1
|
Fabbri R, Scidà A, Saracino E, Conte G, Kovtun A, Candini A, Kirdajova D, Spennato D, Marchetti V, Lazzarini C, Konstantoulaki A, Dambruoso P, Caprini M, Muccini M, Ursino M, Anderova M, Treossi E, Zamboni R, Palermo V, Benfenati V. Graphene oxide electrodes enable electrical stimulation of distinct calcium signalling in brain astrocytes. NATURE NANOTECHNOLOGY 2024; 19:1344-1353. [PMID: 38987650 PMCID: PMC11405283 DOI: 10.1038/s41565-024-01711-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/31/2024] [Indexed: 07/12/2024]
Abstract
Astrocytes are responsible for maintaining homoeostasis and cognitive functions through calcium signalling, a process that is altered in brain diseases. Current bioelectronic tools are designed to study neurons and are not suitable for controlling calcium signals in astrocytes. Here, we show that electrical stimulation of astrocytes using electrodes coated with graphene oxide and reduced graphene oxide induces respectively a slow response to calcium, mediated by external calcium influx, and a sharp one, exclusively due to calcium release from intracellular stores. Our results suggest that the different conductivities of the substrate influence the electric field at the cell-electrolyte or cell-material interfaces, favouring different signalling events in vitro and ex vivo. Patch-clamp, voltage-sensitive dye and calcium imaging data support the proposed model. In summary, we provide evidence of a simple tool to selectively control distinct calcium signals in brain astrocytes for straightforward investigations in neuroscience and bioelectronic medicine.
Collapse
Affiliation(s)
- Roberta Fabbri
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy
| | - Alessandra Scidà
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy
| | - Emanuela Saracino
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy
| | - Giorgia Conte
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy
| | - Alessandro Kovtun
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy
| | - Andrea Candini
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy
| | - Denisa Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, CAS, Prague, Czech Republic
| | - Diletta Spennato
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy
| | - Valeria Marchetti
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, CAS, Prague, Czech Republic
- Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Chiara Lazzarini
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy
| | - Aikaterini Konstantoulaki
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy
| | - Paolo Dambruoso
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy
| | - Marco Caprini
- Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy
| | - Michele Muccini
- Consiglio Nazionale delle Ricerche, Istituto per lo Studio dei Materiali Nanostrutturati, Bologna, Italy
| | - Mauro Ursino
- Dipartimento di Ingegneria dell'Energia Elettrica e dell'Informazione 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, CAS, Prague, Czech Republic
| | - Emanuele Treossi
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy.
| | - Roberto Zamboni
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy
| | - Vincenzo Palermo
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy.
| | - Valentina Benfenati
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività, Bologna, Italy.
| |
Collapse
|
2
|
Zong P, Li CX, Feng J, Cicchetti M, Yue L. TRP Channels in Stroke. Neurosci Bull 2024; 40:1141-1159. [PMID: 37995056 PMCID: PMC11306852 DOI: 10.1007/s12264-023-01151-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 11/24/2023] Open
Abstract
Ischemic stroke is a devastating disease that affects millions of patients worldwide. Unfortunately, there are no effective medications for mitigating brain injury after ischemic stroke. TRP channels are evolutionally ancient biosensors that detect external stimuli as well as tissue or cellular injury. To date, many members of the TRP superfamily have been reported to contribute to ischemic brain injury, including the TRPC subfamily (1, 3, 4, 5, 6, 7), TRPV subfamily (1, 2, 3, 4) and TRPM subfamily (2, 4, 7). These TRP channels share structural similarities but have distinct channel functions and properties. Their activation during ischemic stroke can be beneficial, detrimental, or even both. In this review, we focus on discussing the interesting features of stroke-related TRP channels and summarizing the underlying cellular and molecular mechanisms responsible for their involvement in ischemic brain injury.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, School of Medicine (UConn Health), University of Connecticut, Farmington, CT, 06030, USA.
- Institute for the Brain and Cognitive Sciences, University of Connecticut, 337 Mansfield Road, Unit 1272, Storrs, CT, 06269, USA.
| | - Cindy X Li
- Department of Cell Biology, Calhoun Cardiology Center, School of Medicine (UConn Health), University of Connecticut, Farmington, CT, 06030, USA
| | - Jianlin Feng
- Department of Cell Biology, Calhoun Cardiology Center, School of Medicine (UConn Health), University of Connecticut, Farmington, CT, 06030, USA
| | - Mara Cicchetti
- Department of Cell Biology, Calhoun Cardiology Center, School of Medicine (UConn Health), University of Connecticut, Farmington, CT, 06030, USA
- Department of Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, School of Medicine (UConn Health), University of Connecticut, Farmington, CT, 06030, USA.
| |
Collapse
|
3
|
Takayama Y, Tominaga M. Interaction between TRP channels and anoctamins. Cell Calcium 2024; 121:102912. [PMID: 38823351 DOI: 10.1016/j.ceca.2024.102912] [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: 02/28/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Anoctamin 1 (ANO1) binds to transient receptor potential (TRP) channels (protein-protein interaction) and then is activated by TRP channels (functional interaction). TRP channels are non-selective cation channels that are expressed throughout the body and play roles in multiple physiological functions. Studies on TRP channels increased after the identification of TRP vanilloid 1 (TRPV1) in 1997. Calcium-activated chloride channel anoctamin 1 (ANO1, also called TMEM16A and DOG1) was identified in 2008. ANO1 plays a major role in TRP channel-mediated functions, as first shown in 2014 with the demonstration of a protein-protein interaction between TRPV4 and ANO1. In cells that co-express TRP channels and ANO1, calcium entering cells through activated TRP channels causes ANO1 activation. Therefore, in many tissues, the physiological functions related to TRP channels are modulated through chloride flux associated with ANO1 activation. In this review, we summarize the latest understanding of TRP-ANO1 interactions, particularly interaction of ANO1 with TRPV4, TRP canonical 6 (TRPC6), TRPV3, TRPV1, and TRPC2 in the salivary glands, blood vessels, skin keratinocytes, primary sensory neurons, and vomeronasal organs, respectively.
Collapse
Affiliation(s)
- Yasunori Takayama
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, Japan.
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 5-1 Aza-Higashiyama, Myodaiji, Okazaki, Aichi, Japan; Thermal Biology Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Aza-Higashiyama, Myodaiji, Okazaki, Aichi, Japan; Thermal Biology Research Group, Nagoya Advanced Research and Development Center, Nagoya City University, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, Japan.
| |
Collapse
|
4
|
Muhamad NA, Masutani K, Furukawa S, Yuri S, Toriyama M, Matsumoto C, Itoh S, Shinagawa Y, Isotani A, Toriyama M, Itoh H. Astrocyte-Specific Inhibition of the Primary Cilium Suppresses C3 Expression in Reactive Astrocyte. Cell Mol Neurobiol 2024; 44:48. [PMID: 38822888 PMCID: PMC11144130 DOI: 10.1007/s10571-024-01482-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/21/2024] [Indexed: 06/03/2024]
Abstract
C3-positive reactive astrocytes play a neurotoxic role in various neurodegenerative diseases. However, the mechanisms controlling C3-positive reactive astrocyte induction are largely unknown. We found that the length of the primary cilium, a cellular organelle that receives extracellular signals was increased in C3-positive reactive astrocytes, and the loss or shortening of primary cilium decreased the count of C3-positive reactive astrocytes. Pharmacological experiments suggested that Ca2+ signalling may synergistically promote C3 expression in reactive astrocytes. Conditional knockout (cKO) mice that specifically inhibit primary cilium formation in astrocytes upon drug stimulation exhibited a reduction in the proportions of C3-positive reactive astrocytes and apoptotic cells in the brain even after the injection of lipopolysaccharide (LPS). Additionally, the novel object recognition (NOR) score observed in the cKO mice was higher than that observed in the neuroinflammation model mice. These results suggest that the primary cilium in astrocytes positively regulates C3 expression. We propose that regulating astrocyte-specific primary cilium signalling may be a novel strategy for the suppression of neuroinflammation.
Collapse
Affiliation(s)
- Nor Atiqah Muhamad
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Kohei Masutani
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Shota Furukawa
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Shunsuke Yuri
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Michinori Toriyama
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, 1 Gakuenuegahara, Sanda, Hyogo, 669-1330, Japan
| | - Chuya Matsumoto
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Seiya Itoh
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Yuichiro Shinagawa
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Ayako Isotani
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Manami Toriyama
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan.
| | - Hiroshi Itoh
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan.
| |
Collapse
|
5
|
Zong P, Legere N, Feng J, Yue L. TRP Channels in Excitotoxicity. Neuroscientist 2024:10738584241246530. [PMID: 38682490 DOI: 10.1177/10738584241246530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Glutamate excitotoxicity is a central mechanism contributing to cellular dysfunction and death in various neurological disorders and diseases, such as stroke, traumatic brain injury, epilepsy, schizophrenia, addiction, mood disorders, Huntington's disease, Alzheimer's disease, Parkinson's disease, multiple sclerosis, pathologic pain, and even normal aging-related changes. This detrimental effect emerges from glutamate binding to glutamate receptors, including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, N-methyl-d-aspartate receptors, kainate receptors, and GluD receptors. Thus, excitotoxicity could be prevented by targeting glutamate receptors and their downstream signaling pathways. However, almost all the glutamate receptor antagonists failed to attenuate excitotoxicity in human patients, mainly due to the limited understanding of the underlying mechanisms regulating excitotoxicity. Transient receptor potential (TRP) channels serve as ancient cellular sensors capable of detecting and responding to both external and internal stimuli. The study of human TRP channels has flourished in recent decades since the initial discovery of mammalian TRP in 1995. These channels have been found to play pivotal roles in numerous pathologic conditions, including excitotoxicity. In this review, our focus centers on exploring the intricate interactions between TRP channels and glutamate receptors in excitotoxicity.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, USA
| | - Nicholas Legere
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jianlin Feng
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| |
Collapse
|
6
|
Mishima T, Komano K, Tabaru M, Kofuji T, Saito A, Ugawa Y, Terao Y. Repetitive pulsed-wave ultrasound stimulation suppresses neural activity by modulating ambient GABA levels via effects on astrocytes. Front Cell Neurosci 2024; 18:1361242. [PMID: 38601023 PMCID: PMC11004293 DOI: 10.3389/fncel.2024.1361242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Ultrasound is highly biopermeable and can non-invasively penetrate deep into the brain. Stimulation with patterned low-intensity ultrasound can induce sustained inhibition of neural activity in humans and animals, with potential implications for research and therapeutics. Although mechanosensitive channels are involved, the cellular and molecular mechanisms underlying neuromodulation by ultrasound remain unknown. To investigate the mechanism of action of ultrasound stimulation, we studied the effects of two types of patterned ultrasound on synaptic transmission and neural network activity using whole-cell recordings in primary cultured hippocampal cells. Single-shot pulsed-wave (PW) or continuous-wave (CW) ultrasound had no effect on neural activity. By contrast, although repetitive CW stimulation also had no effect, repetitive PW stimulation persistently reduced spontaneous recurrent burst firing. This inhibitory effect was dependent on extrasynaptic-but not synaptic-GABAA receptors, and the effect was abolished under astrocyte-free conditions. Pharmacological activation of astrocytic TRPA1 channels mimicked the effects of ultrasound by increasing the tonic GABAA current induced by ambient GABA. Pharmacological blockade of TRPA1 channels abolished the inhibitory effect of ultrasound. These findings suggest that the repetitive PW low-intensity ultrasound used in our study does not have a direct effect on neural function but instead exerts its sustained neuromodulatory effect through modulation of ambient GABA levels via channels with characteristics of TRPA1, which is expressed in astrocytes.
