1
|
Zhou Z, Bai Y, Gu X, Ren H, Xi W, Wang Y, Bian L, Liu X, Shen L, Xiang X, Huang W, Luo Z, Han B, Yao H. Membrane Associated RNA-Containing Vesicles Regulate Cortical Astrocytic Microdomain Calcium Transients in Awake Ischemic Stroke Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404391. [PMID: 39444078 PMCID: PMC11633488 DOI: 10.1002/advs.202404391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/01/2024] [Indexed: 10/25/2024]
Abstract
Astrocytic processes minutely regulate neuronal activity via tripartite synaptic structures. The precision-tuning of the function of astrocytic processes is garnering increasing attention because of its significance in promoting brain repair following ischemic stroke. Microdomain calcium (Ca2+) transients in astrocytic processes are pivotal for the functional regulation of these processes. However, the understanding of the alterations and regulatory mechanism of microdomain Ca2+ transients during stroke remains limited. In the present study, a fast high-resolution, miniaturized two-photon microscopy is used to show that the levels of astrocytic microdomain Ca2+ transients are significantly reduced in the peri-infarct area of awake ischemic stroke mice. This finding correlated with the behavioral deficits shown by these mice under freely-moving conditions. Mitochondrial Ca2+ activity is an important factor driving the microdomain Ca2+ transients. DEAD Box 1 (DDX1) bound to circSCMH1 (a circular RNA involved in vascular post-stroke repair) facilitates the formation of membrane-associated RNA-containing vesicles (MARVs) and enhances the activity of astrocytic microdomain Ca2+ transients, thereby promoting behavioral recovery. These results show that targeting astrocytic microdomain Ca2+ transients is a potential therapeutic approach in stroke intervention.
Collapse
Affiliation(s)
- Zhongqiu Zhou
- Department of PharmacologyJiangsu Provincial Key Laboratory of Critical Care MedicineSchool of MedicineSoutheast UniversityNanjing210009China
| | - Ying Bai
- Department of PharmacologyJiangsu Provincial Key Laboratory of Critical Care MedicineSchool of MedicineSoutheast UniversityNanjing210009China
| | - Xiaochun Gu
- Jiangsu Key Laboratory of Molecular and Functional ImagingDepartment of RadiologyZhongda HospitalMedical School of Southeast UniversityNanjing210009China
| | - Hui Ren
- Department of PharmacologyJiangsu Provincial Key Laboratory of Critical Care MedicineSchool of MedicineSoutheast UniversityNanjing210009China
| | - Wen Xi
- Department of PharmacologyJiangsu Provincial Key Laboratory of Critical Care MedicineSchool of MedicineSoutheast UniversityNanjing210009China
| | - Yu Wang
- Department of PharmacologyJiangsu Provincial Key Laboratory of Critical Care MedicineSchool of MedicineSoutheast UniversityNanjing210009China
| | - Liang Bian
- Department of PharmacologyJiangsu Provincial Key Laboratory of Critical Care MedicineSchool of MedicineSoutheast UniversityNanjing210009China
| | - Xue Liu
- Department of PharmacologyJiangsu Provincial Key Laboratory of Critical Care MedicineSchool of MedicineSoutheast UniversityNanjing210009China
| | - Ling Shen
- Department of PharmacologyJiangsu Provincial Key Laboratory of Critical Care MedicineSchool of MedicineSoutheast UniversityNanjing210009China
| | - Xianyuan Xiang
- Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055China
| | - Wenhui Huang
- Department of Molecular PhysiologyCenter for Integrative Physiology and Molecular MedicineUniversity of Saarland66421HomburgGermany
| | - Zhuojuan Luo
- The Key Laboratory of Developmental Genes and Human DiseaseSchool of Life Science and TechnologySoutheast UniversityNanjing210096China
| | - Bing Han
- Department of PharmacologyJiangsu Provincial Key Laboratory of Critical Care MedicineSchool of MedicineSoutheast UniversityNanjing210009China
| | - Honghong Yao
- Department of PharmacologyJiangsu Provincial Key Laboratory of Critical Care MedicineSchool of MedicineSoutheast UniversityNanjing210009China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
- Institute of Life SciencesKey Laboratory of Developmental Genes and Human DiseaseSoutheast UniversityNanjing210096China
| |
Collapse
|
2
|
Wu CH, Liao WH, Chu YC, Hsiao MY, Kung Y, Wang JL, Chen WS. Very Low-Intensity Ultrasound Facilitates Glymphatic Influx and Clearance via Modulation of the TRPV4-AQP4 Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401039. [PMID: 39494466 DOI: 10.1002/advs.202401039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Recently, the glymphatic system has been proposed as a mechanism for waste clearance from the brain parenchyma. Glymphatic dysfunction has previously been shown to be associated with several neurological diseases, including Alzheimer's disease, traumatic brain injury, and stroke. As such, it may serve as an important target for therapeutic interventions. In the present study, very low-intensity ultrasound (VLIUS) (center frequency, 1 MHz; pulse repetition frequency, 1 kHz; duty factor, 1%; spatial peak temporal average intensity [Ispta] = 3.68 mW cm2; and duration, 5 min) is found to significantly enhance the influx of cerebrospinal fluid tracers into the paravascular spaces of the brain, and further facilitate interstitial substance clearance from the brain parenchyma, including exogenous β-amyloid. Notably, no evidence of brain damage is observed following VLIUS stimulation. VLIUS may enhance glymphatic influx via the transient receptor potential vanilloid-4-aquaporin-4 pathway in astrocytes. This mechanism may provide insights into VLIUS-regulated glymphatic function that modifies the natural course of central nervous system disorders related to waste clearance dysfunction.
Collapse
Affiliation(s)
- Chueh-Hung Wu
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, 300, Taiwan
| | - Wei-Hao Liao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
| | - Ya-Cherng Chu
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Ming-Yen Hsiao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
| | - Yi Kung
- Department of Biomechatronic Engineering, National Chiayi University, Chiayi, 600, Taiwan
| | - Jaw-Lin Wang
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Wen-Shiang Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, 350, Taiwan
| |
Collapse
|
3
|
Frank R, Szarvas PA, Pesti I, Zsigmond A, Berkecz R, Menyhárt Á, Bari F, Farkas E. Nimodipine inhibits spreading depolarization, ischemic injury, and neuroinflammation in mouse live brain slice preparations. Eur J Pharmacol 2024; 977:176718. [PMID: 38849040 DOI: 10.1016/j.ejphar.2024.176718] [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: 04/11/2024] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024]
Abstract
Nimodipine is used to prevent delayed ischemic deficit in patients with aneurysmal subarachnoid hemorrhage (aSAH). Spreading depolarization (SD) is recognized as a factor in the pathomechanism of aSAH and other acute brain injuries. Although nimodipine is primarily known as a cerebral vasodilator, it may have a more complex mechanism of action due to the expression of its target, the L-type voltage-gated calcium channels (LVGCCs) in various cells in neural tissue. This study was designed to investigate the direct effect of nimodipine on SD, ischemic tissue injury, and neuroinflammation. SD in control or nimodipine-treated live mouse brain slices was induced under physiological conditions using electrical stimulation, or by subjecting the slices to hypo-osmotic stress or mild oxygen-glucose deprivation (mOGD). SD was recorded applying local field potential recording or intrinsic optical signal imaging. Histological analysis was used to estimate tissue injury, the number of reactive astrocytes, and the degree of microglia activation. Nimodipine did not prevent SD occurrence in mOGD, but it did reduce the rate of SD propagation and the cortical area affected by SD. In contrast, nimodipine blocked SD occurrence in hypo-osmotic stress, but had no effect on SD propagation. Furthermore, nimodipine prevented ischemic injury associated with SD in mOGD. Nimodipine also exhibited anti-inflammatory effects in mOGD by reducing reactive astrogliosis and microglial activation. The results demonstrate that nimodipine directly inhibits SD, independent of nimodipine's vascular effects. Therefore, the use of nimodipine may be extended to treat acute brain injuries where SD plays a central role in injury progression.
