1
|
Otake S, Saito S, Lin X, Saito CT, Kohno S, Takagi W, Hyodo S, Tominaga M, Katsu Y. Functional characterization of thermosensitive TRPV channels from holocephalan elephant shark (Callorhinchus milii) illuminate the ancestral thermosensory system in vertebrates. J Exp Biol 2025; 228:JEB249961. [PMID: 39916595 DOI: 10.1242/jeb.249961] [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: 12/11/2023] [Accepted: 02/02/2025] [Indexed: 03/11/2025]
Abstract
Homeostasis and survival of various animal species have been affected by changes in environmental temperature, causing animals to evolve physiological systems for sensing ambient and body temperature. Temperature-sensitive transient receptor potential (TRP) channels have multimodal properties that are activated by physical stimuli such as temperature, as well as by various chemical substances. Our goal is to understand the diversity of the vertebrate thermosensory system by characterizing the temperature-sensitive TRPV channels of the elephant shark, which belongs to the Holocephali of the cartilaginous fishes. Since elephant sharks are basal jawed vertebrates, analysis of elephant shark TRPs is critical to understanding the evolution of thermosensory systems in vertebrate lineages. We found that temperature stimulation activated elephant shark TRPVs in an electrophysiological analysis similarly to the mammalian ortholog. The thermal activation threshold of elephant shark TRPV1 (31°C) was similar to the thresholds reported for several other fish species, but was much lower than that of mammalian orthologs. Strikingly, the elephant shark TRPV4 was a cooling-activated channel with a threshold of 20°C, whereas, in several tetrapods, it is activated by warmth. These results suggest that the temperature sensitivity of TRPV4 has changed in vertebrate evolutionary lineages. Furthermore, we also found the elephant shark possesses heat-evoked TRPV3 with a threshold of 42°C, which is absent in more derived teleost fishes. Taken together, our findings elucidate that the vertebrate-type thermosensory system has already emerged in the common ancestor of jawed vertebrates, although their temperature-sensing ranges were different from those of mammals.
Collapse
Affiliation(s)
- Sumika Otake
- Graduate School of Life Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Shigeru Saito
- Department of Animal Bioscience, Nagahama Institute of Bio-science and Technology, Nagahama, Shiga 526-0829, Japan
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki 444-8787, Aichi, Japan
- Thermal Biology Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Aichi, Japan
| | - Xiaozhi Lin
- Graduate School of Life Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Claire T Saito
- Department of Animal Bioscience, Nagahama Institute of Bio-science and Technology, Nagahama, Shiga 526-0829, Japan
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki 444-8787, Aichi, Japan
| | - Satomi Kohno
- Department of Biological Sciences, St Cloud State University, St Cloud, MN 56301, USA
| | - Wataru Takagi
- Faculty of Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Chiba 277-8564, Japan
| | - Susumu Hyodo
- Faculty of Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Chiba 277-8564, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki 444-8787, Aichi, Japan
- Thermal Biology Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Aichi, Japan
- Thermal Biology Research Group, Nagoya Advanced Research and Development Center, Nagoya City University, Nagoya 467-8601, Aichi, Japan
| | - Yoshinao Katsu
- Graduate School of Life Science, Hokkaido University, Sapporo 060-0810, Japan
- Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| |
Collapse
|
2
|
Shibasaki K. TRPV4 activation by core body temperature has multimodal functions in the central nervous system. J Physiol Sci 2024; 74:55. [PMID: 39578735 PMCID: PMC11583650 DOI: 10.1186/s12576-024-00948-x] [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: 06/30/2024] [Accepted: 11/01/2024] [Indexed: 11/24/2024]
Abstract
Brain temperature is strictly regulated by various endogenous mechanisms and significantly contributes to brain function in homeothermic animals, making it an important factor for health. Thermosensitive transient receptor potential (TRP) channels convert temperature information into electrical signals through cation influx. In particular, TRPV4 is involved in the regulation of brain function. TRPV4, constitutively active in neurons through its activation by brain temperature, increases neuronal firing. TRPV4KO mice have electroencephalogram abnormalities, resulting in depression-like and social behavioral abnormalities. This basic function of TRPV4, as a translator of brain temperature information, has been implicated in several diseases, including epilepsy and stress-induced depression. In addition to its neuronal functions, TRPV4 has many key functions in glia and vasculature that depend on brain temperature and contribute to brain activity. In this review, I summarize the importance of TRPV4 activities in relation to brain temperature and focus on how hyperthermia-induced TRPV4 dysfunction exacerbates brain diseases.
Collapse
Affiliation(s)
- Koji Shibasaki
- Laboratory of Neurochemistry, Department of Nutrition Science, University of Nagasaki, Nagasaki, 851-2195, Japan.
| |
Collapse
|
3
|
Zhao L, Xu K, Talyzina I, Shi J, Li S, Yang Y, Zhang S, Zheng J, Sobolevsky AI, Chen H, Cui J. Human TRPV4 engineering yields an ultrasound-sensitive actuator for sonogenetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618766. [PMID: 39464052 PMCID: PMC11507911 DOI: 10.1101/2024.10.16.618766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Sonogenetics offers non-invasive and cell-type specific modulation of cells genetically engineered to express ultrasound-sensitive actuators. Finding an ion channel to serve as sonogenetic actuator it critical for advancing this promising technique. Here, we show that ultrasound can activate human TRP channel hTRPV4. By screening different hTRPV4 variants, we identify a mutation F617L that increases mechano-sensitivity of this channel to ultrasound, while reduces its sensitivity to hypo-osmolarity, elevated temperature, and agonist. This altered sensitivity profile correlates with structural differences in hTRPV4-F617L compared to wild-type channels revealed by our cryo-electron microscopy analysis. We also show that hTRPV4-F617L can serve as a sonogenetic actuator for neuromodulation in freely moving mice. Our findings demonstrate the use of structure-guided mutagenesis to engineer ion channels with tailored properties of ideal sonogenetic actuators.
Collapse
|
4
|
Hernández-Vega AM, Llorente I, Sánchez-Hernández R, Segura Y, Tusié-Luna T, Morales-Buenrostro LE, García-Villegas R, Islas LD, Rosenbaum T. Identification and Properties of TRPV4 Mutant Channels Present in Polycystic Kidney Disease Patients. FUNCTION 2024; 5:zqae031. [PMID: 38984987 PMCID: PMC11384909 DOI: 10.1093/function/zqae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024] Open
Abstract
Polycystic kidney disease (PKD), a disease characterized by the enlargement of the kidney through cystic growth is the fourth leading cause of end-stage kidney disease world-wide. Transient receptor potential Vanilloid 4 (TRPV4), a calcium-permeable TRP, channel participates in kidney cell physiology and since TRPV4 forms complexes with another channel whose malfunction is associated to PKD, TRPP2 (or PKD2), we sought to determine whether patients with PKD, exhibit previously unknown mutations in TRPV4. Here, we report the presence of mutations in the TRPV4 gene in patients diagnosed with PKD and determine that they produce gain-of-function (GOF). Mutations in the sequence of the TRPV4 gene have been associated to a broad spectrum of neuropathies and skeletal dysplasias but not PKD, and their biophysical effects on channel function have not been elucidated. We identified and examined the functional behavior of a novel E6K mutant and of the previously known S94L and A217S mutant TRVP4 channels. The A217S mutation has been associated to mixed neuropathy and/or skeletal dysplasia phenotypes, however, the PKD carriers of these variants had not been diagnosed with these reported clinical manifestations. The presence of certain mutations in TRPV4 may influence the progression and severity of PKD through GOF mechanisms. PKD patients carrying TRVP4 mutations are putatively more likely to require dialysis or renal transplant as compared to those without these mutations.