Collapse
Affiliation(s)
- Tatsuya Mishima
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Japan
| | - Kenta Komano
- Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Marie Tabaru
- Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Takefumi Kofuji
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Japan
- Radioisotope Laboratory, Kyorin University School of Medicine, Mitaka, Japan
| | - Ayako Saito
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Japan
| | - Yoshikazu Ugawa
- Department of Human Neurophysiology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yasuo Terao
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Japan
| |
Collapse
|
7
|
Rajasekhar P, Carbone SE, Johnston ST, Nowell CJ, Wiklendt L, Crampin EJ, She Y, DiCello JJ, Saito A, Sorensen L, Nguyen T, Lee KM, Hamilton JA, King SK, Eriksson EM, Spencer NJ, Gulbransen BD, Veldhuis NA, Poole DP. TRPV4 is expressed by enteric glia and muscularis macrophages of the colon but does not play a prominent role in colonic motility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574831. [PMID: 38260314 PMCID: PMC10802399 DOI: 10.1101/2024.01.09.574831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Mechanosensation is an important trigger of physiological processes in the gastrointestinal tract. Aberrant responses to mechanical input are associated with digestive disorders, including visceral hypersensitivity. Transient Receptor Potential Vanilloid 4 (TRPV4) is a mechanosensory ion channel with proposed roles in visceral afferent signaling, intestinal inflammation, and gut motility. While TRPV4 is a potential therapeutic target for digestive disease, current mechanistic understanding of how TRPV4 may influence gut function is limited by inconsistent reports of TRPV4 expression and distribution. Methods In this study we profiled functional expression of TRPV4 using Ca2+ imaging of wholemount preparations of the mouse, monkey, and human intestine in combination with immunofluorescent labeling for established cellular markers. The involvement of TRPV4 in colonic motility was assessed in vitro using videomapping and contraction assays. Results The TRPV4 agonist GSK1016790A evoked Ca2+ signaling in muscularis macrophages, enteric glia, and endothelial cells. TRPV4 specificity was confirmed using TRPV4 KO mouse tissue or antagonist pre-treatment. Calcium responses were not detected in other cell types required for neuromuscular signaling including enteric neurons, interstitial cells of Cajal, PDGFRα+ cells, and intestinal smooth muscle. TRPV4 activation led to rapid Ca2+ responses by a subpopulation of glial cells, followed by sustained Ca2+ signaling throughout the enteric glial network. Propagation of these waves was suppressed by inhibition of gap junctions or Ca2+ release from intracellular stores. Coordinated glial signaling in response to GSK1016790A was also disrupted in acute TNBS colitis. The involvement of TRPV4 in the initiation and propagation of colonic motility patterns was examined in vitro. Conclusions We reveal a previously unappreciated role for TRPV4 in the initiation of distension-evoked colonic motility. These observations provide new insights into the functional role of TRPV4 activation in the gut, with important implications for how TRPV4 may influence critical processes including inflammatory signaling and motility.
Collapse
Affiliation(s)
- Pradeep Rajasekhar
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
- Centre for Dynamic Imaging, WEHI, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Simona E Carbone
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Stuart T Johnston
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Lukasz Wiklendt
- College of Medicine & Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | - Edmund J Crampin
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Yinghan She
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Jesse J DiCello
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Ayame Saito
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Luke Sorensen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Thanh Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Kevin Mc Lee
- Department of Medicine, The University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3010, Australia
| | - John A Hamilton
- Department of Medicine, The University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3010, Australia
| | - Sebastian K King
- Department of Paediatric Surgery, The Royal Children's Hospital, Parkville, VIC 3052, Australia
- Surgical Research, Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Emily M Eriksson
- Population Health and Immunity, WEHI, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Nick J Spencer
- College of Medicine & Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | | | - Nicholas A Veldhuis
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Daniel P Poole
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| |
Collapse
|
8
|
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
|
9
|
Albini M, Krawczun-Rygmaczewska A, Cesca F. Astrocytes and brain-derived neurotrophic factor (BDNF). Neurosci Res 2023; 197:42-51. [PMID: 36780947 DOI: 10.1016/j.neures.2023.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/17/2023] [Accepted: 02/02/2023] [Indexed: 02/13/2023]
Abstract
Astrocytes are emerging in the neuroscience field as crucial modulators of brain functions, from the molecular control of synaptic plasticity to orchestrating brain-wide circuit activity for cognitive processes. The cellular pathways through which astrocytes modulate neuronal activity and plasticity are quite diverse. In this review, we focus on neurotrophic pathways, mostly those mediated by brain-derived neurotrophic factor (BDNF). Neurotrophins are a well-known family of trophic factors with pleiotropic functions in neuronal survival, maturation and activity. Within the brain, BDNF is the most abundantly expressed and most studied of all neurotrophins. While we have detailed knowledge of the effect of BDNF on neurons, much less is known about its physiology on astroglia. However, over the last years new findings emerged demonstrating that astrocytes take an active part into BDNF physiology. In this work, we discuss the state-of-the-art knowledge about astrocytes and BDNF. Indeed, astrocytes sense extracellular BDNF through its specific TrkB receptors and activate intracellular responses that greatly vary depending on the brain area, stage of development and receptors expressed. Astrocytes also uptake and recycle BDNF / proBDNF at synapses contributing to synaptic plasticity. Finally, experimental evidence is now available describing deficits in astrocytic BDNF in several neuropathologies, suggesting that astrocytic BDNF may represent a promising target for clinical translation.
Collapse
Affiliation(s)
- Martina Albini
- Department of Experimental Medicine, University of Genova, Italy; IIT Center for Synaptic Neuroscience and Technology, Genova, Italy
| | - Alicja Krawczun-Rygmaczewska
- IIT Center for Synaptic Neuroscience and Technology, Genova, Italy; Department of Life Sciences, University of Trieste, Italy
| | - Fabrizia Cesca
- IIT Center for Synaptic Neuroscience and Technology, Genova, Italy; Department of Life Sciences, University of Trieste, Italy.
| |
Collapse
|
10
|
Pathak Z, Jadav T, Roy A, Chopra M, Singh N, Sengupta P, Kumar H. Maresin-1 prevents blood-spinal cord barrier disruption associated with TRPV4 elevation in the experimental model of spinal cord injury. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159395. [PMID: 37729963 DOI: 10.1016/j.bbalip.2023.159395] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/22/2023]
Abstract
Recently, we reported the TRPV4 ion channel activation and its association with secondary damage after spinal cord injury (SCI). TRPV4 activation is linked with blood-spinal cord barrier (BSCB) disruption, endothelial damage, and inflammation after SCI. Specialized pro-resolving mediators (SPM) are endogenous lipid mediators released for inflammation resolution. Studies suggest that SPM could act as an endogenous antagonist of ion channels directly or indirectly at the plasma membrane. Herein, we studied the effect of maresin-1, a docosahexaenoic acid (DHA)-derived SPM, in SCI-induced TRPV4 expression and subsequent associated damage. First, employing a particular agonist (4αPDD) in endothelial and neuronal cell lines, we examined the potential of maresin-1 to block TRPV4 activation. Then we quantify the DHA levels in plasma and epicenter of the spinal cord in sham and at 1, 3, 7, 14, 21, and 28-days post-injury (DPI) using LC-MS. Then, we exogenously administered maresin-1 using two dosing regimens i.e., single-dose (1 μg) and multiple-dose (1 μg/day for seven days), to confirm its role in the TRPV4 inhibition and its linked damage. After SCI, DHA levels decrease in the spinal cord epicenter area as well as in the plasma. Treatment with maresin-1 attenuates TRPV4 expression, inflammatory cytokines, and chemokines and impedes neutrophil infiltration. Furthermore, treatment with maresin-1 prevents BSCB disruption, alleviates glial scar formation, and improves functional recovery. Thus, our results suggest that maresin-1 could modulate TRPV4 expression and could be a safe and promising approach to target inflammation and BSCB damage after SCI.
Collapse
Affiliation(s)
- Zarna Pathak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Tarang Jadav
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Manjeet Chopra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Nidhi Singh
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Pinaki Sengupta
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
11
|
Purushotham SS, Buskila Y. Astrocytic modulation of neuronal signalling. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1205544. [PMID: 37332623 PMCID: PMC10269688 DOI: 10.3389/fnetp.2023.1205544] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023]
Abstract
Neuronal signalling is a key element in neuronal communication and is essential for the proper functioning of the CNS. Astrocytes, the most prominent glia in the brain play a key role in modulating neuronal signalling at the molecular, synaptic, cellular, and network levels. Over the past few decades, our knowledge about astrocytes and their functioning has evolved from considering them as merely a brain glue that provides structural support to neurons, to key communication elements. Astrocytes can regulate the activity of neurons by controlling the concentrations of ions and neurotransmitters in the extracellular milieu, as well as releasing chemicals and gliotransmitters that modulate neuronal activity. The aim of this review is to summarise the main processes through which astrocytes are modulating brain function. We will systematically distinguish between direct and indirect pathways in which astrocytes affect neuronal signalling at all levels. Lastly, we will summarize pathological conditions that arise once these signalling pathways are impaired focusing on neurodegeneration.
Collapse
Affiliation(s)
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- The MARCS Institute, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
12
|
Tureckova J, Hermanova Z, Marchetti V, Anderova M. Astrocytic TRPV4 Channels and Their Role in Brain Ischemia. Int J Mol Sci 2023; 24:ijms24087101. [PMID: 37108263 PMCID: PMC10138480 DOI: 10.3390/ijms24087101] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/06/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Transient receptor potential cation channels subfamily V member 4 (TRPV4) are non-selective cation channels expressed in different cell types of the central nervous system. These channels can be activated by diverse physical and chemical stimuli, including heat and mechanical stress. In astrocytes, they are involved in the modulation of neuronal excitability, control of blood flow, and brain edema formation. All these processes are significantly impaired in cerebral ischemia due to insufficient blood supply to the tissue, resulting in energy depletion, ionic disbalance, and excitotoxicity. The polymodal cation channel TRPV4, which mediates Ca2+ influx into the cell because of activation by various stimuli, is one of the potential therapeutic targets in the treatment of cerebral ischemia. However, its expression and function vary significantly between brain cell types, and therefore, the effect of its modulation in healthy tissue and pathology needs to be carefully studied and evaluated. In this review, we provide a summary of available information on TRPV4 channels and their expression in healthy and injured neural cells, with a particular focus on their role in ischemic brain injury.