Collapse
Affiliation(s)
- Rita Frank
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary; Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary.
| | - Péter Archibald Szarvas
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary; Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - István Pesti
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary; Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Anna Zsigmond
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Róbert Berkecz
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary; Department of Forensic Medicine, Albert Szent-Györgyi Health Centre, Kossuth Lajos Sgt. 40, Szeged, Hungary
| | - Ákos Menyhárt
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary; Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ferenc Bari
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary; Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary.
| |
Collapse
|
4
|
Chen Z, Liu B, Zhou D, Lei M, Yang J, Hu Z, Duan W. AQP4 regulates ferroptosis and oxidative stress of Muller cells in diabetic retinopathy by regulating TRPV4. Exp Cell Res 2024; 439:114087. [PMID: 38735619 DOI: 10.1016/j.yexcr.2024.114087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
Diabetic retinopathy (DR) is a common microvascular complication that causes visual impairment or loss. Aquaporin 4 (AQP4) is a regulatory protein involved in water transport and metabolism. In previous studies, we found that AQP4 is related to hypoxia injury in Muller cells. Transient receptor potential cation channel subfamily V member 4 (TRPV4) is a non-selective cation channel protein involved in the regulation of a variety of ophthalmic diseases. However, the effects of AQP4 and TRPV4 on ferroptosis and oxidative stress in high glucose (HG)-treated Muller cells are unclear. In this study, we investigated the functions of AQP4 and TRPV4 in DR. HG was used to treat mouse Muller cells. Reverse transcription quantitative polymerase chain reaction was used to measure AQP4 mRNA expression. Western blotting was used to detect the protein levels of AQP4, PTGS2, GPX4, and TRPV4. Cell count kit-8, flow cytometry, 5,5',6,6'-tetrachloro-1,1,3,3'-tetraethylbenzimidazolyl carbocyanine iodide staining, and glutathione (GSH), superoxide dismutase (SOD), and malondialdehyde (MDA) kits were used to evaluate the function of the Muller cells. Streptozotocin was used to induce DR in rats. Haematoxylin and eosin staining was performed to stain the retina of rats. GSH, SOD, and MDA detection kits, immunofluorescence, and flow cytometry assays were performed to study the function of AQP4 and TRPV4 in DR rats. Results found that AQP4 and TRPV4 were overexpressed in HG-induced Muller cells and streptozotocin-induced DR rats. AQP4 inhibition promoted proliferation and cell cycle progression, repressed cell apoptosis, ferroptosis, and oxidative stress, and alleviated retinal injury in DR rats. Mechanistically, AQP4 positively regulated TRPV4 expression. Overexpression of TRPV4 enhanced ferroptosis and oxidative stress in HG-treated Muller cells, and inhibition of TRPV4 had a protective effect on DR-induced retinal injury in rats. In conclusion, inhibition of AQP4 inhibits the ferroptosis and oxidative stress in Muller cells by downregulating TRPV4, which may be a potential target for DR therapy.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China.
| | - Bingjie Liu
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China
| | - Daijiao Zhou
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China
| | - Mingshu Lei
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China
| | - Jingying Yang
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China
| | - Zhongyin Hu
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China
| | - Wenhua Duan
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China
| |
Collapse
|
5
|
Li B, Zhao A, Tian T, Yang X. Mechanobiological insight into brain diseases based on mechanosensitive channels: Common mechanisms and clinical potential. CNS Neurosci Ther 2024; 30:e14809. [PMID: 38923822 PMCID: PMC11197048 DOI: 10.1111/cns.14809] [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: 02/28/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND As physical signals, mechanical cues regulate the neural cells in the brain. The mechanosensitive channels (MSCs) perceive the mechanical cues and transduce them by permeating specific ions or molecules across the plasma membrane, and finally trigger a series of intracellular bioelectrical and biochemical signals. Emerging evidence supports that wide-distributed, high-expressed MSCs like Piezo1 play important roles in several neurophysiological processes and neurological disorders. AIMS To systematically conclude the functions of MSCs in the brain and provide a novel mechanobiological perspective for brain diseases. METHOD We summarized the mechanical cues and MSCs detected in the brain and the research progress on the functional roles of MSCs in physiological conditions. We then concluded the pathological activation and downstream pathways triggered by MSCs in two categories of brain diseases, neurodegenerative diseases and place-occupying damages. Finally, we outlined the methods for manipulating MSCs and discussed their medical potential with some crucial outstanding issues. RESULTS The MSCs present underlying common mechanisms in different brain diseases by acting as the "transportation hubs" to transduce the distinct signal patterns: the upstream mechanical cues and the downstream intracellular pathways. Manipulating the MSCs is feasible to alter the complicated downstream processes, providing them promising targets for clinical treatment. CONCLUSIONS Recent research on MSCs provides a novel insight into brain diseases. The common mechanisms mediated by MSCs inspire a wide range of therapeutic potentials targeted on MSCs in different brain diseases.
Collapse
Affiliation(s)
- Bolong Li
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- College of Life SciencesUniversity of Chinese Academy of ScienceBeijingChina
| | - An‐ran Zhao
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- College of Life SciencesUniversity of Chinese Academy of ScienceBeijingChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| | - Tian Tian
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| | - Xin Yang
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| |
Collapse
|
6
|
Lénárt N, Cserép C, Császár E, Pósfai B, Dénes Á. Microglia-neuron-vascular interactions in ischemia. Glia 2024; 72:833-856. [PMID: 37964690 DOI: 10.1002/glia.24487] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023]
Abstract
Cerebral ischemia is a devastating condition that results in impaired blood flow in the brain leading to acute brain injury. As the most common form of stroke, occlusion of cerebral arteries leads to a characteristic sequence of pathophysiological changes in the brain tissue. The mechanisms involved, and comorbidities that determine outcome after an ischemic event appear to be highly heterogeneous. On their own, the processes leading to neuronal injury in the absence of sufficient blood supply to meet the metabolic demand of the cells are complex and manifest at different temporal and spatial scales. While the contribution of non-neuronal cells to stroke pathophysiology is increasingly recognized, recent data show that microglia, the main immune cells of the central nervous system parenchyma, play previously unrecognized roles in basic physiological processes beyond their inflammatory functions, which markedly change during ischemic conditions. In this review, we aim to discuss some of the known microglia-neuron-vascular interactions assumed to contribute to the acute and delayed pathologies after cerebral ischemia. Because the mechanisms of neuronal injury have been extensively discussed in several excellent previous reviews, here we focus on some recently explored pathways that may directly or indirectly shape neuronal injury through microglia-related actions. These discoveries suggest that modulating gliovascular processes in different forms of stroke and other neurological disorders might have presently unexplored therapeutic potential in combination with neuroprotective and flow restoration strategies.