Collapse
Affiliation(s)
- Ana M Hernández-Vega
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Itzel Llorente
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Raúl Sánchez-Hernández
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Yayoi Segura
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México14080, Mexico
| | - Teresa Tusié-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México14080, Mexico
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Luis E Morales-Buenrostro
- Departmento de Nefrología y Metabolismo Mineral, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, México
| | - Refugio García-Villegas
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico
| | - León D Islas
- Departamento de Fisiología, Facultad de Medicina. Universidad Nacional Autónoma de México,Ciudad de México 04510, Mexico
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
Collapse
|
5
|
Yang F, Ma Y, Zhang A, Yao J, Jiang S, He C, Peng H, Ren G, Yang Y, Wu A. Engineering magnetic nanosystem for TRPV1 and TRPV4 channel activation. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1987. [PMID: 39136188 DOI: 10.1002/wnan.1987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 09/21/2024]
Abstract
Recently, physical tools for remotely stimulating mechanical force-sensitive and temperature-sensitive proteins to regulate intracellular pathways have opened up novel and exciting avenues for basic research and clinical applications. Among the numerous modes of physical stimulation, magnetic stimulation is significantly attractive for biological applications due to the advantages of depth penetration and spatial-temporally controlled transduction. Herein, the physicochemical parameters (e.g., shape, size, composition) that influence the magnetic properties of magnetic nanosystems as well as the characteristics of transient receptor potential vanilloid-1 (TRPV1) and transient receptor potential vanilloid-4 (TRPV4) channels are systematically summarized, which offer opportunities for magnetic manipulation of cell fate in a precise and effective manner. In addition, representative regulatory applications involving magnetic nanosystem-based TRPV1 and TRPV4 channel activation are highlighted, both at the cellular level and in animal models. Furthermore, perspectives on the further development of this magnetic stimulation mode are commented on, with emphasis on scientific limitations and possible directions for exploitation. This article is categorized under: Diagnostic Tools > Biosensing Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Fang Yang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Yaqi Ma
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Aoran Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Junlie Yao
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shaohua Jiang
- International Innovation Center for Forest Chemicals and Materials, College of Materials Science and Engineering, Nanjing Forestry University, Nanjing, China
| | - Chenglong He
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Hao Peng
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Guiping Ren
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiqian Yang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Aiguo Wu
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
6
|
Hermanova Z, Valihrach L, Kriska J, Maheta M, Tureckova J, Kubista M, Anderova M. The deletion of AQP4 and TRPV4 affects astrocyte swelling/volume recovery in response to ischemia-mimicking pathologies. Front Cell Neurosci 2024; 18:1393751. [PMID: 38818517 PMCID: PMC11138210 DOI: 10.3389/fncel.2024.1393751] [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: 02/29/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024] Open
Abstract
Introduction Astrocytic Transient receptor potential vanilloid 4 (TRPV4) channels, together with Aquaporin 4 (AQP4), are suspected to be the key players in cellular volume regulation, and therefore may affect the development and severity of cerebral edema during ischemia. In this study, we examined astrocytic swelling/volume recovery in mice with TRPV4 and/or AQP4 deletion in response to in vitro ischemic conditions, to determine how the deletion of these channels can affect the development of cerebral edema. Methods We used three models of ischemia-related pathological conditions: hypoosmotic stress, hyperkalemia, and oxygenglucose deprivation (OGD), and observed their effect on astrocyte volume changes in acute brain slices of Aqp4-/-, Trpv4-/- and double knockouts. In addition, we employed single-cell RT-qPCR to assess the effect of TRPV4 and AQP4 deletion on the expression of other ion channels and transporters involved in the homeostatic functioning of astrocytes. Results Quantification of astrocyte volume changes during OGD revealed that the deletion of AQP4 reduces astrocyte swelling, while simultaneous deletion of both AQP4 and TRPV4 leads to a disruption of astrocyte volume recovery during the subsequent washout. Of note, astrocyte exposure to hypoosmotic stress or hyperkalemia revealed no differences in astrocyte swelling in the absence of AQP4, TRPV4, or both channels. Moreover, under ischemia-mimicking conditions, we identified two distinct subpopulations of astrocytes with low and high volumetric responses (LRA and HRA), and their analyses revealed that mainly HRA are affected by the deletion of AQP4, TRPV4, or both channels. Furthermore, gene expression analysis revealed reduced expression of the ion transporters KCC1 and ClC2 as well as the receptors GABAB and NMDA in Trpv4-/- mice. The deletion of AQP4 instead caused reduced expression of the serine/cysteine peptidase inhibitor Serpina3n. Discussion Thus, we showed that in AQP4 or TRPV4 knockouts, not only the specific function of these channels is affected, but also the expression of other proteins, which may modulate the ischemic cascade and thus influence the final impact of ischemia.
Collapse
Affiliation(s)
- Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
- Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Lukas Valihrach
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| | - Mansi Maheta
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| |
Collapse
|
7
|
Chen J, Gao Y, Liu N, Hai D, Wei W, Liu Y, Lan X, Jin X, Yu J, Ma L. Mechanism of NLRP3 Inflammasome in Epilepsy and Related Therapeutic Agents. Neuroscience 2024; 546:157-177. [PMID: 38574797 DOI: 10.1016/j.neuroscience.2024.03.029] [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: 12/31/2023] [Revised: 03/05/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024]
Abstract
Epilepsy is one of the most widespread and complex diseases in the central nervous system (CNS), affecting approximately 65 million people globally, an important factor resulting in neurological disability-adjusted life year (DALY) and progressive cognitive dysfunction. Medication is the most essential treatment. The currently used drugs have shown drug resistance in some patients and only control symptoms; the development of novel and more efficacious pharmacotherapy is imminent. Increasing evidence suggests neuroinflammation is involved in the occurrence and development of epilepsy, and high expression of NLRP3 inflammasome has been observed in the temporal lobe epilepsy (TLE) brain tissue of patients and animal models. The inflammasome is a crucial cause of neuroinflammation by activating IL-1β and IL-18. Many preclinical studies have confirmed that regulating NLRP3 inflammasome pathway can prevent the development of epilepsy, reduce the severity of epilepsy, and play a neuroprotective role. Therefore, regulating NLRP3 inflammasome could be a potential target for epilepsy treatment. In summary, this review describes the priming and activation of inflammasome and its biological function in the progression of epilepsy. In addition, we reviewes the current pharmacological researches for epilepsy based on the regulation of NLRP3 inflammasome, aiming to provide a basis and reference for developing novel antiepileptic drugs.