Collapse
Affiliation(s)
- Jana Tureckova
- Institute of Experimental Medicine, Czech Academy of Sciences, 1083 Videnska, 142 20 Prague, Czech Republic
| | - Zuzana Hermanova
- Institute of Experimental Medicine, Czech Academy of Sciences, 1083 Videnska, 142 20 Prague, Czech Republic
- Second Faculty of Medicine, Charles University, 84 V Uvalu, 150 06 Prague, Czech Republic
| | - Valeria Marchetti
- Institute of Experimental Medicine, Czech Academy of Sciences, 1083 Videnska, 142 20 Prague, Czech Republic
- Second Faculty of Medicine, Charles University, 84 V Uvalu, 150 06 Prague, Czech Republic
| | - Miroslava Anderova
- Institute of Experimental Medicine, Czech Academy of Sciences, 1083 Videnska, 142 20 Prague, Czech Republic
- Second Faculty of Medicine, Charles University, 84 V Uvalu, 150 06 Prague, Czech Republic
| |
Collapse
|
13
|
Jiang J, Yu Y. Pharmacologically targeting transient receptor potential channels for seizures and epilepsy: Emerging preclinical evidence of druggability. Pharmacol Ther 2023; 244:108384. [PMID: 36933703 PMCID: PMC10124570 DOI: 10.1016/j.pharmthera.2023.108384] [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: 01/06/2023] [Revised: 02/19/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
As one of the most prevalent and disabling brain disorders, epilepsy is characterized by spontaneous seizures that result from aberrant, excessive hyperactivity of a group of highly synchronized brain neurons. Remarkable progress in epilepsy research and treatment over the first two decades of this century led to a dramatical expansion in the third-generation antiseizure drugs (ASDs). However, there are still over 30% of patients suffering from seizures resistant to the current medications, and the broad unbearable adversative effects of ASDs significantly impair the quality of life in about 40% of individuals affected by the disease. Prevention of epilepsy in those who are at high risks is another major unmet medical need, given that up to 40% of epilepsy patients are believed to have acquired causes. Therefore, it is important to identify novel drug targets that can facilitate the discovery and development of new therapies engaging unprecedented mechanisms of action that might overcome these significant limitations. Also over the last two decades, calcium signaling has been increasingly recognized as a key contributory factor in epileptogenesis of many aspects. The intracellular calcium homeostasis involves a variety of calcium-permeable cation channels, the most important of which perhaps are the transient receptor potential (TRP) ion channels. This review focuses on recent exciting advances in understanding of TRP channels in preclinical models of seizure disorders. We also provide emerging insights into the molecular and cellular mechanisms of TRP channels-engaged epileptogenesis that might lead to new antiseizure therapies, epilepsy prevention and modification, and even a cure.
Collapse
Affiliation(s)
- Jianxiong Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States.
| | - Ying Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States.
| |
Collapse
|
14
|
Blockage of TRPV4 Downregulates the Nuclear Factor-Kappa B Signaling Pathway to Inhibit Inflammatory Responses and Neuronal Death in Mice with Pilocarpine-Induced Status Epilepticus. Cell Mol Neurobiol 2023; 43:1283-1300. [PMID: 35840809 DOI: 10.1007/s10571-022-01249-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 06/25/2022] [Indexed: 11/03/2022]
Abstract
The blockage of transient receptor potential vanilloid 4 (TRPV4) inhibits inflammation and reduces hippocampal neuronal injury in a pilocarpine-induced mouse model of temporal lobe epilepsy. However, the underlying mechanisms remain largely unclear. NF-κB signaling pathway is responsible for the inflammation and neuronal injury during epilepsy. Here, we explored whether TRPV4 blockage could affect the NF-κB pathway in mice with pilocarpine-induced status epilepticus (PISE). Application of a TRPV4 antagonist markedly attenuated the PISE-induced increase in hippocampal HMGB1, TLR4, phospho (p)-IκK (p-IκK), and p-IκBα protein levels, as well as those of cytoplasmic p-NF-κB p65 (p-p65) and nuclear NF-κB p65 and p50; in contrast, the application of GSK1016790A, a TRPV4 agonist, showed similar changes to PISE mice. Administration of the TLR4 antagonist TAK-242 or the NF-κB pathway inhibitor BAY 11-7082 led to a noticeable reduction in the hippocampal protein levels of cleaved IL-1β, IL-6 and TNF, as well as those of cytoplasmic p-p65 and nuclear p65 and p50 in GSK1016790A-injected mice. Finally, administration of either TAK-242 or BAY 11-7082 greatly increased neuronal survival in hippocampal CA1 and CA2/3 regions in GSK1016790A-injected mice. Therefore, TRPV4 activation increases HMGB1 and TLR4 expression, leading to IκK and IκBα phosphorylation and, consequently, NF-κB activation and nuclear translocation. The resulting increase in pro-inflammatory cytokine production is responsible for TRPV4 activation-induced neuronal injury. We conclude that blocking TRPV4 can downregulate HMGB1/TLR4/IκK/κBα/NF-κB signaling following PISE onset, an effect that may underlie the anti-inflammatory response and neuroprotective ability of TRPV4 blockage in mice with PISE.
Collapse
|
15
|
Liu Y, Peng J, Leng Q, Tian Y, Wu X, Tan R. Effects of Aloe-Emodin on the Expression of Brain Aquaporins and Secretion of Neurotrophic Factors in a Rat Model of Post-Stroke Depression. Int J Mol Sci 2023; 24:5206. [PMID: 36982280 PMCID: PMC10048947 DOI: 10.3390/ijms24065206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/25/2023] [Accepted: 03/01/2023] [Indexed: 03/12/2023] Open
Abstract
Post-stroke depression (PSD) is a common complication of stroke that can damage patients' brains. More and more studies have been conducted on PSD in recent years, but the exact mechanism is still not understood. Currently, animal models provide an alternative approach to better understand the pathophysiology of PSD and may also pave the way for the discovery of new treatments for depression. This study investigated the therapeutic effect and mechanism of aloe-emodin (AE) on PSD rats. Previous studies have shown that AE positively affects PSD in rats by improving depression, increasing their activities and curiosities, enhancing the number of neurons, and ameliorating damage to brain tissue. Meanwhile, AE could up-regulate the expression of brain-derived neurotrophic factor (BDNF) and neurotrophic 3 (NTF3), but it could also down-regulate the expression of aquaporins (AQP3, AQP4, and AQP5), glial fibrillary acidic protein (GFAP), and transient receptor potential vanilloid 4 (TRPV4), which is helpful in maintaining homeostasis and alleviating encephaledema. AE may be a prospective solution in the future for the treatment of PSD patients.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoqing Wu
- College of Life Science and Engineering, Southwest Jiao tong University, Chengdu 610031, China
| | - Rui Tan
- College of Life Science and Engineering, Southwest Jiao tong University, Chengdu 610031, China
| |
Collapse
|
16
|
Zhong X, Wang Y, Liu D, Liang Y, Liu W, Huang Y, Xie L, Cao W, Xu Y, Chen L. HC067047 Ameliorates Sepsis-associated Encephalopathy by Suppressing Endoplasmic Reticulum Stress and Oxidative Stress-Induced Pyroptosis in the Hippocampi of Mice. Neuroscience 2023; 517:117-127. [PMID: 36805006 DOI: 10.1016/j.neuroscience.2023.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
Sepsis-associated encephalopathy (SAE) is a common neurological complication of sepsis and is characterized by hyperneuroinflammation. NLRP3 inflammasome-mediated pyroptosis can induce an inflammatory cascade response and plays a key role in SAE. TRPV4 is involved in the hyperinflammatory response associated with inflammation; however, whether TRPV4 inhibition might alleviate SAE-related brain damage is still unknown. Therefore, we aimed to investigate the role and mechanism of HC067047, a potent inhibitor of TRPV4, in hyperneuroinflammation and blood-brain barrier (BBB) dysfunction in a lipopolysaccharide (LPS)-induced SAE mouse model. We found that HC067047 administration significantly inhibited the expression of TRPV4 and p-CamkIIα in the hippocampi of SAE mice. Furthermore, HC067047 treatment attenuated LPS-induced endoplasmic reticulum (ER) stress and oxidative stress (OS), thus remarkably preventing NLRP3 inflammasome-mediated pyroptosis, as well as the expression of proinflammatory factors (IL-1β and IL-18). Additionally, we found that HC067047 selectively prevented pyroptosis in hippocampal cells, mainly the neurons, oligodendrocytes and the resident microglia. The disruption of BBB integrity in SAE mice was also rescued by HC067047 intervention. Thus, we can conclude that the TRPV4 inhibitor HC067047 could protect against hippocampal cell pyroptosis, which might be due to the attenuation of the NLRP3 inflammasome-mediated pyroptosis pathway caused by ER stress and OS. Our findings suggest a potential preventive role for HC067047 in SAE.
Collapse
Affiliation(s)
- Xiaolin Zhong
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yajuan Wang
- Department of Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Dandan Liu
- Department of Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yue Liang
- Department of Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - WenJia Liu
- Department of Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yanmei Huang
- Department of Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Lihua Xie
- Department of Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Wenyu Cao
- Department of Human Anatomy, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yang Xu
- Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang 421001 Hunan, China.
| | - Ling Chen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
17
|
Križaj D, Cordeiro S, Strauß O. Retinal TRP channels: Cell-type-specific regulators of retinal homeostasis and multimodal integration. Prog Retin Eye Res 2023; 92:101114. [PMID: 36163161 PMCID: PMC9897210 DOI: 10.1016/j.preteyeres.2022.101114] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 02/05/2023]
Abstract
Transient receptor potential (TRP) channels are a widely expressed family of 28 evolutionarily conserved cationic ion channels that operate as primary detectors of chemical and physical stimuli and secondary effectors of metabotropic and ionotropic receptors. In vertebrates, the channels are grouped into six related families: TRPC, TRPV, TRPM, TRPA, TRPML, and TRPP. As sensory transducers, TRP channels are ubiquitously expressed across the body and the CNS, mediating critical functions in mechanosensation, nociception, chemosensing, thermosensing, and phototransduction. This article surveys current knowledge about the expression and function of the TRP family in vertebrate retinas, which, while dedicated to transduction and transmission of visual information, are highly susceptible to non-visual stimuli. Every retinal cell expresses multiple TRP subunits, with recent evidence establishing their critical roles in paradigmatic aspects of vertebrate vision that include TRPM1-dependent transduction of ON bipolar signaling, TRPC6/7-mediated ganglion cell phototransduction, TRP/TRPL phototransduction in Drosophila and TRPV4-dependent osmoregulation, mechanotransduction, and regulation of inner and outer blood-retina barriers. TRP channels tune light-dependent and independent functions of retinal circuits by modulating the intracellular concentration of the 2nd messenger calcium, with emerging evidence implicating specific subunits in the pathogenesis of debilitating diseases such as glaucoma, ocular trauma, diabetic retinopathy, and ischemia. Elucidation of TRP channel involvement in retinal biology will yield rewards in terms of fundamental understanding of vertebrate vision and therapeutic targeting to treat diseases caused by channel dysfunction or over-activation.