Collapse
Affiliation(s)
- Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Csaba Cserép
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Eszter Császár
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Pósfai
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
7
|
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
|
8
|
Huang J, Wang XS, Gao T, Wang X, Yu MY, Song HX, Wang BY, Li LM, Zeng Q, Zhang HN. Astrocyte KDM4A mediates chemokines and drives neutrophil infiltration to aggravate cerebral ischemia and reperfusion injury. J Cereb Blood Flow Metab 2024; 44:491-507. [PMID: 38008899 PMCID: PMC10981400 DOI: 10.1177/0271678x231216158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 11/28/2023]
Abstract
Neutrophils plays a crucial role in acute ischemic brain injury and have emerged as potential treatment targets to mitigate such injuries. Lysine-specific demethylase 4 A (KDM4A), a member of the histone lysine demethylase family of enzymes involved in transcriptional regulation of gene expression, is upregulated during hypoxic events. However, the exact role of KDM4A in the pathological process of ischemic stroke remains largely unexplored. Our findings reveal that there was an upregulation of KDM4A levels in reactive astrocytes within both stroke mouse models and in vitro oxygen-glucose deprivation/regeneration (OGD/R) models. Using a conditional knockout mouse, we observed that astrocytic Kdm4a knockout regulates neutrophil infiltration and alleviates brain injury following middle cerebral artery occlusion reperfusion. Furthermore, Kdm4a deficiency astrocytes displayed lower chemokine C-X-C motif ligand 1 (CXCL1) level upon OGD/R and decreased neutrophil infiltration in a transwell system. Mechanistically, KDM4A, in cooperation with nuclear factor-kappa B (NF-κB), activates Cxcl1 gene expression by demethylating histone H3 lysine 9 trimethylation at Cxcl1 gene promoters in astrocytes upon OGD/R injury. Our findings suggest that astrocyte KDM4A-mediated Cxcl1 activation contributes to neutrophil infiltration via cooperation with NF-κB, and KDM4A in astrocytes may serve as a potential therapeutic target to modulate neutrophil infiltration after stroke.
Collapse
Affiliation(s)
- Jing Huang
- Department of Health Management, Second Affiliated Hospital, Fourth Military Medical University, Xi’an, China
- Department of Neurology, Second Affiliated Hospital, Fourth Military Medical University, Xi’an, China
- Health Management Institute, Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xin-Shang Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Tian Gao
- Department of Health Management, Second Affiliated Hospital, Fourth Military Medical University, Xi’an, China
| | - Xing Wang
- Department of Health Management, Second Affiliated Hospital, Fourth Military Medical University, Xi’an, China
| | - Man-Yang Yu
- Department of Health Management, Second Affiliated Hospital, Fourth Military Medical University, Xi’an, China
| | - Hao-Xin Song
- Department of Health Management, Second Affiliated Hospital, Fourth Military Medical University, Xi’an, China
| | - Bi-Yan Wang
- Department of Health Management, Second Affiliated Hospital, Fourth Military Medical University, Xi’an, China
| | - Ling-Mei Li
- Department of Health Management, Second Affiliated Hospital, Fourth Military Medical University, Xi’an, China
| | - Qiang Zeng
- Health Management Institute, Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hui-Nan Zhang
- Department of Health Management, Second Affiliated Hospital, Fourth Military Medical University, Xi’an, China
- Department of Neurology, Second Affiliated Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
9
|
Shi L, Jiang C, Xu H, Wu J, Lu J, He Y, Yin X, Chen Z, Cao D, Shen X, Hou X, Han J. Hyperoside ameliorates cerebral ischaemic-reperfusion injury by opening the TRPV4 channel in vivo through the IP 3-PKC signalling pathway. PHARMACEUTICAL BIOLOGY 2023; 61:1000-1012. [PMID: 37410551 DOI: 10.1080/13880209.2023.2228379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 05/15/2023] [Accepted: 06/18/2023] [Indexed: 07/08/2023]
Abstract
CONTEXT Hyperoside (Hyp), one of the active flavones from Rhododendron (Ericaceae), has beneficial effects against cerebrovascular disease. However, the effect of Hyp on vasodilatation has not been elucidated. OBJECTIVE To explore the effect of Hyp on vasodilatation in the cerebral basilar artery (CBA) of Sprague-Dawley (SD) rats suffering with ischaemic-reperfusion (IR) injury. MATERIALS AND METHODS Sprague-Dawley rats were randomly divided into sham, model, Hyp, Hyp + channel blocker and channel blocker groups. Hyp (50 mg/kg, IC50 = 18.3 μg/mL) and channel blocker were administered via tail vein injection 30 min before ischaemic, followed by 20 min of ischaemic and 2 h of reperfusion. The vasodilation, hyperpolarization, ELISA assay, haematoxylin-eosin (HE), Nissl staining and channel-associated proteins and qPCR were analysed. Rat CBA smooth muscle cells were isolated to detect the Ca2+ concentration and endothelial cells were isolated to detect apoptosis rate. RESULTS Hyp treatment significantly ameliorated the brain damage induced by IR and evoked endothelium-dependent vasodilation rate (47.93 ± 3.09% vs. 2.99 ± 1.53%) and hyperpolarization (-8.15 ± 1.87 mV vs. -0.55 ± 0.42 mV) by increasing the expression of IP3R, PKC, transient receptor potential vanilloid channel 4 (TRPV4), IKCa and SKCa in the CBA. Moreover, Hyp administration significantly reduced the concentration of Ca2+ (49.08 ± 7.74% vs. 83.52 ± 6.93%) and apoptosis rate (11.27 ± 1.89% vs. 23.44 ± 2.19%) in CBA. Furthermore, these beneficial effects of Hyp were blocked by channel blocker. DISCUSSION AND CONCLUSIONS Although Hyp showed protective effect in ischaemic stroke, more clinical trial certification is needed due to the difference between animals and humans.