Collapse
Affiliation(s)
- Juan Chen
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Yuan Gao
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Ning Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Dongmei Hai
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Wei Wei
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Yue Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Xiaobing Lan
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Xueqin Jin
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China.
| | - Jianqiang Yu
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China.
| | - Lin Ma
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
8
|
Tytti K, Sanna K, Carla G, Jonatan P, Kaisa R, Sari T. Mechanosensitive TRPV4 channel guides maturation and organization of the bilayered mammary epithelium. Sci Rep 2024; 14:6774. [PMID: 38514727 PMCID: PMC10957991 DOI: 10.1038/s41598-024-57346-x] [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: 07/03/2023] [Accepted: 03/18/2024] [Indexed: 03/23/2024] Open
Abstract
Biophysical cues from the cell microenvironment are detected by mechanosensitive components at the cell surface. Such machineries convert physical information into biochemical signaling cascades within cells, subsequently leading to various cellular responses in a stimulus-dependent manner. At the surface of extracellular environment and cell cytoplasm exist several ion channel families that are activated by mechanical signals to direct intracellular events. One of such channel is formed by transient receptor potential cation channel subfamily V member, TRPV4 that is known to act as a mechanosensor in wide variaty of tissues and control ion-influx in a spatio-temporal way. Here we report that TRPV4 is prominently expressed in the stem/progenitor cell populations of the mammary epithelium and seems important for the lineage-specific differentiation, consequently affecting mechanical features of the mature mammary epithelium. This was evident by the lack of several markers for mature myoepithelial and luminal epithelial cells in TRPV4-depleted cell lines. Interestingly, TRPV4 expression is controlled in a tension-dependent manner and it also impacts differentation process dependently on the stiffness of the microenvironment. Furthermore, such cells in a 3D compartment were disabled to maintain normal mammosphere structures and displayed abnormal lumen formation, size of the structures and disrupted cellular junctions. Mechanosensitive TRPV4 channel therefore act as critical player in the homeostasis of normal mammary epithelium through sensing the physical environment and guiding accordingly differentiation and structural organization of the bilayered mammary epithelium.
Collapse
Affiliation(s)
- Kärki Tytti
- Department of Applied Physics, School of Science, Aalto University, Espoo, Finland
| | - Koskimäki Sanna
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Guenther Carla
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Pirhonen Jonatan
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Rajakylä Kaisa
- School of Social Services and Health Care, Tampere University of Applied Sciences, Tampere, Finland
| | - Tojkander Sari
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland.
| |
Collapse
|
9
|
Shibasaki K. Regulation of Neural Functions by Brain Temperature and Thermo-TRP Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1461:199-211. [PMID: 39289283 DOI: 10.1007/978-981-97-4584-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Body temperature is an important determinant in regulating the activities of animals. In humans, a mild 0.5 °C hyperthermia can cause headaches, demonstrating that the maintenance of normal body temperature is a key for our health. In a more extreme example, accidental acute hypothermia can lead to severe shivering, loss of consciousness, or death, although the details of these mechanisms are poorly understood. We previously found that the TRPV4 ion channel is constitutively activated by normal body temperature. The activation threshold of TRPV4 is >34 °C in the brain, which enables TRPV4 to convert thermal information into cellular signaling. Here we review the data that describe how the deletion of TRPV4 evokes abnormal behavior in mice. These studies demonstrate that the maintenance of body temperature and the sensory system for detecting body temperature, such as via TRPV4, are critical components for normal cellular function. Moreover, abnormal TRPV4 activation exacerbates cell death, epilepsy, stroke, or brain edema. Notably, TRPV4 can detect mechanical stimuli and contributes to various neural functions similar to the mechanosensitive characteristics of TRPV2. In this review, I summarize the findings related to TRPV2/TRPV4 and neural functions.
Collapse
Affiliation(s)
- Koji Shibasaki
- Laboratory of Neurochemistry, Department of Nutrition Science, University of Nagasaki, Nagasaki, Japan.
| |
Collapse
|
10
|
Matsumoto T, Taguchi K, Kobayashi T. Role of TRPV4 on vascular tone regulation in pathophysiological states. Eur J Pharmacol 2023; 959:176104. [PMID: 37802278 DOI: 10.1016/j.ejphar.2023.176104] [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: 08/18/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023]
Abstract
Vascular tone regulation is a key event in controlling blood flow in the body. Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) help regulate the vascular tone. Abnormal vascular responsiveness to various stimuli, including constrictors and dilators, has been observed in pathophysiological states although EC and VSMC coordinate to maintain the exquisite balance between contraction and relaxation in vasculatures. Thus, investigating the mechanisms underlying vascular tone abnormality is very important in maintaining vascular health and treating vasculopathy. Increased intracellular free Ca2+ concentration ([Ca2+]i) is one of the major triggers initiating each EC and VSMC response. Transient receptor potential vanilloid family member 4 (TRPV4) is a Ca2+-permeable non-selective ion channel, which is activated by several stimuli, and is presented in both ECs and VSMCs. Therefore, TRPV4 plays an important role in vascular responses. Emerging evidence indicates the role of TRPV4 on the functions of ECs and VSMCs in various pathophysiological states, including hypertension, diabetes, and obesity. This review focused on the link between TRPV4 and the functions of ECs/VSMCs, particularly its role in vascular tone and responsiveness to vasoactive substances.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Pharmaceutical Education and Research, Pharmaceutical Education and Research Center, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan.
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| |
Collapse
|
11
|
Taivanbat B, Yamazaki S, Nasanbat B, Uchiyama A, Amalia SN, Nasan-Ochir M, Inoue Y, Ishikawa M, Kosaka K, Sekiguchi A, Ogino S, Yokoyama Y, Torii R, Hosoi M, Shibasaki K, Motegi SI. Transient receptor potential vanilloid 4 promotes cutaneous wound healing by regulating keratinocytes and fibroblasts migration and collagen production in fibroblasts in a mouse model. J Dermatol Sci 2023; 112:54-62. [PMID: 37839930 DOI: 10.1016/j.jdermsci.2023.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 08/29/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Transient receptor potential vanilloid 4 (TRPV4), a cation ion channel, is expressed in different cells, and it regulates the development of different diseases. We recently found a high TRPV4 expression in the wounded skin area. However, the role of TRPV4 in cutaneous wound healing is unknown. OBJECTIVE To investigate the role of TRPV4 in cutaneous wound healing in a mouse model. METHODS Skin wound healing experiment and histopathological studies were performed between WT and TRPV4 KO mice. The effect of TRPV4 antagonist and agonist on cell migration, proliferation, and differentiation were examined in vitro. RESULTS TRPV4 expression was enhanced in wounded area in the skin. TRPV4 KO mice had impaired cutaneous wound healing compared with the WT mice. Further, they had significantly suppressed re-epithelialization and formation of granulation tissue, amount of collagen deposition, and number of α-SMA-positive myofibroblasts in skin wounds. qPCR revealed that the KO mice had decreased mRNA expression of COL1A1 and ACTA2 in skin wounds. In vitro, treatment with selective TRPV4 antagonist suppressed migrating capacity, scratch stimulation enhanced the expression of phospho-ERK in keratinocytes, and TGF-β stimulation enhanced the mRNA expression of COL1A1 and ACTA2 in fibroblasts. Selective TRPV4 agonist suppressed cell migration in keratinocytes, and did not enhance proliferation and migration, but promoted differentiation in fibroblasts. CONCLUSION TRPV4 mediates keratinocytes and fibroblasts migration and increases collagen deposition in the wound area, thereby promoting cutaneous wound healing.