Collapse
Affiliation(s)
- David Križaj
- Departments of Ophthalmology, Neurobiology, and Bioengineering, University of Utah, Salt Lake City, USA
| | - Soenke Cordeiro
- Institute of Physiology, Faculty of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Olaf Strauß
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, a Corporate Member of Freie Universität, Humboldt-University, The Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
18
|
Goenaga J, Araque A, Kofuji P, Herrera Moro Chao D. Calcium signaling in astrocytes and gliotransmitter release. Front Synaptic Neurosci 2023; 15:1138577. [PMID: 36937570 PMCID: PMC10017551 DOI: 10.3389/fnsyn.2023.1138577] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Glia are as numerous in the brain as neurons and widely known to serve supportive roles such as structural scaffolding, extracellular ionic and neurotransmitter homeostasis, and metabolic support. However, over the past two decades, several lines of evidence indicate that astrocytes, which are a type of glia, play active roles in neural information processing. Astrocytes, although not electrically active, can exhibit a form of excitability by dynamic changes in intracellular calcium levels. They sense synaptic activity and release neuroactive substances, named gliotransmitters, that modulate neuronal activity and synaptic transmission in several brain areas, thus impacting animal behavior. This "dialogue" between astrocytes and neurons is embodied in the concept of the tripartite synapse that includes astrocytes as integral elements of synaptic function. Here, we review the recent work and discuss how astrocytes via calcium-mediated excitability modulate synaptic information processing at various spatial and time scales.
Collapse
|
19
|
Cullimore B, Baumann J, Rudzitis CN, Jo AO, Kirdajova D, Križaj D. Mechanotransduction mechanisms in central nervous system glia. Neural Regen Res 2022; 18:1031-1032. [PMID: 36254988 PMCID: PMC9827760 DOI: 10.4103/1673-5374.355758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Brenan Cullimore
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Jackson Baumann
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA,Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Christopher N. Rudzitis
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Andrew O. Jo
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Denisa Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - David Križaj
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA,Department of Bioengineering, University of Utah, Salt Lake City, UT, USA,Department of Neurobiology, University of Utah, Salt Lake City, UT, USA,Correspondence to: David Križaj, .
| |
Collapse
|
20
|
Lapajne L, Rudzitis CN, Cullimore B, Ryskamp D, Lakk M, Redmon SN, Yarishkin O, Krizaj D. TRPV4: Cell type-specific activation, regulation and function in the vertebrate eye. CURRENT TOPICS IN MEMBRANES 2022; 89:189-219. [PMID: 36210149 PMCID: PMC9879314 DOI: 10.1016/bs.ctm.2022.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The architecture of the vertebrate eye is optimized for efficient delivery and transduction of photons and processing of signaling cascades downstream from phototransduction. The cornea, lens, retina, vasculature, ciliary body, ciliary muscle, iris and sclera have specialized functions in ocular protection, transparency, accommodation, fluid regulation, metabolism and inflammatory signaling, which are required to enable function of the retina-light sensitive tissue in the posterior eye that transmits visual signals to relay centers in the midbrain. This process can be profoundly impacted by non-visual stimuli such as mechanical (tension, compression, shear), thermal, nociceptive, immune and chemical stimuli, which target these eye regions to induce pain and precipitate vision loss in glaucoma, diabetic retinopathy, retinal dystrophies, retinal detachment, cataract, corneal dysfunction, ocular trauma and dry eye disease. TRPV4, a polymodal nonselective cation channel, integrate non-visual inputs with homeostatic and signaling functions of the eye. The TRPV4 gene is expressed in most if not all ocular tissues, which vary widely with respect to the mechanisms of TRPV4 channel activation, modulation, oligomerization, and participation in protein- and lipid interactions. Under- and overactivation of TRPV4 may affect intraocular pressure, maintenance of blood-retina barriers, lens accommodation, neuronal function and neuroinflammation. Because TRPV4 dysregulation precipitates many pathologies across the anterior and posterior eye, the channel could be targeted to mitigate vision loss.
Collapse
Affiliation(s)
- Luka Lapajne
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States; Department of Ophthalmology, University Medical Centre, University of Ljubljana, Ljubljana, Slovenia
| | - Christopher N Rudzitis
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Brenan Cullimore
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Daniel Ryskamp
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Monika Lakk
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Sarah N Redmon
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Oleg Yarishkin
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - David Krizaj
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States; Department of Neurobiology, University of Utah, Salt Lake City, UT, United States; Department of Bioengineering, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
21
|
Jo AO, Lakk M, Rudzitis CN, Križaj D. TRPV4 and TRPC1 channels mediate the response to tensile strain in mouse Müller cells. Cell Calcium 2022; 104:102588. [PMID: 35398674 PMCID: PMC9119919 DOI: 10.1016/j.ceca.2022.102588] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/10/2022] [Accepted: 04/01/2022] [Indexed: 11/24/2022]
Abstract
Müller glia, a pillar of metabolic, volume regulatory and immune/inflammatory signaling in the mammalian retina, are among the earliest responders to mechanical stressors in the eye. Ocular trauma, edema, detachment and glaucoma evoke early inflammatory activation of Müller cells yet the identity of their mechanotransducers and signaling mechanisms downstream remains unknown. Here, we investigate expression of genes that encode putative stretch-activated calcium channels (SACs) in mouse Müller cells and study their responses to dynamical tensile loading in cells loaded with a calcium indicator dye. Transcript levels in purified glia were Trpc1>Piezo1>Trpv2>Trpv4>>Trpv1>Trpa1. Cyclic radial deformation of matrix-coated substrates produced dose-dependent increases in [Ca2+]i that were suppressed by the TRPV4 channel antagonist HC-067047 and by ablation of the Trpv4 gene. Stretch-evoked calcium responses were also reduced by knockdown and pharmacological inhibition of TRPC1 channels whereas the TRPV2 inhibitor tranilast had no effect. These data demonstrate that Müller cells are intrinsically mechanosensitive, with the response to tensile loading mediated through synergistic activation of TRPV4 and TRPC1 channels. Coupling between mechanical stress and Müller Ca2+ homeostasis has treatment implications, since many neuronal injury paradigms in the retina involve calcium dysregulation associated with inflammatory and immune signaling.
Collapse
Affiliation(s)
- Andrew O Jo
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132
| | - Monika Lakk
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132
| | - Christopher N Rudzitis
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132; Interdepartmental Program in Neuroscience
| | - David Križaj
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132; Interdepartmental Program in Neuroscience; Department of Neurobiology, University of Utah, Salt Lake City, UT 84112; Department of Bioengineering, University of Utah, Salt Lake City, UT 84112.
| |
Collapse
|
22
|
An P, Zhao XC, Liu MJ, You YQ, Li JY, Gong HS. Dexmedetomidine Alleviates Intracerebral Hemorrhage-Induced Anxiety-Like Behaviors in Mice Through the Inhibition of TRPV4 Opening. Front Pharmacol 2022; 13:852401. [PMID: 35431940 PMCID: PMC9012538 DOI: 10.3389/fphar.2022.852401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/21/2022] [Indexed: 11/30/2022] Open
Abstract
Post-stroke anxiety severely affects recovery in patients with intracerebral hemorrhage (ICH). Dexmedetomidine (Dex), a highly selective alpha 2 adrenal receptor (α2-AR) agonist, was recently found to exert an excellent protective effect against mental disorders including anxiety. The transient receptor potential vanilloid 4 (TRPV4) channel is involved in a series of diseases such as asthma, cancer, anxiety, and cardiac hypertrophy. This study examines whether Dex improved ICH-induced anxiety via the inhibition of TRPV4 channel opening. A rodent model of moderate ICH in the basal ganglia was established using autologous blood injection (20 μl). Mice were treated with Dex (25 μg/kg, intraperitoneal injection) every day for 3 days post-ICH. GSK1016790A (1 μmol/2 μl), an agonist of TRPV4, was administered via the left lateral ventricle. Thirty days post-ICH, post-stroke anxiety was evaluated by elevated plus-maze and open-field tests. Following behavioral tests, superoxide dismutase (SOD), malondialdehyde (MDA), astrocytic activation, and A1-and A2-type astrocytes were determined. Primary astrocytes were exposed to hemin to simulate ICH in vitro. Compared with sham-treated mice, Dex administration ameliorates ICH-induced decreases of distance and time in the open-arm, reduces distance and time in the central zone, increases astrocytic activation and A1-type astrocytes, elevates MDA content, downregulates total SOD contents, and decreases A2-type astrocytes. However, GSK1016790A partially reversed the neuroprotective effects of Dex. In addition, Dex significantly inhibited hemin-induced astrocytic activation in vitro. Dex improves ICH-induced anxiety-like behaviors in mice, and the mechanism might be associated with the inhibition of TRPV4-channel opening.
Collapse
Affiliation(s)
- Ping An
- Department of Neurobiology, School of Life Science, China Medical University, Shenyang, China
| | - Xiao-Chun Zhao
- Department of Anesthesiology, School and Hospital of Stomatology, China Medical University, Shenyang, China
- *Correspondence: Xiao-Chun Zhao,
| | - Man-Jia Liu
- Department of Anesthesiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - Yu-Qing You
- Department of Anesthesiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - Jing-Ya Li
- Department of Anesthesiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - He-Song Gong
- Department of Anesthesiology, ShengJing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
23
|
Nguyen TN, Siddiqui G, Veldhuis NA, Poole DP. Diverse Roles of TRPV4 in Macrophages: A Need for Unbiased Profiling. Front Immunol 2022; 12:828115. [PMID: 35126384 PMCID: PMC8811046 DOI: 10.3389/fimmu.2021.828115] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/24/2021] [Indexed: 12/27/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a non-selective mechanosensitive ion channel expressed by various macrophage populations. Recent reports have characterized the role of TRPV4 in shaping the activity and phenotype of macrophages to influence the innate immune response to pathogen exposure and inflammation. TRPV4 has been studied extensively in the context of inflammation and inflammatory pain. Although TRPV4 activity has been generally described as pro-inflammatory, emerging evidence suggests a more complex role where this channel may also contribute to anti-inflammatory activities. However, detailed understanding of how TRPV4 may influence the initiation, maintenance, and resolution of inflammatory disease remains limited. This review highlights recent insights into the cellular processes through which TRPV4 contributes to pathological conditions and immune processes, with a focus on macrophage biology. The potential use of high-throughput and omics methods as an unbiased approach for studying the functional outcomes of TRPV4 activation is also discussed.