Collapse
Affiliation(s)
- Lei Shi
- Pharmacology 3rd Grade Laboratory of the State Administration of Traditional Chinese Medicine, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, China
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Chenchen Jiang
- Pharmacology 3rd Grade Laboratory of the State Administration of Traditional Chinese Medicine, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, China
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Hanghang Xu
- Pharmacology 3rd Grade Laboratory of the State Administration of Traditional Chinese Medicine, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, China
| | - Jiangping Wu
- Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, China
| | - Jiajun Lu
- Pharmacology 3rd Grade Laboratory of the State Administration of Traditional Chinese Medicine, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, China
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yuxiang He
- Pharmacology 3rd Grade Laboratory of the State Administration of Traditional Chinese Medicine, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, China
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Xiuyun Yin
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, China
- Drug Research and Development Center, Wannan Medical College, Wuhu, China
| | - Zhuo Chen
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, China
- Drug Research and Development Center, Wannan Medical College, Wuhu, China
| | - Di Cao
- Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, China
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, China
- Drug Research and Development Center, Wannan Medical College, Wuhu, China
| | - Xuebin Shen
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, China
- Drug Research and Development Center, Wannan Medical College, Wuhu, China
| | - Xuefeng Hou
- Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, China
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, China
- Drug Research and Development Center, Wannan Medical College, Wuhu, China
| | - Jun Han
- Pharmacology 3rd Grade Laboratory of the State Administration of Traditional Chinese Medicine, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Wannan Medical College, Wuhu, China
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, China
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, China
- Drug Research and Development Center, Wannan Medical College, Wuhu, China
| |
Collapse
|
10
|
Eitelmann S, Everaerts K, Petersilie L, Rose CR, Stephan J. Ca 2+-dependent rapid uncoupling of astrocytes upon brief metabolic stress. Front Cell Neurosci 2023; 17:1151608. [PMID: 37886111 PMCID: PMC10598858 DOI: 10.3389/fncel.2023.1151608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 08/23/2023] [Indexed: 10/28/2023] Open
Abstract
Astrocytic gap junctional coupling is a major element in neuron-glia interaction. There is strong evidence that impaired coupling is involved in neurological disorders. Reduced coupling was, e.g., demonstrated for core regions of ischemic stroke that suffer from massive cell death. In the surrounding penumbra, cells may recover, but recovery is hampered by spreading depolarizations, which impose additional metabolic stress onto the tissue. Spreading depolarizations are characterized by transient breakdown of cellular ion homeostasis, including pH and Ca2+, which might directly affect gap junctional coupling. Here, we exposed acute mouse neocortical tissue slices to brief metabolic stress and examined its effects on the coupling strength between astrocytes. Changes in gap junctional coupling were assessed by recordings of the syncytial isopotentiality. Moreover, quantitative ion imaging was performed in astrocytes to analyze the mechanisms triggering the observed changes. Our experiments show that a 2-minute perfusion of tissue slices with blockers of glycolysis and oxidative phosphorylation causes a rapid uncoupling in half of the recorded cells. They further indicate that uncoupling is not mediated by the accompanying (moderate) intracellular acidification. Dampening large astrocytic Ca2+ loads by removal of extracellular Ca2+ or blocking Ca2+ influx pathways as well as a pharmacological inhibition of calmodulin, however, prevent the uncoupling. Taken together, we conclude that astrocytes exposed to brief episodes of metabolic stress can undergo a rapid, Ca2+/calmodulin-dependent uncoupling. Such uncoupling may help to confine and reduce cellular damage in the ischemic penumbra in vivo.
Collapse
Affiliation(s)
| | | | | | - Christine R. Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jonathan Stephan
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
11
|
Wu X, Li JR, Fu Y, Chen DY, Nie H, Tang ZP. From static to dynamic: live observation of the support system after ischemic stroke by two photon-excited fluorescence laser-scanning microscopy. Neural Regen Res 2023; 18:2093-2107. [PMID: 37056116 PMCID: PMC10328295 DOI: 10.4103/1673-5374.369099] [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: 10/14/2022] [Revised: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 02/17/2023] Open
Abstract
Ischemic stroke is one of the most common causes of mortality and disability worldwide. However, treatment efficacy and the progress of research remain unsatisfactory. As the critical support system and essential components in neurovascular units, glial cells and blood vessels (including the blood-brain barrier) together maintain an optimal microenvironment for neuronal function. They provide nutrients, regulate neuronal excitability, and prevent harmful substances from entering brain tissue. The highly dynamic networks of this support system play an essential role in ischemic stroke through processes including brain homeostasis, supporting neuronal function, and reacting to injuries. However, most studies have focused on postmortem animals, which inevitably lack critical information about the dynamic changes that occur after ischemic stroke. Therefore, a high-precision technique for research in living animals is urgently needed. Two-photon fluorescence laser-scanning microscopy is a powerful imaging technique that can facilitate live imaging at high spatiotemporal resolutions. Two-photon fluorescence laser-scanning microscopy can provide images of the whole-cortex vascular 3D structure, information on multicellular component interactions, and provide images of structure and function in the cranial window. This technique shifts the existing research paradigm from static to dynamic, from flat to stereoscopic, and from single-cell function to multicellular intercommunication, thus providing direct and reliable evidence to identify the pathophysiological mechanisms following ischemic stroke in an intact brain. In this review, we discuss exciting findings from research on the support system after ischemic stroke using two-photon fluorescence laser-scanning microscopy, highlighting the importance of dynamic observations of cellular behavior and interactions in the networks of the brain's support systems. We show the excellent application prospects and advantages of two-photon fluorescence laser-scanning microscopy and predict future research developments and directions in the study of ischemic stroke.
Collapse
Affiliation(s)
- Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jia-Rui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yu Fu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Dan-Yang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hao Nie
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhou-Ping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
12
|
Pape N, Rose CR. Activation of TRPV4 channels promotes the loss of cellular ATP in organotypic slices of the mouse neocortex exposed to chemical ischemia. J Physiol 2023; 601:2975-2990. [PMID: 37195195 DOI: 10.1113/jp284430] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/15/2023] [Indexed: 05/18/2023] Open
Abstract
The vertebrate brain has an exceptionally high energy need. During ischemia, intracellular ATP concentrations decline rapidly, resulting in the breakdown of ion gradients and cellular damage. Here, we employed the nanosensor ATeam1.03YEMK to analyse the pathways driving the loss of ATP upon transient metabolic inhibition in neurons and astrocytes of the mouse neocortex. We demonstrate that brief chemical ischemia, induced by combined inhibition of glycolysis and oxidative phosphorylation, results in a transient decrease in intracellular ATP. Neurons experienced a larger relative decline and showed less ability to recover from prolonged (>5 min) metabolic inhibition than astrocytes. Blocking voltage-gated Na+ channels or NMDA receptors ameliorated the ATP decline in neurons and astrocytes, while blocking glutamate uptake aggravated the overall reduction in neuronal ATP, confirming the central role of excitatory neuronal activity in the cellular energy loss. Unexpectedly, pharmacological inhibition of transient receptor potential vanilloid 4 (TRPV4) channels significantly reduced the ischemia-induced decline in ATP in both cell types. Imaging with Na+ -sensitive indicator dye ING-2 furthermore showed that TRPV4 inhibition also reduced ischemia-induced increases in intracellular Na+ . Altogether, our results demonstrate that neurons exhibit a higher vulnerability to brief metabolic inhibition than astrocytes. Moreover, they reveal an unexpected strong contribution of TRPV4 channels to the loss of cellular ATP and suggest that the demonstrated TRPV4-related ATP consumption is most likely a direct consequence of Na+ influx. Activation of TRPV4 channels thus provides a hitherto unacknowledged contribution to the cellular energy loss during energy failure, generating a significant metabolic cost in ischemic conditions. KEY POINTS: In the ischemic brain, cellular ATP concentrations decline rapidly, which results in the collapse of ion gradients and promotes cellular damage and death. We analysed the pathways driving the loss of ATP upon transient metabolic inhibition in neurons and astrocytes of the mouse neocortex. Our results confirm the central role of excitatory neuronal activity in the cellular energy loss and demonstrate that neurons experience a larger decline in ATP and are more vulnerable to brief metabolic stress than astrocytes. Our study also reveals a new, previously unknown involvement of osmotically activated transient receptor potential vanilloid 4 (TRPV4) channels to the reduction in cellular ATP in both cell types and indicates that this is a consequence of TRPV4-mediated Na+ influx. We conclude that activation of TRPV4 channels provides a considerable contribution to the cellular energy loss, thereby generating a significant metabolic cost in ischemic conditions.