Collapse
Affiliation(s)
- Bayarmaa Taivanbat
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Sahori Yamazaki
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Bolor Nasanbat
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akihiko Uchiyama
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan.
| | - Syahla Nisaa Amalia
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | | | - Yuta Inoue
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Mai Ishikawa
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Keiji Kosaka
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akiko Sekiguchi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Sachiko Ogino
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoko Yokoyama
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ryoko Torii
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Mari Hosoi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Koji Shibasaki
- Laboratory of Neurochemistry, Department of Nutrition Science, University of Nagasaki, Nagasaki, Japan
| | - Sei-Ichiro Motegi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
12
|
Li M, Zheng J, Wu T, He Y, Guo J, Xu J, Gao C, Qu S, Zhang Q, Zhao J, Cheng W. Activation of TRPV4 Induces Exocytosis and Ferroptosis in Human Melanoma Cells. Int J Mol Sci 2022; 23:ijms23084146. [PMID: 35456964 PMCID: PMC9030060 DOI: 10.3390/ijms23084146] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 12/20/2022] Open
Abstract
TRPV4 (transient receptor potential vanilloid 4), a calcium permeable TRP ion channel, is known to play a key role in endocytosis. However, whether it contributes to exocytosis remains unclear. Here, we report that activation of TRPV4 induced massive exocytosis in both melanoma A375 cell and heterologous expression systems. We show here that, upon application of TRPV4-specific agonists, prominent vesicle priming from endoplasmic reticulum (ER) was observed, followed by morphological changes of mitochondrial crista may lead to cell ferroptosis. We further identified interactions between TRPV4 and folding/vesicle trafficking proteins, which were triggered by calcium entry through activated TRPV4. This interplay, in turn, enhanced TRPV4-mediated activation of folding and vesicle trafficking proteins to promote exocytosis. Our study revealed a signaling mechanism underlying stimulus-triggered exocytosis in melanoma and highlighted the role of cellular sensor TRPV4 ion channel in mediating ferroptosis.
Collapse
|
13
|
Uchida Y, Izumizaki M. Effect of menstrual cycle and female hormones on TRP and TREK channels in modifying thermosensitivity and physiological functions in women. J Therm Biol 2021; 100:103029. [PMID: 34503776 DOI: 10.1016/j.jtherbio.2021.103029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
Thermoregulation is crucial for human survival at various ambient temperatures. Transient receptor potential (TRP) and TWIK-related K+ (TREK) channels expressed in sensory neurons play a role in peripheral thermosensitivity for temperature detection. In addition, these channels have various physiological roles in the skeletal, nervous, immune, vascular, digestive, and urinary systems. In women, the female hormones estradiol (E2) and progesterone (P4), which fluctuate during the menstrual cycle, affect various physiological functions, such as thermoregulation in hot and cold environments. The present review describes the effect of female hormones on TRP and TREK channels and related physiological functions. The P4 decreased thermosensitivity via TRPV1. E2 facilitates temporomandibular joint disease (TRPV1), breast cancer (TRPM8), and calcium absorption in the digestive system (TRPV5 and TRPV6), inhibits the facilitation of vasoconstriction (TRPM3), nerve inflammation (TRPM4), sweetness sensitivity (TRPM5), and menstrual disorders (TRPC1), and prevents insulin resistance (TRPC5) via each channel. P4 inhibits vasoconstriction (TRPM3), sweetness sensitivity (TRPM5), ciliary motility in the lungs (TRPV4), menstrual disorder (TRPC1), and immunity (TRPC3), and facilitates breast cancer (TRPV6) via each channel as indicated. The effects of female hormones on TREK channels and physiological functions are still under investigation. In summary, female hormones influence physiological functions via some TRP channels; however, the literature is not comprehensive and future studies are needed, especially those related to thermoregulation in women.
Collapse
Affiliation(s)
- Yuki Uchida
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan.
| | - Masahiko Izumizaki
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Kärki T, Tojkander S. TRPV Protein Family-From Mechanosensing to Cancer Invasion. Biomolecules 2021; 11:1019. [PMID: 34356643 PMCID: PMC8301805 DOI: 10.3390/biom11071019] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 02/08/2023] Open
Abstract
Biophysical cues from the cellular microenvironment are detected by mechanosensitive machineries that translate physical signals into biochemical signaling cascades. At the crossroads of extracellular space and cell interior are located several ion channel families, including TRP family proteins, that are triggered by mechanical stimuli and drive intracellular signaling pathways through spatio-temporally controlled Ca2+-influx. Mechanosensitive Ca2+-channels, therefore, act as critical components in the rapid transmission of physical signals into biologically compatible information to impact crucial processes during development, morphogenesis and regeneration. Given the mechanosensitive nature of many of the TRP family channels, they must also respond to the biophysical changes along the development of several pathophysiological conditions and have also been linked to cancer progression. In this review, we will focus on the TRPV, vanilloid family of TRP proteins, and their connection to cancer progression through their mechanosensitive nature.
Collapse
Affiliation(s)
- Tytti Kärki
- Department of Applied Physics, School of Science, Aalto University, 00076 Espoo, Finland;
| | - Sari Tojkander
- Department of Veterinary Biosciences, Section of Pathology, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
15
|
Malik M, Roh M, England SK. Uterine contractions in rodent models and humans. Acta Physiol (Oxf) 2021; 231:e13607. [PMID: 33337577 PMCID: PMC8047897 DOI: 10.1111/apha.13607] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022]
Abstract
Aberrant uterine contractions can lead to preterm birth and other labour complications and are a significant cause of maternal morbidity and mortality. To investigate the mechanisms underlying dysfunctional uterine contractions, researchers have used experimentally tractable small animal models. However, biological differences between humans and rodents change how researchers select their animal model and interpret their results. Here, we provide a general review of studies of uterine excitation and contractions in mice, rats, guinea pigs, and humans, in an effort to introduce new researchers to the field and help in the design and interpretation of experiments in rodent models.
Collapse
Affiliation(s)
- Manasi Malik
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMOUSA
| | - Michelle Roh
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMOUSA
| | - Sarah K. England
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
16
|
Wang S, He H, Long J, Sui X, Yang J, Lin G, Wang Q, Wang Y, Luo Y. TRPV4 Regulates Soman-Induced Status Epilepticus and Secondary Brain Injury via NMDA Receptor and NLRP3 Inflammasome. Neurosci Bull 2021; 37:905-920. [PMID: 33761112 DOI: 10.1007/s12264-021-00662-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 12/06/2020] [Indexed: 01/12/2023] Open
Abstract
Nerve agents are used in civil wars and terrorist attacks, posing a threat to public safety. Acute exposure to nerve agents such as soman (GD) causes serious brain damage, leading to death due to intense seizures induced by acetylcholinesterase inhibition and neuronal injury resulting from increased excitatory amino-acid levels and neuroinflammation. However, data on the anticonvulsant and neuroprotective efficacies of currently-used countermeasures are limited. Here, we evaluated the potential effects of transient receptor vanilloid 4 (TRPV4) in the treatment of soman-induced status epilepticus (SE) and secondary brain injury. We demonstrated that TRPV4 expression was markedly up-regulated in rat hippocampus after soman-induced seizures. Administration of the TRPV4 antagonist GSK2193874 prior to soman exposure significantly decreased the mortality rate in rats and reduced SE intensity. TRPV4-knockout mice also showed lower incidence of seizures and higher survival rates than wild-type mice following soman exposure. Further in vivo and in vitro experiments demonstrated that blocking TRPV4 prevented NMDA receptor-mediated glutamate excitotoxicity. The protein levels of the NLRP3 inflammasome complex and its downstream cytokines IL-1β and IL-18 increased in soman-exposed rat hippocampus. However, TRPV4 inhibition or deletion markedly reversed the activation of the NLRP3 inflammasome pathway. In conclusion, our study suggests that the blockade of TRPV4 protects against soman exposure and reduces brain injury following SE by decreasing NMDA receptor-mediated excitotoxicity and NLRP3-mediated neuroinflammation. To our knowledge, this is the first study regarding the "dual-switch" function of TRPV4 in the treatment of soman intoxication.