Collapse
Affiliation(s)
- Thanh-Nhan Nguyen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Nicholas A. Veldhuis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
- *Correspondence: Daniel P. Poole, ; Nicholas A. Veldhuis,
| | - Daniel P. Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
- *Correspondence: Daniel P. Poole, ; Nicholas A. Veldhuis,
| |
Collapse
|
24
|
Acharya TK, Sahu RP, Kumar S, Kumar S, Rokade TP, Chakraborty R, Dubey NK, Shikha D, Chawla S, Goswami C. Function and regulation of thermosensitive ion channel TRPV4 in the immune system. CURRENT TOPICS IN MEMBRANES 2022; 89:155-188. [DOI: 10.1016/bs.ctm.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
25
|
Li W, Xu Y, Liu Z, Shi M, Zhang Y, Deng Y, Zhong X, Chen L, He J, Zeng J, Luo M, Cao W, Wan W. TRPV4 inhibitor HC067047 produces antidepressant-like effect in LPS-induced depression mouse model. Neuropharmacology 2021; 201:108834. [PMID: 34637786 DOI: 10.1016/j.neuropharm.2021.108834] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 01/07/2023]
Abstract
Inflammation is a crucial component that contributes to the pathogenesis of major depressive disorder. It has been revealed that the nonselective cation channel transient receptor potential vanilloid 4 (TRPV4) profoundly affects a variety of physiological processes, including inflammation. However, its roles and mechanisms in LPS-induced depression are still unclear. Here, for the first time, we found that there was a significant increase in TRPV4 in the hippocampus in a depression mouse model induced by LPS. TRPV4 inhibitor HC067047 or knockdown the hippocampal TRPV4 with TRPV4 shRNA could effectively rescue the aberrant behaviors. Furthermore, TRPV4 inhibitor HC067047 reduced the activation of astrocyte and microglia, decreased expression of CaMKII-NLRP3 inflammasome and increased the expression of neurogenesis marker DCX in the hippocampus. In addition, enhanced neuroinflammation in the serum was also reversed by TRPV4 inhibitor HC067047. Thus, we consider that TRPV4 has an important role in contributing to the depression-like behavior following LPS-induced systemic inflammation.
Collapse
Affiliation(s)
- Wei Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Yang Xu
- Institute of Neuroscience, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Zhenghai Liu
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Mengmeng Shi
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Yuan Zhang
- Department of Pathology, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Yingcheng Deng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Xiaolin Zhong
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, 421001, Hengyang, Hunan, China
| | - Ling Chen
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, 421001, Hengyang, Hunan, China
| | - Jie He
- Department of Pathology, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Jiayu Zeng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Mingying Luo
- Department of Anatomy & Histology & Embryology, Kunming Medical University, 650500, Kunming, Yunnan, China
| | - Wenyu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China.
| | - Wei Wan
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China; Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, 571199, Haikou, China.
| |
Collapse
|
26
|
Hoshi Y, Shibasaki K, Gailly P, Ikegaya Y, Koyama R. Thermosensitive receptors in neural stem cells link stress-induced hyperthermia to impaired neurogenesis via microglial engulfment. SCIENCE ADVANCES 2021; 7:eabj8080. [PMID: 34826234 PMCID: PMC8626080 DOI: 10.1126/sciadv.abj8080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Social stress impairs hippocampal neurogenesis and causes psychiatric disorders such as depression. Recent studies have highlighted the significance of increased body temperature in stress responses; however, whether and how social stress–induced hyperthermia affects hippocampal neurogenesis remains unknown. Here, using transgenic mice in which the thermosensitive transient receptor potential vanilloid 4 (TRPV4) is conditionally knocked out in Nestin-expressing neural stem cells (NSCs), we found that social defeat stress (SDS)–induced hyperthermia activates TRPV4 in NSCs in the dentate gyrus and thereby impairs hippocampal neurogenesis. Specifically, SDS activated TRPV4 in NSCs and induced the externalization of phosphatidylserine in NSCs, which was recognized by the brain-resident macrophage, microglia, and promoted the microglial engulfment of NSCs. SDS-induced impairment of hippocampal neurogenesis was ameliorated by NSC-specific knockout of TRPV4 or pharmacological removal of microglia. Thus, this study reveals a previously unknown role of thermosensitive receptors expressed by NSCs in stress responses.
Collapse
Affiliation(s)
- Yutaka Hoshi
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Koji Shibasaki
- Laboratory of Neurochemistry, Graduate School of Human Health Science, University of Nagasaki, Nagasaki 851-2195, Japan
| | - Philippe Gailly
- Laboratory of Cell Physiology, Institute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo 113-0033, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka 565-0871, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
- Corresponding author.
| |
Collapse
|
27
|
Schmaul S, Hanuscheck N, Bittner S. Astrocytic potassium and calcium channels as integrators of the inflammatory and ischemic CNS microenvironment. Biol Chem 2021; 402:1519-1530. [PMID: 34455729 DOI: 10.1515/hsz-2021-0256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022]
Abstract
Astrocytes are key regulators of their surroundings by receiving and integrating stimuli from their local microenvironment, thereby regulating glial and neuronal homeostasis. Cumulating evidence supports a plethora of heterogenic astrocyte subpopulations that differ morphologically and in their expression patterns of receptors, transporters and ion channels, as well as in their functional specialisation. Astrocytic heterogeneity is especially relevant under pathological conditions. In experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS), morphologically distinct astrocytic subtypes were identified and could be linked to transcriptome changes during different disease stages and regions. To allow for continuous awareness of changing stimuli across age and diseases, astrocytes are equipped with a variety of receptors and ion channels allowing the precise perception of environmental cues. Recent studies implicate the diverse repertoire of astrocytic ion channels - including transient receptor potential channels, voltage-gated calcium channels, inwardly rectifying K+ channels, and two-pore domain potassium channels - in sensing the brain state in physiology, inflammation and ischemia. Here, we review current evidence regarding astrocytic potassium and calcium channels and their functional contribution in homeostasis, neuroinflammation and stroke.
Collapse
Affiliation(s)
- Samantha Schmaul
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Nicholas Hanuscheck
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| |
Collapse
|
28
|
Abstract
The cerebral microcirculation undergoes dynamic changes in parallel with the development of neurons, glia, and their energy metabolism throughout gestation and postnatally. Cerebral blood flow (CBF), oxygen consumption, and glucose consumption are as low as 20% of adult levels in humans born prematurely but eventually exceed adult levels at ages 3 to 11 years, which coincide with the period of continued brain growth, synapse formation, synapse pruning, and myelination. Neurovascular coupling to sensory activation is present but attenuated at birth. By 2 postnatal months, the increase in CBF often is disproportionately smaller than the increase in oxygen consumption, in contrast to the relative hyperemia seen in adults. Vascular smooth muscle myogenic tone increases in parallel with developmental increases in arterial pressure. CBF autoregulatory response to increased arterial pressure is intact at birth but has a more limited range with arterial hypotension. Hypoxia-induced vasodilation in preterm fetal sheep with low oxygen consumption does not sustain cerebral oxygen transport, but the response becomes better developed for sustaining oxygen transport by term. Nitric oxide tonically inhibits vasomotor tone, and glutamate receptor activation can evoke its release in lambs and piglets. In piglets, astrocyte-derived carbon monoxide plays a central role in vasodilation evoked by glutamate, ADP, and seizures, and prostanoids play a large role in endothelial-dependent and hypercapnic vasodilation. Overall, homeostatic mechanisms of CBF regulation in response to arterial pressure, neuronal activity, carbon dioxide, and oxygenation are present at birth but continue to develop postnatally as neurovascular signaling pathways are dynamically altered and integrated. © 2021 American Physiological Society. Compr Physiol 11:1-62, 2021.
Collapse
|
29
|
Borodinova AA, Balaban PM, Bezprozvanny IB, Salmina AB, Vlasova OL. Genetic Constructs for the Control of Astrocytes' Activity. Cells 2021; 10:cells10071600. [PMID: 34202359 PMCID: PMC8306323 DOI: 10.3390/cells10071600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022] Open
Abstract
In the current review, we aim to discuss the principles and the perspectives of using the genetic constructs based on AAV vectors to regulate astrocytes’ activity. Practical applications of optogenetic approaches utilizing different genetically encoded opsins to control astroglia activity were evaluated. The diversity of astrocytic cell-types complicates the rational design of an ideal viral vector for particular experimental goals. Therefore, efficient and sufficient targeting of astrocytes is a multiparametric process that requires a combination of specific AAV serotypes naturally predisposed to transduce astroglia with astrocyte-specific promoters in the AAV cassette. Inadequate combinations may result in off-target neuronal transduction to different degrees. Potentially, these constraints may be bypassed with the latest strategies of generating novel synthetic AAV serotypes with specified properties by rational engineering of AAV capsids or using directed evolution approach by searching within a more specific promoter or its replacement with the unique enhancer sequences characterized using modern molecular techniques (ChIP-seq, scATAC-seq, snATAC-seq) to drive the selective transgene expression in the target population of cells or desired brain regions. Realizing these strategies to restrict expression and to efficiently target astrocytic populations in specific brain regions or across the brain has great potential to enable future studies.
Collapse
Affiliation(s)
- Anastasia A. Borodinova
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia;
| | - Pavel M. Balaban
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia;
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (I.B.B.); (A.B.S.); (O.L.V.)
- Correspondence:
| | - Ilya B. Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (I.B.B.); (A.B.S.); (O.L.V.)
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Alla B. Salmina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (I.B.B.); (A.B.S.); (O.L.V.)
- Research Institute of Molecular Medicine and Pathobiochemistry, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
- Research Center of Neurology, 125367 Moscow, Russia
| | - Olga L. Vlasova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (I.B.B.); (A.B.S.); (O.L.V.)
| |
Collapse
|
30
|
Kashio M. Thermosensation involving thermo-TRPs. Mol Cell Endocrinol 2021; 520:111089. [PMID: 33227348 DOI: 10.1016/j.mce.2020.111089] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 02/02/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022]
Abstract
The transient receptor potential (TRP) channels constitute a superfamily of large ion channels that are activated by a wide range of chemical, mechanical and thermal stimuli. TRP channels with temperature sensitivity are called thermo-TRPs. They are involved in diverse physiological functions through their detection of external environmental temperature and internal body temperature. Each thermo-TRP has its own characteristic temperature threshold for activation. As a group, they cover temperatures ranging from cold to nociceptive high temperatures. Recently, many studies have identified the functions of thermo-TRPs residing in deep organs where they are exposed to body temperature. Importantly, temperature thresholds of thermo-TRPs can be regulated by physiological factors enabling their function at relatively constant body temperature. Moreover, several thermo-TRPs are reportedly engaged in body temperature regulation. This review will summarize the current understanding of thermo-TRPs, including their roles in thermosensation and functional regulation of physiological responses at body temperature and the regulation of body temperature.