Collapse
Affiliation(s)
- Nils Pape
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstraße 1, Düsseldorf, Germany
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstraße 1, Düsseldorf, Germany
| |
Collapse
|
13
|
Fomitcheva IV, Sword J, Shi Y, Kirov SA. Plasticity of perisynaptic astroglia during ischemia-induced spreading depolarization. Cereb Cortex 2023; 33:5469-5483. [PMID: 36368909 PMCID: PMC10152098 DOI: 10.1093/cercor/bhac434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/13/2022] Open
Abstract
High astroglial capacity for glutamate and potassium clearance aids in recovering spreading depolarization (SD)-evoked disturbance of ion homeostasis during stroke. Since perisynaptic astroglia cannot be imaged with diffraction-limited light microscopy, nothing is known about the impact of SD on the ultrastructure of a tripartite synapse. We used serial section electron microscopy to assess astroglial synaptic coverage in the sensorimotor cortex of urethane-anesthetized male and female mice during and after SD evoked by transient bilateral common carotid artery occlusion. At the subcellular level, astroglial mitochondria were remarkably resilient to SD compared to dendritic mitochondria that were fragmented by SD. Overall, 482 synapses in `Sham' during `SD' and `Recovery' groups were randomly selected and analyzed in 3D. Perisynaptic astroglia was present at the axon-spine interface (ASI) during SD and after recovery. Astrocytic processes were more likely found at large synapses on mushroom spines after recovery, while the length of the ASI perimeter surrounded by astroglia has also significantly increased at large synapses. These findings suggest that as larger synapses have a bigger capacity for neurotransmitter release during SD, they attract astroglial processes to their perimeter during recovery, limiting extrasynaptic glutamate escape and further enhancing the astrocytic ability to protect synapses in stroke.
Collapse
Affiliation(s)
- Ioulia V Fomitcheva
- Department of Neurosurgery, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| | - Jeremy Sword
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| | - Yang Shi
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
- Division of Biostatistics and Data Science, Department of Population Health Sciences, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| | - Sergei A Kirov
- Department of Neurosurgery, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| |
Collapse
|
14
|
Xiong H, Tang F, Guo Y, Xu R, Lei P. Neural Circuit Changes in Neurological Disorders: Evidence from in vivo Two-photon Imaging. Ageing Res Rev 2023; 87:101933. [PMID: 37061201 DOI: 10.1016/j.arr.2023.101933] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 04/11/2023] [Indexed: 04/17/2023]
Abstract
Neural circuits, such as synaptic plasticity and neural activity, are critical components of healthy brain function. The consequent dynamic remodeling of neural circuits is an ongoing procedure affecting neuronal activities. Disruption of this essential process results in diseases. Advanced microscopic applications such as two-photon laser scanning microscopy have recently been applied to understand neural circuit changes during disease since it can visualize fine structural and functional cellular activation in living animals. In this review, we have summarized the latest work assessing the dynamic rewiring of postsynaptic dendritic spines and modulation of calcium transients in neurons of the intact living brain, focusing on their potential roles in neurological disorders (e.g. Alzheimer's disease, stroke, and epilepsy). Understanding the fine changes that occurred in the brain during disease is crucial for future clinical intervention developments.
Collapse
Affiliation(s)
- Huan Xiong
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China; Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Fei Tang
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Yujie Guo
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China.
| |
Collapse
|
15
|
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: 3.5] [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
|
16
|
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
|
17
|
Zeng ML, Kong S, Chen TX, Peng BW. Transient Receptor Potential Vanilloid 4: a Double-Edged Sword in the Central Nervous System. Mol Neurobiol 2023; 60:1232-1249. [PMID: 36434370 DOI: 10.1007/s12035-022-03141-6] [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: 08/09/2022] [Accepted: 11/17/2022] [Indexed: 11/26/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a nonselective cation channel that can be activated by diverse stimuli, such as heat, mechanical force, hypo-osmolarity, and arachidonic acid metabolites. TRPV4 is widely expressed in the central nervous system (CNS) and participates in many significant physiological processes. However, accumulative evidence has suggested that deficiency, abnormal expression or distribution, and overactivation of TRPV4 are involved in pathological processes of multiple neurological diseases. Here, we review the latest studies concerning the known features of this channel, including its expression, structure, and its physiological and pathological roles in the CNS, proposing an emerging therapeutic strategy for CNS diseases.
Collapse
Affiliation(s)
- Meng-Liu Zeng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shuo Kong
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China
| | - Tao-Xiang Chen
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China.
| |
Collapse
|
18
|
Xu Q, Zou Y, Miao Z, Jiang L, Zhao X. Transient receptor potential ion channels and cerebral stroke. Brain Behav 2023; 13:e2843. [PMID: 36527242 PMCID: PMC9847613 DOI: 10.1002/brb3.2843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
METHODS The databases Pubmed, and the National Library of Medicine were searched for literature. All papers on celebral stroke and transient receptor potential ion channels were considered. RESULTS Stroke is the second leading cause of death and disability, with an increasing incidence in developing countries. About 75 per cent of strokes are caused by occlusion of cerebral arteries, and substantial advances have been made in elucidating mechanisms how stroke affects the brain. Transient receptor potential (TRP) ion channels are calcium-permeable channels highly expressed in brain that drives Ca2+ entry into multiple cellular compartments. TRPC1/3/4/6, TRPV1/2/4, and TRPM2/4/7 channels have been implicated in stroke pathophysiology. CONCLUSIONS Although the precise mechanism of transient receptor potential ion channels in cerebral stroke is still unclear, it has the potential to be a therapeutic target for patients with stroke if developed appropriately. Hence, more research is needed to prove its efficacy in this context.