Collapse
Affiliation(s)
- Shuai Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Huanhuan He
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Jianhai Long
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Xin Sui
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Jun Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Guodong Lin
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Qian Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| |
Collapse
|
17
|
The Role of Thermosensitive Ion Channels in Mammalian Thermoregulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:355-370. [DOI: 10.1007/978-981-16-4254-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
18
|
Dupont C, Novak K, Denman K, Myers JH, Sullivan JM, Walker PV, Brown NL, Ladle DR, Bogdanik L, Lutz CM, A Voss A, Sumner CJ, Rich MM. TRPV4 Antagonism Prevents Mechanically Induced Myotonia. Ann Neurol 2020; 88:297-308. [PMID: 32418267 PMCID: PMC7657963 DOI: 10.1002/ana.25780] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/27/2020] [Accepted: 05/07/2020] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Myotonia is caused by involuntary firing of skeletal muscle action potentials and causes debilitating stiffness. Current treatments are insufficiently efficacious and associated with side effects. Myotonia can be triggered by voluntary movement (electrically induced myotonia) or percussion (mechanically induced myotonia). Whether distinct molecular mechanisms underlie these triggers is unknown. Our goal was to identify ion channels involved in mechanically induced myotonia and to evaluate block of the channels involved as a novel approach to therapy. METHODS We developed a novel system to enable study of mechanically induced myotonia using both genetic and pharmacologic mouse models of myotonia congenita. We extended ex vivo studies of excitability to in vivo studies of muscle stiffness. RESULTS As previous work suggests activation of transient receptor potential vanilloid 4 (TRPV4) channels by mechanical stimuli in muscle, we examined the role of this cation channel. Mechanically induced myotonia was markedly suppressed in TRPV4-null muscles and in muscles treated with TRPV4 small molecule antagonists. The suppression of mechanically induced myotonia occurred without altering intrinsic muscle excitability, such that myotonia triggered by firing of action potentials (electrically induced myotonia) was unaffected. When injected intraperitoneally, TRPV4 antagonists lessened the severity of myotonia in vivo by approximately 80%. INTERPRETATION These data demonstrate that there are distinct molecular mechanisms triggering electrically induced and mechanically induced myotonia. Our data indicates that activation of TRPV4 during muscle contraction plays an important role in triggering myotonia in vivo. Elimination of mechanically induced myotonia by TRPV4 inhibition offers a new approach to treating myotonia. ANN NEUROL 2020;88:297-308.
Collapse
Affiliation(s)
- Chris Dupont
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - Kevin Novak
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - Kirsten Denman
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - Jessica H Myers
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - Jeremy M Sullivan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Phillip V Walker
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - Nicklaus L Brown
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - David R Ladle
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | | | | | - Andrew A Voss
- Department of Biology, Wright State University, Dayton, OH, USA
| | - Charlotte J Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark M Rich
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| |
Collapse
|
19
|
TRPV4 promotes acoustic wave-mediated BBB opening via Ca 2+/PKC-δ pathway. J Adv Res 2020; 26:15-28. [PMID: 33133680 PMCID: PMC7584681 DOI: 10.1016/j.jare.2020.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 05/14/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction Numerous studies have shown the ability of low-energy acoustic waves such as focused ultrasound or shockwave to transiently open blood-brain barrier (BBB) and facilitate drug delivery to the brain. Preclinical and clinical evidences have well demonstrated the efficacy and safety in treating various brain disorders. However, the molecular mechanisms of acoustic waves on the BBB are still not fully understood. Objectives The present study aimed at exploring the possible molecular mechanisms of acoustic wave stimulation on brains. Methods: Briefly describe the experimental design The left hemisphere of the rat‘s brain was treated with pulsed ultrasound from a commercial focused shockwave or a planar ultrasound device, and the right hemisphere served as a control. One hour after the mechanical wave stimulation or overnight, the rats were sacrificed and the brains were harvested for protein or histological analysis. Agonists and antagonists related to the signal transduction pathways of tight junction proteins were used to investigate the possible intracellular mechanisms. Results Intracellular signal transduction analysis shows calcium influx through transient receptor potential vanilloid 4 (TRPV4) channels, and the activation of PKC-δ pathway to mediate dissociation of ZO-1 and occludin after acoustic wave stimulation. The activation of TRPV4 or PKC-δ signaling further increased the expression level of TRPV4, suggesting a feedback loop to regulate BBB permeability. Moreover, the tight junction proteins dissociation can be reversed by administration of PKC-δ inhibitor and TRPV4 antagonist. Conclusion The present study shows the crucial role of TRPV4 in acoustic wave-mediated BBB permeability, specifically its effect on compromising tight junction proteins, ZO-1 and occludin. Our findings provide a new molecular perspective to explain acoustic wave-mediated BBB opening. Moreover, activation of TRPV4 by agonists may reduce the threshold intensity level of acoustic waves for BBB opening, which may prevent undesirable mechanical damages while maintaining efficient BBB opening.
Collapse
|
20
|
Meng C, Xia Q, Wu H, Huang H, Liu H, Li Y, Zhang F, Song W. Photobiomodulation with 630-nm LED radiation inhibits the proliferation of human synoviocyte MH7A cells possibly via TRPV4/PI3K/AKT/mTOR signaling pathway. Lasers Med Sci 2020; 35:1927-1936. [PMID: 32162133 DOI: 10.1007/s10103-020-02977-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/31/2020] [Indexed: 01/19/2023]
Abstract
Phototherapy has been used to treat postoperative pain and inflammatory response in rheumatoid arthritis. Confidence in this approach, however, is impaired by lack of understanding of the light-triggered cellular and molecular mechanisms. The purpose of this study was to characterize the response of human synoviocyte MH7A cells to visible LED red light in an attempt to elucidate the associated action mechanism. Human synoviocyte MH7A cells were treated with 630-nm LED light after stimulation of tumor necrosis factor-α (TNF-α). The effects of light radiation on cell proliferation and migration were detected by MTT assay and scratch test. The expressions of inflammatory cytokines were measured using RT-qPCR. This was followed by detection of the levels of extracellular proteins IL-6 and IL-8 after differential radiation. Furthermore, the expression levels and activation of proteins on PI3K/AKT/mTOR signaling pathway were examined with Western blot. In terms of the proliferation and migration, repeated radiation with LED red light (630 nm, 26 and 39 J/cm2) exerted an inhibitory effect on synoviocyte MH7A cells. Expression of inflammatory factors (IL-6, IL-1β, IL-8, and MMP-3) was reduced; meanwhile, the expression of anti-inflammatory factor IL-10 was promoted. At the protein level, treatment with 39 J/cm2 of LED red light could decrease the level of extracellular protein (IL-6 and IL-8) and affect the expression and phosphorylation of proteins on TRPV4/PI3K/AKT/mTOR signaling pathway induced by TNF-α. These results demonstrated that LED red light (630 nm) inhibits proliferation and migration of MH7A cells. The growth-inhibiting effects of LED red light on human synoviocyte MH7A cells appear to be associated with regulation of the TRPV4/PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Caiyun Meng
- Department of Microbiology, Wu Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Qing Xia
- Department of Microbiology, Wu Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Hao Wu
- Department of Microbiology, Wu Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - He Huang
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hailiang Liu
- Department of Microbiology, the Heilongjiang Key Laboratory of Immunity and Infection, Harbin Medical University, Harbin, China
| | - Yujun Li
- Department of Microbiology, the Heilongjiang Key Laboratory of Immunity and Infection, Harbin Medical University, Harbin, China
| | - Fengmin Zhang
- Department of Microbiology, the Heilongjiang Key Laboratory of Immunity and Infection, Harbin Medical University, Harbin, China
| | - Wuqi Song
- Department of Microbiology, the Heilongjiang Key Laboratory of Immunity and Infection, Harbin Medical University, Harbin, China.