Collapse
Affiliation(s)
- Makiko Kashio
- Department of Physiology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.
| |
Collapse
|
31
|
Zhou L, Xu W, An D, Sha S, Men C, Li Y, Wang X, Du Y, Chen L. Transient receptor potential vanilloid 4 activation inhibits the delayed rectifier potassium channels in hippocampal pyramidal neurons: An implication in pathological changes following pilocarpine-induced status epilepticus. J Neurosci Res 2020; 99:914-926. [PMID: 33393091 DOI: 10.1002/jnr.24749] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/10/2020] [Accepted: 10/18/2020] [Indexed: 11/06/2022]
Abstract
Activation of transient receptor potential vanilloid 4 (TRPV4) can increase hippocampal neuronal excitability. TRPV4 has been reported to be involved in the pathogenesis of epilepsy. Voltage-gated potassium channels (VGPCs) play an important role in regulating neuronal excitability and abnormal VGPCs expression or function is related to epilepsy. Here, we examined the effect of TRPV4 activation on the delayed rectifier potassium current (IK ) in hippocampal pyramidal neurons and on the Kv subunits expression in male mice. We also explored the role of TRPV4 in changes in Kv subunits expression in male mice following pilocarpine-induced status epilepticus (PISE). Application of TRPV4 agonists, GSK1016790A and 5,6-EET, markedly reduced IK in hippocampal pyramidal neurons and shifted the voltage-dependent inactivation curve to the hyperpolarizing direction. GSK1016790A- and 5,6-EET-induced inhibition of IK was blocked by TRPV4 specific antagonists, HC-067047 and RN1734. GSK1016790A-induced inhibition of IK was markedly attenuated by calcium/calmodulin-dependent kinase II (CaMKII) antagonist. Application of GSK1016790A for up to 1 hr did not change the hippocampal protein levels of Kv1.1, Kv1.2, or Kv2.1. Intracerebroventricular injection of GSK1016790A for 3 d reduced the hippocampal protein levels of Kv1.2 and Kv2.1, leaving that of Kv1.1 unchanged. Kv1.2 and Kv2.1 protein levels as well as IK reduced markedly in hippocampi on day 3 post PISE, which was significantly reversed by HC-067047. We conclude that activation of TRPV4 inhibits IK in hippocampal pyramidal neurons, possibly by activating CaMKII. TRPV4-induced decrease in Kv1.2 and Kv2.1 expression and IK may be involved in the pathological changes following PISE.
Collapse
Affiliation(s)
- Li Zhou
- Department of Physiology, Nanjing Medical University, Nanjing, P.R. China
| | - Weixing Xu
- Department of Physiology, Nanjing Medical University, Nanjing, P.R. China
| | - Dong An
- Department of Physiology, Nanjing Medical University, Nanjing, P.R. China
| | - Sha Sha
- Department of Physiology, Nanjing Medical University, Nanjing, P.R. China
| | - Chen Men
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Yingchun Li
- Department of Physiology, Nanjing Medical University, Nanjing, P.R. China
| | - Xiaoli Wang
- Department of Physiology, Nanjing Medical University, Nanjing, P.R. China
| | - Yimei Du
- Research Center of Ion Channelopathy, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Lei Chen
- Department of Physiology, Nanjing Medical University, Nanjing, P.R. China.,Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, P.R. China
| |
Collapse
|
32
|
Feng X, Takayama Y, Ohno N, Kanda H, Dai Y, Sokabe T, Tominaga M. Increased TRPV4 expression in non-myelinating Schwann cells is associated with demyelination after sciatic nerve injury. Commun Biol 2020; 3:716. [PMID: 33247229 PMCID: PMC7695724 DOI: 10.1038/s42003-020-01444-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/30/2020] [Indexed: 11/23/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a non-selective calcium-permeable cation channel that is widely expressed and activated in various neurons and glial cells in the nervous system. Schwann cells (SCs) are primary glia cells that wrap around axons to form the myelin sheath in the peripheral nervous system. However, whether TRPV4 is expressed and functions in SCs is unclear. Here, we demonstrate functional expression of TRPV4 in mouse SCs and investigated its physiological significance. Deletion of TRPV4 did not affect normal myelin development for SCs in sciatic nerves in mice. However, after sciatic nerve cut injury, TRPV4 expression levels were remarkably increased in SCs following nerve demyelination. Ablation of TRPV4 expression impaired the demyelinating process after nerve injury, resulting in delayed remyelination and functional recovery of sciatic nerves. These results suggest that local activation of TRPV4 could be an attractive pharmacological target for therapeutic intervention after peripheral nerve injury. Feng et al. report that TRPV4 plays an important role in Schwann cells (SCs) during nerve demyelination and remyelination in mice. Using sciatic nerve cut injury mouse models, they find that TRPV4 expression is remarkably increased in demyelinating SCs during sciatic nerve injury; and ablation of TRPV4 expression impairs the demyelinating process after nerve injury, resulting in their delayed remyelination and functional recovery.
Collapse
Affiliation(s)
- Xiaona Feng
- Department of Physiological Sciences, SOKENDAI, Okazaki, Japan.,Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan.,Thermal Biology Group, Exploratory Research Center on Life and Living Systems (ExCELLS), Okazaki, Japan
| | - Yasunori Takayama
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| | - Nobuhiko Ohno
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Anatomy, Division of Histology and Cell Biology, Jichi Medical University, School of Medicine, Shimotsuke, Japan
| | - Hirosato Kanda
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Japan
| | - Yi Dai
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Japan
| | - Takaaki Sokabe
- Department of Physiological Sciences, SOKENDAI, Okazaki, Japan.,Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan.,Thermal Biology Group, Exploratory Research Center on Life and Living Systems (ExCELLS), Okazaki, Japan
| | - Makoto Tominaga
- Department of Physiological Sciences, SOKENDAI, Okazaki, Japan. .,Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan. .,Thermal Biology Group, Exploratory Research Center on Life and Living Systems (ExCELLS), Okazaki, Japan.
| |
Collapse
|
33
|
Schmidt E, Oheim M. Infrared Excitation Induces Heating and Calcium Microdomain Hyperactivity in Cortical Astrocytes. Biophys J 2020; 119:2153-2165. [PMID: 33130118 DOI: 10.1016/j.bpj.2020.10.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/01/2020] [Accepted: 10/07/2020] [Indexed: 11/16/2022] Open
Abstract
Unraveling how neural networks process and represent sensory information and how these cellular signals instruct behavioral output is a main goal in neuroscience. Two-photon activation of optogenetic actuators and calcium (Ca2+) imaging with genetically encoded indicators allow, respectively, the all-optical stimulation and readout of activity from genetically identified cell populations. However, these techniques locally expose the brain to high near-infrared light doses, raising the concern of light-induced adverse effects on the biology under study. Combining 2P imaging of Ca2+ transients in GCaMP6f-expressing cortical astrocytes and unbiased machine-based event detection, we demonstrate the subtle build-up of aberrant microdomain Ca2+ transients in the fine astroglial processes that depended on the average rather than peak laser power. Illumination conditions routinely being used in biological 2P microscopy (920-nm excitation, ∼100-fs, and ∼10 mW average power) increased the frequency of microdomain Ca2+ events but left their amplitude, area, and duration largely unchanged. Ca2+ transients in the otherwise silent soma were secondary to this peripheral hyperactivity that occurred without overt morphological damage. Continuous-wave (nonpulsed) 920-nm illumination at the same average power was as damaging as femtosecond pulses, unraveling the dominance of a heating-mediated damage mechanism. In an astrocyte-specific inositol 3-phosphate receptor type-2 knockout mouse, near-infrared light-induced Ca2+ microdomains persisted in the small processes, underpinning their resemblance to physiological inositol 3-phosphate receptor type-2-independent Ca2+ signals, whereas somatic hyperactivity was abolished. We conclude that, contrary to what has generally been believed in the field, shorter pulses and lower average power can help to alleviate damage and allow for longer recording windows at 920 nm.
Collapse
Affiliation(s)
- Elke Schmidt
- Université de Paris, SPPIN - Saints-Pères Paris Institute for the Neurosciences, CNRS, Paris, France
| | - Martin Oheim
- Université de Paris, SPPIN - Saints-Pères Paris Institute for the Neurosciences, CNRS, Paris, France.
| |
Collapse
|
34
|
Gualdani R, Seghers F, Yerna X, Schakman O, Tajeddine N, Achouri Y, Tissir F, Devuyst O, Gailly P. Mechanical activation of TRPV4 channels controls albumin reabsorption by proximal tubule cells. Sci Signal 2020; 13:13/653/eabc6967. [DOI: 10.1126/scisignal.abc6967] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Defects in protein reabsorption by the proximal tubule are toxic for epithelial cells in the nephron and may result in nephropathy. In this study, we showed that the ion channel TRPV4 modulated the endocytosis of albumin and low–molecular weight proteins in the proximal tubule. TRPV4 was found at the basolateral side of proximal tubule cells, and its mechanical activation by cell stretching induced Ca2+ entry into the cytosol, which promoted endocytosis. Trpv4−/− mice presented with mild proximal tubule dysfunction under basal conditions. To challenge endocytic function, the permeability of the glomerular filter was altered by systemic delivery of angiotensin II. The proteinuria induced by this treatment was more severe in Trpv4−/− than in Trpv4+/+ mice. Injecting antibodies against the glomerular basement membrane to induce glomerulonephritis is a more pathophysiologically relevant method of impairing glomerular filter permeability. Albuminuria was more severe in mice that lacked TRPV4 specifically in the proximal tubule than in control mice. These results emphasize the importance of TRPV4 in sensing pressure in the proximal tubule in response to variations in the amount of ultrafiltrate and unveil a mechanism that controls protein reabsorption.
Collapse
Affiliation(s)
- Roberta Gualdani
- Université catholique de Louvain, Institute of Neuroscience, Cell Physiology, av. Mounier 53/B1.53.17, B-1200 Brussels, Belgium
| | - François Seghers
- Université catholique de Louvain, Institute of Neuroscience, Cell Physiology, av. Mounier 53/B1.53.17, B-1200 Brussels, Belgium
| | - Xavier Yerna
- Université catholique de Louvain, Institute of Neuroscience, Cell Physiology, av. Mounier 53/B1.53.17, B-1200 Brussels, Belgium
| | - Olivier Schakman
- Université catholique de Louvain, Institute of Neuroscience, Cell Physiology, av. Mounier 53/B1.53.17, B-1200 Brussels, Belgium
| | - Nicolas Tajeddine
- Université catholique de Louvain, Institute of Neuroscience, Cell Physiology, av. Mounier 53/B1.53.17, B-1200 Brussels, Belgium
| | - Younès Achouri
- Université catholique de Louvain, de Duve Institute, Transgenic Core Facility, av. Hippocrate 75/B1.75.09, B-1200 Brussels, Belgium
| | - Fadel Tissir
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, av. Hippocrate 73/B1.73.16, B-1200 Brussels, Belgium
| | - Olivier Devuyst
- University of Zurich, Institute of Physiology, Winterthurerstr. 190, CH-8057 Zurich, Switzerland
| | - Philippe Gailly
- Université catholique de Louvain, Institute of Neuroscience, Cell Physiology, av. Mounier 53/B1.53.17, B-1200 Brussels, Belgium
| |
Collapse
|
35
|
Okubo Y. Astrocytic Ca2+ signaling mediated by the endoplasmic reticulum in health and disease. J Pharmacol Sci 2020; 144:83-88. [DOI: 10.1016/j.jphs.2020.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/19/2022] Open
|
36
|
Roles of TRP Channels in Neurological Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7289194. [PMID: 32963700 PMCID: PMC7492880 DOI: 10.1155/2020/7289194] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/02/2020] [Indexed: 11/17/2022]
Abstract
Transient receptor potential (TRP) proteins consist of a superfamily of cation channels that have been involved in diverse physiological processes in the brain as well as in the pathogenesis of neurological disease. TRP channels are widely expressed in the brain, including neurons and glial cells, as well as in the cerebral vascular endothelium and smooth muscle. Members of this channel superfamily show a wide variety of mechanisms ranging from ligand binding to voltage, physical, and chemical stimuli, implying the promising therapeutic potential of TRP in neurological diseases. In this review, we focus on the physiological functions of TRP channels in the brain and the pathological roles in neurological disorders to explore future potential neuroprotective strategies.