Collapse
Affiliation(s)
- Qin'yi Xu
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Yan Zou
- Department of Neurosurgery, The Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zeng'li Miao
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Lei Jiang
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Xu'dong Zhao
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
19
|
Ek-Vitorin JF, Shahidullah M, Lopez Rosales JE, Delamere NA. Patch clamp studies on TRPV4-dependent hemichannel activation in lens epithelium. Front Pharmacol 2023; 14:1101498. [PMID: 36909173 PMCID: PMC9998544 DOI: 10.3389/fphar.2023.1101498] [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: 11/17/2022] [Accepted: 02/06/2023] [Indexed: 02/26/2023] Open
Abstract
ATP release from the lens via hemichannels has been explained as a response to TRPV4 activation when the lens is subjected to osmotic swelling. To explore the apparent linkage between TRPV4 activation and connexin hemichannel opening we performed patch-clamp recordings on cultured mouse lens epithelial cells exposed to the TRPV4 agonist GSK1016790A (GSK) in the presence or absence of the TRPV4 antagonist HC067047 (HC). GSK was found to cause a fast, variable and generally large non-selective increase of whole cell membrane conductance evident as a larger membrane current (Im) over a wide voltage range. The response was prevented by HC. The GSK-induced Im increase was proportionally larger at negative voltages and coincided with fast depolarization and the simultaneous disappearance of an outward current, likely a K+ current. The presence of this outward current in control conditions appeared to be a reliable predictor of a cell's response to GSK treatment. In some studies, recordings were obtained from single cells by combining cell-attached and whole-cell patch clamp configurations. This approach revealed events with a channel conductance 180-270 pS following GSK application through the patch pipette on the cell-attached side. The findings are consistent with TRPV4-dependent opening of Cx43 hemichannels.
Collapse
Affiliation(s)
- Jose F Ek-Vitorin
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - Mohammad Shahidullah
- Department of Physiology, University of Arizona, Tucson, AZ, United States.,Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ, United States
| | | | - Nicholas A Delamere
- Department of Physiology, University of Arizona, Tucson, AZ, United States.,Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
20
|
Sucha P, Hermanova Z, Chmelova M, Kirdajova D, Camacho Garcia S, Marchetti V, Vorisek I, Tureckova J, Shany E, Jirak D, Anderova M, Vargova L. The absence of AQP4/TRPV4 complex substantially reduces acute cytotoxic edema following ischemic injury. Front Cell Neurosci 2022; 16:1054919. [PMID: 36568889 PMCID: PMC9773096 DOI: 10.3389/fncel.2022.1054919] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Astrocytic Aquaporin 4 (AQP4) and Transient receptor potential vanilloid 4 (TRPV4) channels form a functional complex that likely influences cell volume regulation, the development of brain edema, and the severity of the ischemic injury. However, it remains to be fully elucidated whether blocking these channels can serve as a therapeutic approach to alleviate the consequences of having a stroke. Methods and results In this study, we used in vivo magnetic resonance imaging (MRI) to quantify the extent of brain lesions one day (D1) and seven days (D7) after permanent middle cerebral artery occlusion (pMCAO) in AQP4 or TRPV4 knockouts and mice with simultaneous deletion of both channels. Our results showed that deletion of AQP4 or TRPV4 channels alone leads to a significant worsening of ischemic brain injury at both time points, whereas their simultaneous deletion results in a smaller brain lesion at D1 but equal tissue damage at D7 when compared with controls. Immunohistochemical analysis 7 days after pMCAO confirmed the MRI data, as the brain lesion was significantly greater in AQP4 or TRPV4 knockouts than in controls and double knockouts. For a closer inspection of the TRPV4 and AQP4 channel complex in the development of brain edema, we applied a real-time iontophoretic method in situ to determine ECS diffusion parameters, namely volume fraction (α) and tortuosity (λ). Changes in these parameters reflect alterations in cell volume, and tissue structure during exposure of acute brain slices to models of ischemic conditions in situ, such as oxygen-glucose deprivation (OGD), hypoosmotic stress, or hyperkalemia. The decrease in α was comparable in double knockouts and controls when exposed to hypoosmotic stress or hyperkalemia. However, during OGD, there was no decrease in α in the double knockouts as observed in the controls, which suggests less swelling of the cellular components of the brain. Conclusion Although simultaneous deletion of AQP4 and TRPV4 did not improve the overall outcome of ischemic brain injury, our data indicate that the interplay between AQP4 and TRPV4 channels plays a critical role during neuronal and non-neuronal swelling in the acute phase of ischemic injury.
Collapse
Affiliation(s)
- Petra Sucha
- Second Faculty of Medicine, Charles University, Prague, Czechia,Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czechia
| | - Zuzana Hermanova
- Second Faculty of Medicine, Charles University, Prague, Czechia,Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czechia
| | - Martina Chmelova
- Second Faculty of Medicine, Charles University, Prague, Czechia,Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czechia
| | - Denisa Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czechia
| | - Sara Camacho Garcia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czechia
| | - Valeria Marchetti
- Second Faculty of Medicine, Charles University, Prague, Czechia,Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czechia
| | - Ivan Vorisek
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czechia
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czechia
| | - Eyar Shany
- Department of Diagnostic and Interventional Radiology, Institute of Clinical and Experimental Medicine, Prague, Czechia
| | - Daniel Jirak
- Department of Diagnostic and Interventional Radiology, Institute of Clinical and Experimental Medicine, Prague, Czechia,First Faculty of Medicine, Institute of Biophysics and Informatics, Charles University, Prague, Czechia
| | - Miroslava Anderova
- Second Faculty of Medicine, Charles University, Prague, Czechia,Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czechia,*Correspondence: Miroslava Anderova,
| | - Lydia Vargova
- Second Faculty of Medicine, Charles University, Prague, Czechia,Department of Cellular Neurophysiology, Institute of Experimental Medicine of the CAS, Prague, Czechia
| |
Collapse
|
21
|
Kazandzhieva K, Mammadova-Bach E, Dietrich A, Gudermann T, Braun A. TRP channel function in platelets and megakaryocytes: basic mechanisms and pathophysiological impact. Pharmacol Ther 2022; 237:108164. [PMID: 35247518 DOI: 10.1016/j.pharmthera.2022.108164] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/29/2022] [Accepted: 02/28/2022] [Indexed: 12/30/2022]
Abstract
Transient receptor potential (TRP) proteins form a superfamily of cation channels that are expressed in a wide range of tissues and cell types. During the last years, great progress has been made in understanding the molecular complexity and the functions of TRP channels in diverse cellular processes, including cell proliferation, migration, adhesion and activation. The diversity of functions depends on multiple regulatory mechanisms by which TRP channels regulate Ca2+ entry mechanisms and intracellular Ca2+ dynamics, either through membrane depolarization involving cation influx or store- and receptor-operated mechanisms. Abnormal function or expression of TRP channels results in vascular pathologies, including hypertension, ischemic stroke and inflammatory disorders through effects on vascular cells, including the components of blood vessels and platelets. Moreover, some TRP family members also regulate megakaryopoiesis and platelet production, indicating a complex role of TRP channels in pathophysiological conditions. In this review, we describe potential roles of TRP channels in megakaryocytes and platelets, as well as their contribution to diseases such as thrombocytopenia, thrombosis and stroke. We also critically discuss the potential of TRP channels as possible targets for disease prevention and treatment.