| |
Collapse
|
21
|
TRPV4 activation by thermal and mechanical stimuli in disease progression. J Transl Med 2020; 100:218-223. [PMID: 31896814 DOI: 10.1038/s41374-019-0362-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/04/2019] [Accepted: 12/15/2019] [Indexed: 12/17/2022] Open
Abstract
Body temperature is an important determinant in regulating the activities of animals. In humans, a mild 0.5 °C hyperthermia can cause headaches, demonstrating that the maintenance of normal body temperature is a key for our health. In a more extreme example, accidental acute hypothermia can lead to severe shivering, loss of consciousness, or death, although the details of these mechanisms are poorly understood. We previously found that the TRPV4 ion channel is constitutively activated by normal body temperature. The activation threshold of TRPV4 is >34 °C in the brain, which enables TRPV4 to convert thermal information into cellular signaling. Here we review the data which describe how the deletion of TRPV4 evokes abnormal behavior in mice. These studies demonstrate that the maintenance of body temperature and the sensory system for detecting body temperature, such as via TRPV4, are critical components for normal cellular function. Moreover, abnormal TRPV4 activation exacerbates cell death, epilepsy, stroke, brain edema, or cardiac fibroblast activity. In this review, we also summarize the findings related to TRPV4 and disease.
Collapse
|
22
|
Negri S, Faris P, Berra-Romani R, Guerra G, Moccia F. Endothelial Transient Receptor Potential Channels and Vascular Remodeling: Extracellular Ca 2 + Entry for Angiogenesis, Arteriogenesis and Vasculogenesis. Front Physiol 2020; 10:1618. [PMID: 32038296 PMCID: PMC6985578 DOI: 10.3389/fphys.2019.01618] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022] Open
Abstract
Vasculogenesis, angiogenesis and arteriogenesis represent three crucial mechanisms involved in the formation and maintenance of the vascular network in embryonal and post-natal life. It has long been known that endothelial Ca2+ signals are key players in vascular remodeling; indeed, multiple pro-angiogenic factors, including vascular endothelial growth factor, regulate endothelial cell fate through an increase in intracellular Ca2+ concentration. Transient Receptor Potential (TRP) channel consist in a superfamily of non-selective cation channels that are widely expressed within vascular endothelial cells. In addition, TRP channels are present in the two main endothelial progenitor cell (EPC) populations, i.e., myeloid angiogenic cells (MACs) and endothelial colony forming cells (ECFCs). TRP channels are polymodal channels that can assemble in homo- and heteromeric complexes and may be sensitive to both pro-angiogenic cues and subtle changes in local microenvironment. These features render TRP channels the most versatile Ca2+ entry pathway in vascular endothelial cells and in EPCs. Herein, we describe how endothelial TRP channels stimulate vascular remodeling by promoting angiogenesis, arteriogenesis and vasculogenesis through the integration of multiple environmental, e.g., extracellular growth factors and chemokines, and intracellular, e.g., reactive oxygen species, a decrease in Mg2+ levels, or hypercholesterolemia, stimuli. In addition, we illustrate how endothelial TRP channels induce neovascularization in response to synthetic agonists and small molecule drugs. We focus the attention on TRPC1, TRPC3, TRPC4, TRPC5, TRPC6, TRPV1, TRPV4, TRPM2, TRPM4, TRPM7, TRPA1, that were shown to be involved in angiogenesis, arteriogenesis and vasculogenesis. Finally, we discuss the role of endothelial TRP channels in aberrant tumor vascularization by focusing on TRPC1, TRPC3, TRPV2, TRPV4, TRPM8, and TRPA1. These observations suggest that endothelial TRP channels represent potential therapeutic targets in multiple disorders featured by abnormal vascularization, including cancer, ischemic disorders, retinal degeneration and neurodegeneration.
Collapse
Affiliation(s)
- Sharon Negri
- Laboratory of General Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Pawan Faris
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Roberto Berra-Romani
- Department of Biomedicine, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Germano Guerra
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
23
|
Tsushima H, Mori-Kawabe M. Central Transient Receptor Potential Vanilloid 4 Contributes to Systemic Water Homeostasis through Urinary Excretion. Biol Pharm Bull 2019; 42:1877-1882. [DOI: 10.1248/bpb.b19-00409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hiromi Tsushima
- Laboratory of Pharmacology, College of Pharmacy, Kinjo Gakuin University
| | - Mayumi Mori-Kawabe
- Department of Cellular and Molecular Pharmacology, Nagoya City University Graduate School of Medical Sciences
| |
Collapse
|
24
|
Andersson KE. TRP Channels as Lower Urinary Tract Sensory Targets. Med Sci (Basel) 2019; 7:E67. [PMID: 31121962 PMCID: PMC6572419 DOI: 10.3390/medsci7050067] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/19/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022] Open
Abstract
Several members of the transient receptor potential (TRP) superfamily, including TRPV1, TRPV2, TRPV4, TRM4, TRPM8 and TRPA1, are expressed in the lower urinary tract (LUT), not only in neuronal fibers innervating the bladder and urethra, but also in the urothelial and muscular layers of the bladder and urethral walls. In the LUT, TRP channels are mainly involved in nociception and mechanosensory transduction. Animal studies have suggested the therapeutic potential of several TRP channels for the treatment of both bladder over- and underactivity and bladder pain disorders,; however translation of this finding to clinical application has been slow and the involvement of these channels in normal human bladder function, and in various pathologic states have not been established. The development of selective TRP channel agonists and antagonists is ongoing and the use of such agents can be expected to offer new and important information concerning both normal physiological functions and possible therapeutic applications.
Collapse
Affiliation(s)
- Karl-Erik Andersson
- Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA.
- Institute of Laboratory Medicine, Lund University, 223 62 Lund, Sweden.
| |
Collapse
|
25
|
Sun XY, Sun LL, Qi H, Gao Q, Wang GX, Wei NN, Wang K. Antipruritic Effect of Natural Coumarin Osthole through Selective Inhibition of Thermosensitive TRPV3 Channel in the Skin. Mol Pharmacol 2018; 94:1164-1173. [PMID: 30108138 DOI: 10.1124/mol.118.112466] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/16/2018] [Indexed: 12/14/2022] Open
Abstract
Coumarin osthole is a dominant bioactive ingredient of the natural Cnidium monnieri plant commonly used for traditional Chinese herbal medicines for therapies and treatments including antipruritus and antidermatitis. However, the molecular mechanism underlying the action of osthole remains unclear. In this study, we report that osthole exerts an antipruritic effect through selective inhibition of Ca2+-permeable and thermosensitive transient receptor potential vanilloid 3 (TRPV3) cation channels that are primarily expressed in the keratinocytes of the skin. Coumarin osthole was identified as an inhibitor of TRPV3 channels transiently expressed in HEK293 cells in a calcium fluorescent assay. Inhibition of the TRPV3 current by osthole and its selectivity were further confirmed by whole-cell patch clamp recordings of TRPV3-expressing HEK293 cells and mouse primary cultured keratinocytes. Behavioral evaluation demonstrated that inhibition of TRPV3 by osthole or silencing by knockout of the TRPV3 gene significantly reduced the scratching induced by either acetone-ether-water or histamine in localized rostral neck skin in mice. Taken together, our findings provide a molecular basis for use of natural coumarin osthole from the C. monnieri plant in antipruritic or skin care therapy, thus establishing a significant role of the TRPV3 channel in chronic itch signaling or acute histamine-dependent itch sensation.
