP amplitude and duration decrease similarly during IR illumination, it is discovered that the recovery of the AP duration after the IR pulses is slower than that of the AP amplitude. An IR-induced decrease in the input resistance (8.8%) is detected and discussed together with the temperature dependent changes in channel kinetics as contributing factors for the inhibition reported here.
Collapse
Affiliation(s)
- Xuedong Zhu
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, USA
- Neurophotonics Center, Boston University, 24 Cummington Mall, Boston, MA 02215, USA
- Photonics Center, Boston University, 8 Saint Mary’s Street, Boston, MA 02215, USA
| | - Jen-Wei Lin
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Michelle Y. Sander
- Neurophotonics Center, Boston University, 24 Cummington Mall, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, Boston University, 8 Saint Mary’s Street, Boston, MA 02215, USA
- Photonics Center, Boston University, 8 Saint Mary’s Street, Boston, MA 02215, USA
- Division of Materials Science and Engineering, Boston University, 15 Saint Mary’s Street, Brookline, MA 02446, USA
| |
Collapse
|
31
|
Jiang W, Rajguru SM. Eye Movements Evoked by Pulsed Infrared Radiation of the Rat Vestibular System. Ann Biomed Eng 2018; 46:1406-1418. [PMID: 29845411 PMCID: PMC6095805 DOI: 10.1007/s10439-018-2059-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/24/2018] [Indexed: 10/16/2022]
Abstract
Light at infrared wavelengths has been demonstrated to modulate the pattern of neural signals transmitted from the angular motion sensing semicircular canals of the vestibular system to the brain. In the present study, we have characterized physiological eye movements evoked by focused, pulsed infrared radiation (IR) stimuli directed at an individual semicircular canal in a mammalian model. Pulsed IR (1863 nm) trains were directed at the posterior semicircular canal in a rat using 200-400 µm optical fibers. Evoked bilateral eye movements were measured using a custom-modified video-oculography system. The activation of vestibulo-ocular motor pathways by frequency modulated pulsed IR directed at single posterior semicircular canals evoked significant, characteristic bilateral eye movements. In this case, the resulting eye movements were disconjugate with ipsilateral eye moving upwards with a rotation towards the stimulated ear and the contralateral eye moving downwards. The eye movements were stable through several hours of repeated stimulation and could be maintained with 30 + minutes of continuous, frequency-modulated IR stimulation. Following the measurements, the distance of the fiber from target structures and orientation of the beam relative to vestibular structures were determined using micro-computed tomography. Results highlight the spatial selectivity of optical stimulation. Our results demonstrate a novel strategy for direct optical stimulation of the vestibular pathway in rodents and lays the groundwork for future applications of optical neural stimulation in inner ear research and therapeutic applications.
Collapse
Affiliation(s)
- Weitao Jiang
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, MEA 204, Coral Gables, FL, 33146, USA
| | - Suhrud M Rajguru
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, MEA 204, Coral Gables, FL, 33146, USA.
- Department of Otolaryngology, University of Miami, 1600 NW 10th Ave, RMSB 3160, Miami, FL, 33136, USA.
| |
Collapse
|