Collapse
Affiliation(s)
- Kalina Kazandzhieva
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Elmina Mammadova-Bach
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany; Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Alexander Dietrich
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany; German Center for Lung Research (DZL), Munich, Germany
| | - Thomas Gudermann
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany; German Center for Lung Research (DZL), Munich, Germany.
| | - Attila Braun
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
22
|
Eitelmann S, Stephan J, Everaerts K, Durry S, Pape N, Gerkau NJ, Rose CR. Changes in Astroglial K + upon Brief Periods of Energy Deprivation in the Mouse Neocortex. Int J Mol Sci 2022; 23:ijms23094836. [PMID: 35563238 PMCID: PMC9102782 DOI: 10.3390/ijms23094836] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
Malfunction of astrocytic K+ regulation contributes to the breakdown of extracellular K+ homeostasis during ischemia and spreading depolarization events. Studying astroglial K+ changes is, however, hampered by a lack of suitable techniques. Here, we combined results from fluorescence imaging, ion-selective microelectrodes, and patch-clamp recordings in murine neocortical slices with the calculation of astrocytic [K+]. Brief chemical ischemia caused a reversible ATP reduction and a transient depolarization of astrocytes. Moreover, astrocytic [Na+] increased by 24 mM and extracellular [Na+] decreased. Extracellular [K+] increased, followed by an undershoot during recovery. Feeding these data into the Goldman-Hodgkin-Katz equation revealed a baseline astroglial [K+] of 146 mM, an initial K+ loss by 43 mM upon chemical ischemia, and a transient K+ overshoot of 16 mM during recovery. It also disclosed a biphasic mismatch in astrocytic Na+/K+ balance, which was initially ameliorated, but later aggravated by accompanying changes in pH and bicarbonate, respectively. Altogether, our study predicts a loss of K+ from astrocytes upon chemical ischemia followed by a net gain. The overshooting K+ uptake will promote low extracellular K+ during recovery, likely exerting a neuroprotective effect. The resulting late cation/anion imbalance requires additional efflux of cations and/or influx of anions, the latter eventually driving delayed astrocyte swelling.
Collapse
|
23
|
Aisenberg WH, McCray BA, Sullivan JM, Diehl E, DeVine LR, Alevy J, Bagnell AM, Carr P, Donohue JK, Goretzki B, Cole RN, Hellmich UA, Sumner CJ. Multiubiquitination of TRPV4 reduces channel activity independent of surface localization. J Biol Chem 2022; 298:101826. [PMID: 35300980 PMCID: PMC9010760 DOI: 10.1016/j.jbc.2022.101826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
Ubiquitin (Ub)-mediated regulation of plasmalemmal ion channel activity canonically occurs via stimulation of endocytosis. Whether ubiquitination can modulate channel activity by alternative mechanisms remains unknown. Here, we show that the transient receptor potential vanilloid 4 (TRPV4) cation channel is multiubiquitinated within its cytosolic N-terminal and C-terminal intrinsically disordered regions (IDRs). Mutagenizing select lysine residues to block ubiquitination of the N-terminal but not C-terminal IDR resulted in a marked elevation of TRPV4-mediated intracellular calcium influx, without increasing cell surface expression levels. Conversely, enhancing TRPV4 ubiquitination via expression of an E3 Ub ligase reduced TRPV4 channel activity but did not decrease plasma membrane abundance. These results demonstrate Ub-dependent regulation of TRPV4 channel function independent of effects on plasma membrane localization. Consistent with ubiquitination playing a key negative modulatory role of the channel, gain-of-function neuropathy-causing mutations in the TRPV4 gene led to reduced channel ubiquitination in both cellular and Drosophila models of TRPV4 neuropathy, whereas increasing mutant TRPV4 ubiquitination partially suppressed channel overactivity. Together, these data reveal a novel mechanism via which ubiquitination of an intracellular flexible IDR domain modulates ion channel function independently of endocytic trafficking and identify a contributory role for this pathway in the dysregulation of TRPV4 channel activity by neuropathy-causing mutations.
Collapse
Affiliation(s)
- William H Aisenberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brett A McCray
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeremy M Sullivan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Erika Diehl
- Department of Chemistry, Biochemistry Section, Johannes Gutenberg-Universität Mainz, Mainz, Germany
| | - Lauren R DeVine
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jonathan Alevy
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anna M Bagnell
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Patrice Carr
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jack K Donohue
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Benedikt Goretzki
- Institute of Organic Chemistry and Macromolecular Chemistry, Cluster of Excellence 'Balance of the Microverse', Friedrich-Schiller-Universität, Jena, Germany; Center for Biomolecular Magnetic Resonance (BMRZ), Goethe-Universität, Frankfurt am Main, Germany
| | - Robert N Cole
- Institute of Organic Chemistry and Macromolecular Chemistry, Cluster of Excellence 'Balance of the Microverse', Friedrich-Schiller-Universität, Jena, Germany
| | - Ute A Hellmich
- Institute of Organic Chemistry and Macromolecular Chemistry, Cluster of Excellence 'Balance of the Microverse', Friedrich-Schiller-Universität, Jena, Germany; Center for Biomolecular Magnetic Resonance (BMRZ), Goethe-Universität, Frankfurt am Main, Germany
| | - Charlotte J Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
24
|
Salman MM, Kitchen P, Halsey A, Wang MX, Törnroth-Horsefield S, Conner AC, Badaut J, Iliff JJ, Bill RM. Emerging roles for dynamic aquaporin-4 subcellular relocalization in CNS water homeostasis. Brain 2022; 145:64-75. [PMID: 34499128 PMCID: PMC9088512 DOI: 10.1093/brain/awab311] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 06/28/2021] [Accepted: 07/31/2021] [Indexed: 11/25/2022] Open
Abstract
Aquaporin channels facilitate bidirectional water flow in all cells and tissues. AQP4 is highly expressed in astrocytes. In the CNS, it is enriched in astrocyte endfeet, at synapses, and at the glia limitans, where it mediates water exchange across the blood-spinal cord and blood-brain barriers (BSCB/BBB), and controls cell volume, extracellular space volume, and astrocyte migration. Perivascular enrichment of AQP4 at the BSCB/BBB suggests a role in glymphatic function. Recently, we have demonstrated that AQP4 localization is also dynamically regulated at the subcellular level, affecting membrane water permeability. Ageing, cerebrovascular disease, traumatic CNS injury, and sleep disruption are established and emerging risk factors in developing neurodegeneration, and in animal models of each, impairment of glymphatic function is associated with changes in perivascular AQP4 localization. CNS oedema is caused by passive water influx through AQP4 in response to osmotic imbalances. We have demonstrated that reducing dynamic relocalization of AQP4 to the BSCB/BBB reduces CNS oedema and accelerates functional recovery in rodent models. Given the difficulties in developing pore-blocking AQP4 inhibitors, targeting AQP4 subcellular localization opens up new treatment avenues for CNS oedema, neurovascular and neurodegenerative diseases, and provides a framework to address fundamental questions about water homeostasis in health and disease.