Collapse
|
37
|
Semyanov A, Henneberger C, Agarwal A. Making sense of astrocytic calcium signals — from acquisition to interpretation. Nat Rev Neurosci 2020; 21:551-564. [DOI: 10.1038/s41583-020-0361-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2020] [Indexed: 12/31/2022]
|
38
|
Huang Q, Wang X, Lin X, Zhang J, You X, Shao A. The Role of Transient Receptor Potential Channels in Blood-Brain Barrier Dysfunction after Ischemic Stroke. Biomed Pharmacother 2020; 131:110647. [PMID: 32858500 DOI: 10.1016/j.biopha.2020.110647] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/11/2020] [Accepted: 08/16/2020] [Indexed: 12/25/2022] Open
Abstract
Stroke is the leading cause of long-term disability, demanding an ever-increasing need to find treatment. Transient receptor potential (TRP) channels are nonselective Ca2+-permeable channels, among which TRPC, TRPM, and TRPV are widely expressed in the brain. Dysfunction of the blood brain barrier (BBB) is a core feature of stroke and is associated with severity of injury. As studies have shown, TRP channels influence various neuronal functions by regulating the BBB. Here, we briefly review the role of TRP channel in the BBB dysfunction after stroke, and explore the therapeutic potential of TRP-targeted therapy.
Collapse
Affiliation(s)
- Qingxia Huang
- Department of Echocardiography, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xianyi Lin
- Department of anesthesiology, Sir run run shaw hospital, school of medicine, zhejiang university, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Brain Research Institute, Zhejiang University, Hangzhou, China; Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
| | - Xiangdong You
- Department of Echocardiography, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
39
|
Transient receptor potential vanilloid 4 agonist GSK1016790A improves neurological outcomes after intracerebral hemorrhage in mice. Biochem Biophys Res Commun 2020; 529:590-595. [PMID: 32736678 DOI: 10.1016/j.bbrc.2020.06.103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/21/2020] [Indexed: 01/31/2023]
Abstract
Intracerebral hemorrhage (ICH) is one of the most severe subtypes of stroke with high morbidity and mortality. Although a lot of drug discovery studies have been conducted, the drugs with satisfactory therapeutic effects for motor paralysis after ICH have yet to reach clinical application. Transient receptor potential vanilloid 4 (TRPV4), a Ca2+-permeable cation channel and activated by hypoosmolarity and warm temperature, is expressed in various cell types. The present study investigated whether TRPV4 would participate in the brain damage in a mouse model of ICH. ICH was induced by intrastriatal treatment of collagenase. Administration of GSK1016790A, a selective TRPV4 agonist, attenuated neurological and motor deficits. The inhibitory effects of the TRPV4 agonist in collagenase-injected WT mice were completely disappeared in TRPV4-KO mice. The TRPV4 agonist did not alter brain injury volume and brain edema at 1 and 3 days after ICH induction. The TRPV4 agonist did not show any differences with respect to the increased number of Iba1-positive microglia/macrophages, GFAP-positive astrocytes, and Gr1-positive neutrophils at 1 and 3 days after ICH induction. Quantitative RT-PCR experiments revealed that the TRPV4 agonist significantly upregulated the expression level of c-fos, a marker of neuronal activity, while the agonist gave no effects on the expression level of cytokines/chemokines at 1 day after ICH induction, These results suggest that stimulation of TRPV4 would ameliorate ICH-induced brain injury, presumably by increased neuronal activity and TRPV4 provides a novel therapeutic target for the treatment for ICH.
Collapse
|
40
|
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: 24] [Impact Index Per Article: 6.0] [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
|
41
|
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: 3.5] [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
|
42
|
Kumar H, Lim CS, Choi H, Joshi HP, Kim KT, Kim YH, Park CK, Kim HM, Han IB. Elevated TRPV4 Levels Contribute to Endothelial Damage and Scarring in Experimental Spinal Cord Injury. J Neurosci 2020; 40:1943-1955. [PMID: 31974206 PMCID: PMC7046444 DOI: 10.1523/jneurosci.2035-19.2020] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/10/2019] [Accepted: 01/04/2020] [Indexed: 11/21/2022] Open
Abstract
Currently, the role of transient receptor potential vanilloid type 4 (TRPV4), a nonselective cation channel in the pathology of spinal cord injury (SCI), is not recognized. Herein, we report the expression and contribution of TRPV4 in the pathology of scarring and endothelial and secondary damage after SCI. TRPV4 expression increased during the inflammatory phase in female rats after SCI and was expressed primarily by cells at endothelial-microglial junctions. Two-photon microscopy of intracellular-free Ca2+ levels revealed a biphasic increase at similar time points after SCI. Expression of TRPV4 at the injury epicenter, but not intracellular-free Ca2+, progressively increases with the severity of the injury. Activation of TRPV4 with specific agonist altered the organization of endothelial cells, affected tight junctions in the hCMEC/D3 BBB cell line in vitro, and increases the scarring in rat spinal cord as well as induced endothelial damage. By contrast, suppression of TRPV4 with a specific antagonist or in female Trpv4 KO mouse attenuated inflammatory cytokines and chemokines, prevented the degradation of tight junction proteins, and preserve blood-spinal cord barrier integrity, thereby attenuate the scarring after SCI. Likewise, secondary damage was reduced, and behavioral outcomes were improved in Trpv4 KO mice after SCI. These results suggest that increased TRPV4 expression disrupts endothelial cell organization during the early inflammatory phase of SCI, resulting in tissue damage, vascular destabilization, blood-spinal cord barrier breakdown, and scarring. Thus, TRPV4 inhibition/knockdown represents a promising therapeutic strategy to stabilize/protect endothelial cells, attenuate nociception and secondary damage, and reduce scarring after SCI.SIGNIFICANCE STATEMENT TRPV4, a calcium-permeable nonselective cation channel, is widely expressed in both excitable and nonexcitable cells. Spinal cord injury (SCI) majorly caused by trauma/accidents is associated with changes in osmolarity, mechanical injury, and shear stress. After SCI, TRPV4 was increased and were found to be linked with the severity of injury at the epicenter at the time points that were reported to be critical for repair/treatment. Activation of TRPV4 was damaging to endothelial cells that form the blood-spinal cord barrier and thus contributes to scarring (glial and fibrotic). Importantly, inhibition/knockdown of TRPV4 prevented these effects. Thus, the manipulation of TRPV4 signaling might lead to new therapeutic strategies or combinatorial therapies to protect endothelial cells and enhance repair after SCI.
Collapse
Affiliation(s)
- Hemant Kumar
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488
| | - Chang Su Lim
- Department of Energy System Research and Department of Chemistry, Ajou University, Suwon, Gyeonggi-do, Republic of Korea, 16499
| | - Hyemin Choi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488
| | - Hari Prasad Joshi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488
| | - Kyoung-Tae Kim
- Department of Neurosurgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea, 41944
- Department of Neurosurgery, Kyungpook National University Hospital, Daegu, Republic of Korea, 41944, and
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, Republic of Korea, 13120
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, Republic of Korea, 13120
| | - Hwan Myung Kim
- Department of Energy System Research and Department of Chemistry, Ajou University, Suwon, Gyeonggi-do, Republic of Korea, 16499
| | - In-Bo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488,
| |
Collapse
|
43
|
Temperature elevation in epileptogenic foci exacerbates epileptic discharge through TRPV4 activation. J Transl Med 2020; 100:274-284. [PMID: 31641226 DOI: 10.1038/s41374-019-0335-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 12/15/2022] Open
Abstract
Physiological brain temperature is an important determinant of brain function, and it is well established that changes in brain temperature dynamically influence hippocampal neuronal activity. We previously demonstrated that the thermosensor TRPV4 is activated at physiological brain temperature in hippocampal neurons thereby controlling neuronal excitability in vitro. Here, we examined whether TRPV4 regulates neuronal excitability through its activation by brain temperature in vivo. We locally cooled the hippocampus using our novel electrical device and demonstrated constitutive TRPV4 activation in normal mouse brain. We generated a model of partial epilepsy by utilizing kindling stimuli in the ventral hippocampus of wild type (WT) or TRPV4-deficient (TRPV4KO) mice and obtained electroencephalograms (EEG). The frequencies of epileptic EEG in WT mice were significantly larger than those in TRPV4KO mice. These results indicate that TRPV4 activation is involved in disease progression of epilepsy. We expected that disease progression would enhance hyperexcitability and lead to hyperthermia in the epileptogenic foci. To confirm this hypothesis, we developed a new device to measure exact brain temperature only in a restricted local area. From the recording results by the new device, we found that the brain temperatures in epileptogenic zones were dramatically elevated compared with normal regions. Furthermore, we demonstrated that the temperature elevation was critical for disease progression. Based on these results, we speculate that brain cooling treatment at epileptogenic foci would effectively suppress epileptic discharges through inhibition of TRPV4. Notably, the cooling treatment drastically suppressed neuronal discharges dependent on the inactivation of TRPV4.
Collapse
|
44
|
Chen YL, Sonkusare SK. Endothelial TRPV4 channels and vasodilator reactivity. CURRENT TOPICS IN MEMBRANES 2020; 85:89-117. [PMID: 32402646 PMCID: PMC9748413 DOI: 10.1016/bs.ctm.2020.01.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) ion channels on the endothelial cell membrane are widely regarded as a crucial Ca2+ influx pathway that promotes endothelium-dependent vasodilation. The downstream vasodilatory targets of endothelial TRPV4 channels vary among different vascular beds, potentially contributing to endothelial cell heterogeneity. Although numerous studies have examined the role of endothelial TRPV4 channels using specific pharmacological tools over the past decade, their physiological significance remains unclear, mainly due to a lack of endothelium-specific knockouts. Moreover, the loss of endothelium-dependent vasodilation is a significant contributor to vascular dysfunction in cardiovascular disease. The activity of endothelial TRPV4 channels is impaired in cardiovascular disease; therefore, strategies targeting the mechanisms that reduce endothelial TRPV4 channel activity may restore vascular function and provide therapeutic benefit. In this chapter, we discuss endothelial TRPV4 channel-dependent signaling mechanisms, the heterogeneity in endogenous activators and targets of endothelial TRPV4 channels, and the role of endothelial TRPV4 channels in the pathogenesis of cardiovascular diseases. We also discuss potentially interesting future research directions that may provide novel insights into the physiological and pathological roles of endothelial TRPV4 channels.