Collapse
Affiliation(s)
- Xiao-Ying Sun
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Li-Lan Sun
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Hang Qi
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Qin Gao
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Gong-Xin Wang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Ning-Ning Wei
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - KeWei Wang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| |
Collapse
|
26
|
Retinal Detachment-Induced Müller Glial Cell Swelling Activates TRPV4 Ion Channels and Triggers Photoreceptor Death at Body Temperature. J Neurosci 2018; 38:8745-8758. [PMID: 30143574 PMCID: PMC6181316 DOI: 10.1523/jneurosci.0897-18.2018] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 08/21/2018] [Accepted: 08/21/2018] [Indexed: 12/24/2022] Open
Abstract
Using region-specific injection of hyaluronic acid, we developed a mouse model of acute retinal detachment (RD) to investigate molecular mechanisms of photoreceptor cell death triggered by RD. We focused on the transient receptor potential vanilloid 4 (TRPV4) ion channel, which functions as a thermosensor, osmosensor, and/or mechanosensor. After RD, the number of apoptotic photoreceptors was reduced by ∼50% in TRPV4KO mice relative to wild-type mice, indicating the possible involvement of TRPV4 activation in RD-induced photoreceptor cell death. Furthermore, TRPV4 expressed in Müller glial cells can be activated by mechanical stimuli caused by RD-induced swelling of these cells, resulting in release of the cytokine MCP-1, which is reported as a mediator of Müller glia-derived strong mediator for RD-induced photoreceptor death. We also found that the TRPV4 activation by the Müller glial swelling was potentiated by body temperature. Together, our results suggest that RD adversely impacts photoreceptor viability via TRPV4-dependent cytokine release from Müller glial cells and that TRPV4 is part of a novel molecular pathway that could exacerbate the effects of hypoxia on photoreceptor survival after RD. SIGNIFICANCE STATEMENT Identification of the mechanisms of photoreceptor death in retinal detachment is required for establishment of therapeutic targets for preventing loss of visual acuity. In this study, we found that TRPV4 expressed in Müller glial cells can be activated by mechanical stimuli caused by RD-induced swelling of these cells, resulting in release of the cytokine MCP-1, which is reported as a mediator of Müller glia-derived strong mediator for RD-induced photoreceptor death. We also found that the TRPV4 activation by the Müller glial swelling was potentiated by body temperature. Hence, TRPV4 inhibition could suppress cell death in RD pathological conditions and suggests that TRPV4 in Müller glial cells might be a novel therapeutic target for preventing photoreceptor cell death after RD.
Collapse
|
27
|
Li P, Zhang H, Wang D, Tao Y, Zhang L, Zhang W, Wang X. An efficient nonlinear hybridization chain reaction-based sensitive fluorescent assay for in situ estimation of calcium channel protein expression on bone marrow cells. Anal Chim Acta 2018; 1041:25-32. [PMID: 30340687 DOI: 10.1016/j.aca.2018.08.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 02/03/2023]
Abstract
A sensitive and highly efficient approach to monitor the expression of proteins on live cells was urgently needed to demonstrate its factor and mechanism and most important for clinical diagnostics and molecular biology. Herein, we developed a simple and highly efficient strategy, nonlinear hybridization chain reaction (nonlinear HCR), for the sensitive determination of proteins on live cells with transient receptor potential vanilloid 4 (TRPV4) and RAW264.7 cells as a model. Unlike the normal hybridization chain reaction (HCR) with multiplicative amplification, an exponential amplified fluorescent response could be obtained in theory based on the proposed nonlinear HCR. As a result, the nonlinear HCR generated a significant enhancement about 3 times compared with the normal HCR and 10 times compared with the directly immunofluorescence assay. Based on the proposed nonlinear HCR, the fluorescent signals increased with the concentration of TRPV4 in the range from 10 pg/mL to 100 ng/mL with a detection limit of 2.8 pg/mL, which would be useful for the sensitive detection of proteins in cell lysis or on cell surface. At the same time, the significant improvements via nonlinear HCR were achieved in the fluorescent imaging system compared with traditional immunofluorescence staining and normal HCR, proving the significant value of nonlinear HCR-based amplification strategy. Success in the establishment of the highly efficient nonlinear HCR strategy offered a simple and sensitive approach to demonstrate the concentration of special proteins on cell and other proteins and nucleotide potentially, revealing a simple and efficient technology for research fields of clinical diagnostics and molecular biology.
Collapse
Affiliation(s)
- Ping Li
- Department of Maxillofacial &E.N.T Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300020, PR China
| | - Hua Zhang
- Department of Maxillofacial &E.N.T Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300020, PR China
| | - Dong Wang
- Department of Maxillofacial &E.N.T Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300020, PR China
| | - Yingjie Tao
- Department of Maxillofacial &E.N.T Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300020, PR China
| | - Lun Zhang
- Department of Maxillofacial &E.N.T Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300020, PR China
| | - Wenchao Zhang
- Department of Maxillofacial &E.N.T Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300020, PR China.
| | - Xudong Wang
- Department of Maxillofacial &E.N.T Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300020, PR China.
| |
Collapse
|
28
|
Ischemic Brain Injury Leads to Brain Edema via Hyperthermia-Induced TRPV4 Activation. J Neurosci 2018; 38:5700-5709. [PMID: 29793978 DOI: 10.1523/jneurosci.2888-17.2018] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 04/26/2018] [Accepted: 05/03/2018] [Indexed: 01/27/2023] Open
Abstract
Brain edema is characterized by an increase in net brain water content, which results in an increase in brain volume. Although brain edema is associated with a high fatality rate, the cellular and molecular processes of edema remain largely unclear. Here, we developed an in vitro model of ischemic stroke-induced edema in which male mouse brain slices were treated with oxygen-glucose deprivation (OGD) to mimic ischemia. We continuously measured the cross-sectional area of the brain slice for 150 min under macroscopic microscopy, finding that OGD induces swelling of brain slices. OGD-induced swelling was prevented by pharmacologically blocking or genetically knocking out the transient receptor potential vanilloid 4 (TRPV4), a member of the thermosensitive TRP channel family. Because TRPV4 is activated at around body temperature and its activation is enhanced by heating, we next elevated the temperature of the perfusate in the recording chamber, finding that hyperthermia induces swelling via TRPV4 activation. Furthermore, using the temperature-dependent fluorescence lifetime of a fluorescent-thermosensitive probe, we confirmed that OGD treatment increases the temperature of brain slices through the activation of glutamate receptors. Finally, we found that brain edema following traumatic brain injury was suppressed in TRPV4-deficient male mice in vivo Thus, our study proposes a novel mechanism: hyperthermia activates TRPV4 and induces brain edema after ischemia.SIGNIFICANCE STATEMENT Brain edema is characterized by an increase in net brain water content, which results in an increase in brain volume. Although brain edema is associated with a high fatality rate, the cellular and molecular processes of edema remain unclear. Here, we developed an in vitro model of ischemic stroke-induced edema in which mouse brain slices were treated with oxygen-glucose deprivation. Using this system, we showed that the increase in brain temperature and the following activation of the thermosensitive cation channel TRPV4 (transient receptor potential vanilloid 4) are involved in the pathology of edema. Finally, we confirmed that TRPV4 is involved in brain edema in vivo using TRPV4-deficient mice, concluding that hyperthermia activates TRPV4 and induces brain edema after ischemia.