Collapse
Affiliation(s)
- Yen-Lin Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, United States
| | - Swapnil K. Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, United States,Department of Molecular Physiology and Biological Physics, University of Virginia-School of Medicine, Charlottesville, VA, United States,Corresponding author:
| |
Collapse
|
45
|
Men C, Wang Z, Zhou L, Qi M, An D, Xu W, Zhan Y, Chen L. Transient receptor potential vanilloid 4 is involved in the upregulation of connexin expression following pilocarpine-induced status epilepticus in mice. Brain Res Bull 2019; 152:128-133. [PMID: 31299321 DOI: 10.1016/j.brainresbull.2019.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/03/2019] [Accepted: 07/05/2019] [Indexed: 10/26/2022]
Abstract
Epilepsy is characterized by spontaneous seizures. Changes in the expression of the connexins (Cxs) have been reported to be involved in epileptogenesis. It has previously been shown that the transient receptor potential vanilloid 4 (TRPV4) plays an important role in the modulation of neuronal excitability, and that application of a TRPV4 antagonist blocks hyperthermia-induced seizures. Accordingly, in the present study, we sought to explore whether TRPV4 is involved in the regulation of Cx expression following pilocarpine-induced status epilepticus (PISE) in mice. We observed that TRPV4 protein levels in hippocampi increased 3 h to 30 d following PISE, peaking 1-3 d after induction, and that pre-application of the TRPV4 antagonist HC-067047 increased the latency to develop SE induced by pilocarpine and reduced the success rate of PISE preparation. We demonstrated that Cx43 protein levels followed a time profile similar to that of TRPV4, and further showed that the increase in Cx43 protein levels on 3 d post-PISE was markedly attenuated by HC-067047. In contrast, the corresponding increase in Cx32 protein levels lagged substantially behind, and these levels were unaffected by HC-067047. Similarly, the TRPV4 agonist GSK1016790A increased the mRNA and protein levels of Cx43, but not those of Cx32. We thus conclude that the upregulation of Cx43 expression by TRPV4 may be involved in the pathophysiology of epilepsy.
Collapse
Affiliation(s)
- Chen Men
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Zhouqing Wang
- Department of Physiology, Nanjing Medical University, Nanjing, PR China
| | - Li Zhou
- Department of Physiology, Nanjing Medical University, Nanjing, PR China
| | - Mengwen Qi
- Department of Physiology, Nanjing Medical University, Nanjing, PR China
| | - Dong An
- Department of Physiology, Nanjing Medical University, Nanjing, PR China
| | - Weixing Xu
- Department of Physiology, Nanjing Medical University, Nanjing, PR China
| | - Yiyang Zhan
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| | - Lei Chen
- Department of Physiology, Nanjing Medical University, Nanjing, PR China; Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
46
|
Hypothalamic TRPV4 channels participate in the medial preoptic activation of warmth-defence responses in Wistar male rats. Pflugers Arch 2019; 471:1191-1203. [PMID: 31428866 DOI: 10.1007/s00424-019-02303-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 10/26/2022]
Abstract
Recently, we have described, in non-genetically modified rats, that peripheral transient receptor potential vanilloid-4 (TRPV4) channels are activated and trigger warmth-defence responses at ambient temperatures of 26-30 °C. Evidence points to the presence of TRPV4 in the medial preoptic area, a region described to be involved in the activation of thermoeffector pathways, including those involved in heat loss. Thus, we tested the hypothesis that TRPV4 in the medial preoptic area modulates thermoregulation under warm conditions. To this end, under two ambient temperatures (21 and 28 °C), body temperature was measured in rats following blockade of preoptic TRPV4 with two antagonists, HC-067047 and GSK 2193874. Oxygen consumption, heat loss index and preferred ambient temperature were also determined in order to assess thermoeffector activity. Antagonism of central TRPV4 caused an increase in body temperature in rats exposed to 28 °C, but not in those exposed to 21 °C. The body temperature increase at 28 °C was accompanied by an increase in oxygen consumption and an earlier reduction of the heat loss index. In behavioural experiments, control animals previously exposed to warm ambient temperatures (28-30 °C) for 2 h selected colder temperatures in a thermogradient compared to those injected with HC-067047. Our results support the idea that preoptic TRPV4 modulates thermoregulation in a warm environment by activating both autonomic and behavioural heat loss responses. Thus, according to the present study and to that published recently by our group, the activation of warmth-defence responses by TRPV4 seems to be dependent on the activity of both peripheral and central channels.
Collapse
|
47
|
Toft-Bertelsen TL, Larsen BR, MacAulay N. Sensing and regulation of cell volume - we know so much and yet understand so little: TRPV4 as a sensor of volume changes but possibly without a volume-regulatory role? Channels (Austin) 2019; 12:100-108. [PMID: 29424275 PMCID: PMC5972811 DOI: 10.1080/19336950.2018.1438009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cellular volume changes lead to initiation of cell volume regulatory events, the molecular identity of which remains unresolved. We here discuss experimental challenges associated with investigation of volume regulation during application of large, non-physiological osmotic gradients. The TRPV4 ion channel responds to volume increase irrespectively of the molecular mechanism underlying cell swelling, and is thus considered a sensor of volume changes. Evidence pointing towards the involvement of TRPV4 in subsequent volume regulatory mechanisms is intriguing, yet far from conclusive. We here present an experimental setting with astrocytic cell swelling in the absence of externally applied osmotic gradients, and the lack of evidence for involvement of TRPV4 in this regulatory volume response. Our aim with these new data and the preceding discussion is to stimulate further experimental effort in this area of research to clarify the role of TRPV4 and other channels and transporters in regulatory volume responses.
Collapse
Affiliation(s)
| | - Brian R Larsen
- a Department of Neuroscience , University of Copenhagen , Copenhagen , Denmark
| | - Nanna MacAulay
- a Department of Neuroscience , University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
48
|
TRPV4-induced inflammatory response is involved in neuronal death in pilocarpine model of temporal lobe epilepsy in mice. Cell Death Dis 2019; 10:386. [PMID: 31097691 PMCID: PMC6522539 DOI: 10.1038/s41419-019-1612-3] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/21/2019] [Accepted: 04/24/2019] [Indexed: 01/02/2023]
Abstract
Activation of transient receptor potential vanilloid 4 (TRPV4) induces neuronal injury. TRPV4 activation enhances inflammatory response and promotes the proinflammatory cytokine release in various types of tissue and cells. Hyperneuroinflammation contributes to neuronal damage in epilepsy. Herein, we examined the contribution of neuroinflammation to TRPV4-induced neurotoxicity and its involvement in the inflammation and neuronal damage in pilocarpine model of temporal lobe epilepsy in mice. Icv. injection of TRPV4 agonist GSK1016790A (GSK1016790A-injected mice) increased ionized calcium binding adapter molecule-1 (Iba-1) and glial fibrillary acidic protein (GFAP) protein levels and Iba-1-positive (Iba-1+) and GFAP-positive (GFAP+) cells in hippocampi, which indicated TRPV4-induced microglial cell and astrocyte activation. The protein levels of nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3 (NLRP3) inflammasome components NLRP3, apoptosis-related spotted protein (ASC) and cysteinyl aspartate-specific protease-1 (caspase-1) were increased in GSK1016790A-injected mice, which indicated NLRP3 inflammasome activation. GSK1016790A also increased proinflammatory cytokine IL-1β, TNF-α and IL-6 protein levels, which were blocked by caspase-1 inhibitor Ac-YVAD-cmk. GSK1016790A-induced neuronal death was attenuated by Ac-YVAD-cmk. Icv. injection of TRPV4-specific antagonist HC-067047 markedly increased the number of surviving cells 3 d post status epilepticus in pilocarpine model of temporal lobe epilepsy in mice (pilocarpine-induced status epilepticus, PISE). HC-067047 also markedly blocked the increase in Iba-1 and GFAP protein levels, as well as Iba-1+ and GFAP+ cells 3 d post-PISE. Finally, the increased protein levels of NLRP3, ASC and caspase-1 as well as IL-1β, TNF-α and IL-6 were markedly blocked by HC-067047. We conclude that TRPV4-induced neuronal death is mediated at least partially by enhancing the neuroinflammatory response, and this action is involved in neuronal injury following status epilepticus.
Collapse
|
49
|
Okubo Y, Iino M. Visualization of astrocytic intracellular Ca 2+ mobilization. J Physiol 2019; 598:1671-1681. [PMID: 30825213 DOI: 10.1113/jp277609] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 02/06/2019] [Indexed: 11/08/2022] Open
Abstract
Astrocytes generate robust intracellular Ca2+ concentration changes (Ca2+ signals), which are assumed to regulate astrocytic functions that play crucial roles in the regulation of brain functions. One frequently used strategy for exploring the role of astrocytic Ca2+ signalling is the use of mice deficient in the type 2 inositol 1,4,5-trisphosphate receptor (IP3 R2). These IP3 R2-knockout (KO) mice are reportedly devoid of Ca2+ mobilization from the endoplasmic reticulum (ER) in astrocytes. However, they have shown no functional deficits in several studies, causing a heated debate as to the functional relevance of ER-mediated Ca2+ signalling in astrocytes. Recently, the assumption that Ca2+ mobilization from the ER is absent in IP3 R2-KO astrocytes has been re-evaluated using intraorganellar Ca2+ imaging techniques. The new results indicated that IP3 R2-independent Ca2+ release may generate Ca2+ nanodomains around the ER, which may help explain the absence of functional deficits in IP3 R2-KO mice.
Collapse
Affiliation(s)
- Yohei Okubo
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, 133-0033, Japan
| | - Masamitsu Iino
- Division of Cellular and Molecular Pharmacology, Nihon University School of Medicine, Tokyo, 173-8610, Japan
| |
Collapse
|
50
|
Hirata Y, Suzuki Y, Tominaga M, Oku Y. TRPM8 channel is involved in the ventilatory response to CO 2 mediating hypercapnic Ca 2+ responses. Respir Physiol Neurobiol 2019; 263:20-25. [PMID: 30844520 DOI: 10.1016/j.resp.2019.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/22/2019] [Accepted: 03/04/2019] [Indexed: 02/03/2023]
Abstract
The role of TRP channels in the ventilatory response to CO2 was investigated in vivo. To this end, the respiration of unrestrained adult TRPM8-, TRPV1- and TRPV4-channel knockout mice was measured using whole-body plethysmography. Under control conditions and hyperoxic hypercapnia, no difference in respiratory parameters was observed between adult wild-type mice and TRPV1- and TRPV4-channel knockout mice. However, TRPM8-channel knockout mice showed decreased tidal volume under both hypercapnia and resting conditions. In addition, the expression of TRPM8, TRPV1 and TRPV4 mRNAs was detected in EGFP-positive glial cells in the medulla of GFAP promoter-EGFP transgenic mice by real-time PCR. Furthermore, we measured intracellular Ca2+ responses of TRPM8-overexpressing HEK-293 cells to hypercapnic acidosis. Subpopulations of cells that exhibited hypercapnic acidosis-induced Ca2+ response also responded to the application of menthol. These results suggest that TRPM8 partially mediates the ventilatory response to CO2 via changes in intracellular Ca2+ and is a chemosensing protein that may be involved in detecting endogenous CO2 production.
Collapse
Affiliation(s)
- Yutaka Hirata
- Department of Physiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan.
| | - Yoshiro Suzuki
- Division of Cell Signaling, National Institute for Physiological Sciences (Exploratory Research Center for Life and Living Systems), National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, 444-8787, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences (Exploratory Research Center for Life and Living Systems), National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, 444-8787, Japan
| | - Yoshitaka Oku
- Department of Physiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan.
| |
Collapse
|