Collapse
|
29
|
Zhao H, Zhang K, Tang R, Meng H, Zou Y, Wu P, Hu R, Liu X, Feng H, Chen Y. TRPV4 Blockade Preserves the Blood-Brain Barrier by Inhibiting Stress Fiber Formation in a Rat Model of Intracerebral Hemorrhage. Front Mol Neurosci 2018; 11:97. [PMID: 29636662 PMCID: PMC5880899 DOI: 10.3389/fnmol.2018.00097] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 03/12/2018] [Indexed: 12/25/2022] Open
Abstract
Blood–brain barrier (BBB) disruption and subsequent brain edema play important roles in the secondary neuronal death and neurological dysfunction that are observed following intracerebral hemorrhage (ICH). In previous studies, transient receptor potential vanilloid 4 (TRPV4), a calcium-permeable mechanosensitive channel, was shown to induce cytotoxicity in many types of cells and to play a role in orchestrating barrier functions. In the present study, we explored the role of TRPV4 in ICH-induced brain injury, specifically investigating its effect on BBB disruption. Autologous arterial blood was injected into the basal ganglia of rats to mimic ICH. Adult male Sprague Dawley rats were randomly assigned to sham and experimental groups for studies on the time course of TRPV4 expression after ICH. The selective TRPV4 antagonist HC-067047 and TRPV4 siRNA were administered to evaluate the effects of TRPV4 inhibition. GSK1016790A, a TRPV4 agonist, was administered to naive rats to verify the involvement of TRPV4-induced BBB disruption. A PKC inhibitor, dihydrochloride (H7), and a selective RhoA inhibitor, C3 transferase, were administered to clarify the involvement of the PKCα/RhoA/MLC2 pathway following ICH. Post-ICH assessments including functional tests, brain edema measurements, Evans blue extravasation, western blotting and immunohistochemical assays were performed. TRPV4 inhibition remarkably ameliorated neurological symptoms, brain edema, and neuronal death, as well as BBB disruption, 24–72 h following ICH. Meanwhile, TRPV4 blockade preserved the expression of adherens and tight junction proteins, as well as BBB integrity, by inhibiting stress fiber formation, which might be correlated with the regulation of components of the PKCα/RhoA/MLC2 pathway. Furthermore, adherens and tight junction protein degradation induced by GSK1016790A treatment in naive rats was also related to PKCα/RhoA/MLC2-pathway-mediated stress fiber formation. Based on these findings, therapeutic interventions targeting TRPV4 may represent a novel approach to ameliorate secondary brain injury following ICH.
Collapse
Affiliation(s)
- Hengli Zhao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Kaiyuan Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Rongrui Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hui Meng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yongjie Zou
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Pengfei Wu
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Rong Hu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xin Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
30
|
TRPV4 is functionally expressed in oligodendrocyte precursor cells and increases their proliferation. Pflugers Arch 2018; 470:705-716. [PMID: 29569183 DOI: 10.1007/s00424-018-2130-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 10/17/2022]
Abstract
Oligodendrocytes, which differentiate from oligodendrocyte precursor cells (OPCs), ensheath axons with myelin, play an essential role in rapid conduction of action potentials and metabolically support neurons. Elucidation of the mechanisms underlying the proliferation, migration, differentiation, and survival of OPCs is considered indispensable for determining the causes of central nervous system diseases. However, the relationship between these functions of OPCs and their intracellular Ca2+ signaling has not been fully elucidated. Here, we investigated the function of transient receptor potential vanilloid 4 (TRPV4), a Ca2+-permeable channel that responds to hypo-osmolarity, mild temperature, mechanical stimulation, and endogenous arachidonic acid metabolites, in OPCs. Trpv4 mRNA was detected in OPCs in vivo and in primary cultured rat OPCs. In Ca2+ imaging experiments, treatment with the selective TRPV4 agonist GSK1016790A induced sustained elevation of the intracellular Ca2+ concentration in OPCs in a concentration-dependent manner, which was almost completely suppressed by co-treatment with the selective TRPV4 antagonist HC067047. Stimulation of TRPV4 by GSK1016790A augmented OPC proliferation, which was abolished by co-treatment with HC067047, the intracellular Ca2+ chelator BAPTA-AM, and the protein kinase C inhibitor bisindolylmaleimide II. By contrast, GSK1016790A did not significantly affect the migration or differentiation of OPCs. Taken together, these results suggest that TRPV4 is functionally expressed in OPCs and increases the proliferation of these cells without affecting their ability to differentiate into oligodendrocytes.
Collapse
|
31
|
Sugio S, Nagasawa M, Kojima I, Ishizaki Y, Shibasaki K. Transient receptor potential vanilloid 2 activation by focal mechanical stimulation requires interaction with the actin cytoskeleton and enhances growth cone motility. FASEB J 2016; 31:1368-1381. [PMID: 28007781 DOI: 10.1096/fj.201600686rr] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 12/12/2016] [Indexed: 11/11/2022]
Abstract
We have previously reported that transient receptor potential vanilloid 2 (TRPV2) can be activated by mechanical stimulation, which enhances axonal outgrowth in developing neurons; however, the molecular mechanisms that govern the contribution of TRPV2 activation to axonal outgrowth remain unclear. In the present study, we examined this mechanism by using PC12 cells as a neuronal model. Overexpression of TRPV2 enhanced axonal outgrowth in a mechanical stimulus-dependent manner. Accumulation of TRPV2 at the cell surface was 4-fold greater in the growth cone compared with the soma. In the growth cone, TRPV2 is not static, but dynamically accumulates (within ∼100 ms) to the site of mechanical stimulation. The dynamic and acute clustering of TRPV2 can enhance very weak mechanical stimuli via focal accumulation of TRPV2. Focal application of mechanical stimuli dramatically increased growth cone motility and caused actin reorganization via activation of TRPV2. We also found that TRPV2 physically interacts with actin and that changes in the actin cytoskeleton are required for its activation. Here, we demonstrated for the first time to our knowledge that TRPV2 clustering is induced by mechanical stimulation generated by axonal outgrowth and that TRPV2 activation is triggered by actin rearrangements that result from mechanical stimulation. Moreover, TRPV2 activation enhances growth cone motility and actin accumulation to promote axonal outgrowth. Sugio, S., Nagasawa, M., Kojima, I., Ishizaki, Y., Shibasaki, K. Transient receptor potential vanilloid 2 activation by focal mechanical stimulation requires interaction with the actin cytoskeleton and enhances growth cone motility.
Collapse
Affiliation(s)
- Shouta Sugio
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Masami Nagasawa
- Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Itaru Kojima
- Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Koji Shibasaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan;
| |
Collapse
|