Collapse
Affiliation(s)
- Mootaz M Salman
- Department of Physiology, Anatomy and Genetics,
University of Oxford, Oxford OX1 3PT, UK
| | - Philip Kitchen
- School of Biosciences, College of Health and Life
Sciences, Aston University, Aston Triangle,
Birmingham B4 7ET, UK
| | - Andrea Halsey
- Institute of Clinical Sciences, College of Medical
and Dental Sciences, University of Birmingham,
Edgbaston, Birmingham B15 2TT, UK
| | - Marie Xun Wang
- Department of Psychiatry and Behavioral Sciences,
University of Washington School of Medicine, Seattle, WA, USA
| | | | - Alex C Conner
- Institute of Clinical Sciences, College of Medical
and Dental Sciences, University of Birmingham,
Edgbaston, Birmingham B15 2TT, UK
| | - Jerome Badaut
- CNRS-UMR 5536-Centre de Résonance
Magnétique des systèmes Biologiques, Université de
Bordeaux, 33076 Bordeaux, France
| | - Jeffrey J Iliff
- Department of Psychiatry and Behavioral Sciences,
University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, University of Washington
School of Medicine, Seattle, WA, USA
- VISN 20 Mental Illness Research, Education and
Clinical Center, VA Puget Sound Health Care System, Seattle, WA,
USA
| | - Roslyn M Bill
- School of Biosciences, College of Health and Life
Sciences, Aston University, Aston Triangle,
Birmingham B4 7ET, UK
| |
Collapse
|
25
|
Meyer J, Gerkau NJ, Kafitz KW, Patting M, Jolmes F, Henneberger C, Rose CR. Rapid Fluorescence Lifetime Imaging Reveals That TRPV4 Channels Promote Dysregulation of Neuronal Na + in Ischemia. J Neurosci 2022; 42:552-566. [PMID: 34872928 PMCID: PMC8805620 DOI: 10.1523/jneurosci.0819-21.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/28/2021] [Accepted: 11/17/2021] [Indexed: 11/21/2022] Open
Abstract
Fluorescence imaging is an indispensable method for analysis of diverse cellular and molecular processes, enabling, for example, detection of ions, second messengers, or metabolites. Intensity-based approaches, however, are prone to artifacts introduced by changes in fluorophore concentrations. This drawback can be overcome by fluorescence lifetime imaging (FLIM) based on time-correlated single-photon counting. FLIM often necessitates long photon collection times, resulting in strong temporal binning of dynamic processes. Recently, rapidFLIM was introduced, exploiting ultra-low dead-time photodetectors together with rapid electronics. Here, we demonstrate the applicability of rapidFLIM, combined with new and improved correction schemes, for spatiotemporal fluorescence lifetime imaging of low-emission fluorophores in a biological system. Using tissue slices of hippocampi of mice of either sex, loaded with the Na+ indicator ING2, we show that improved rapidFLIM enables quantitative, dynamic imaging of neuronal Na+ signals at a full-frame temporal resolution of 0.5 Hz. Induction of transient chemical ischemia resulted in unexpectedly large Na+ influx, accompanied by considerable cell swelling. Both Na+ loading and cell swelling were dampened on inhibition of TRPV4 channels. Together, rapidFLIM enabled the spatiotemporal visualization and quantification of neuronal Na+ transients at unprecedented speed and independent from changes in cell volume. Moreover, our experiments identified TRPV4 channels as hitherto unappreciated contributors to neuronal Na+ loading on metabolic failure, suggesting this pathway as a possible target to ameliorate excitotoxic damage. Finally, rapidFLIM will allow faster and more sensitive detection of a wide range of dynamic signals with other FLIM probes, most notably those with intrinsic low-photon emission.SIGNIFICANCE STATEMENT FLIM is an indispensable method for analysis of cellular processes. FLIM often necessitates long photon collection periods, requiring the sacrifice of temporal resolution at the expense of spatial information. Here, we demonstrate the applicability of the recently introduced rapidFLIM for quantitative, dynamic imaging with low-emission fluorophores in brain slices. RapidFLIM, combined with improved correction schemes, enabled intensity-independent recording of neuronal Na+ transients at unprecedented full-frame rates of 0.5 Hz. It also allowed quantitative imaging independent from changes in cell volume, revealing a surprisingly strong and hitherto uncovered contribution of TRPV4 channels to Na+ loading on energy failure. Collectively, our study thus provides a novel, unexpected insight into the mechanisms that are responsible for Na+ changes on energy depletion.
Collapse
Affiliation(s)
- Jan Meyer
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Niklas J Gerkau
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Karl W Kafitz
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | | | | | - Christian Henneberger
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases, 53175 Bonn, Germany
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, England
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
26
|
Michinaga S, Onishi K, Shimizu K, Mizuguchi H, Hishinuma S. Pharmacological Inhibition of Transient Receptor Potential Vanilloid 4 Reduces Vasogenic Edema after Traumatic Brain Injury in Mice. Biol Pharm Bull 2021; 44:1759-1766. [PMID: 34719652 DOI: 10.1248/bpb.b21-00512] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vasogenic edema results from blood-brain barrier (BBB) disruption after traumatic brain injury (TBI), and although it can be fatal, no promising therapeutic drugs have been developed as yet. Transient receptor potential vanilloid 4 (TRPV4) is a calcium-permeable channel that is sensitive to temperature and osmotic pressure. As TRPV4 is known to be responsible for various pathological conditions following brain injury, we investigated the effects of pharmacological TRPV4 antagonists on TBI-induced vasogenic edema in this study. A TBI model was established by inflicting fluid percussion injury (FPI) in the mouse cerebrum and cultured astrocytes. Vasogenic brain edema and BBB disruption were assessed based on brain water content and Evans blue (EB) extravasation into brain tissue, respectively. After FPI, brain water content and EB extravasation increased. Repeated intracerebroventricular administration of the specific TRPV4 antagonists HC-067047 and RN-1734 dose-dependently reduced brain water content and alleviated EB extravasation in FPI mice. Additionally, real-time PCR analysis indicated that administration of HC-067047 and RN-1734 reversed the FPI-induced increase in mRNA levels of endogenous causal factors for BBB disruption, including matrix metalloproteinase-9 (MMP-9), vascular endothelial growth factor-A (VEGF-A), and endothelin-1 (ET-1). In astrocytes, TRPV4 level was observed to be higher than that in brain microvascular endothelial cells. Treatment with HC-067047 and RN-1734 inhibited the increase in mRNA levels of MMP-9, VEGF-A, and ET-1 in cultured astrocytes subjected to in vitro FPI. These results suggest that pharmacological inhibition of TRPV4 is expected to be a promising therapeutic strategy for treating TBI-induced vasogenic edema.
Collapse
Affiliation(s)
| | - Kazuya Onishi
- Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University
| | - Kahori Shimizu
- Laboratory of Biochemistry, Faculty of Pharmacy, Osaka Ohtani University
| | - Hiroyuki Mizuguchi
- Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University
| | | |
Collapse
|
27
|
Engels M, Kalia M, Rahmati S, Petersilie L, Kovermann P, van Putten MJAM, Rose CR, Meijer HGE, Gensch T, Fahlke C. Glial Chloride Homeostasis Under Transient Ischemic Stress. Front Cell Neurosci 2021; 15:735300. [PMID: 34602981 PMCID: PMC8481871 DOI: 10.3389/fncel.2021.735300] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
High water permeabilities permit rapid adjustments of glial volume upon changes in external and internal osmolarity, and pathologically altered intracellular chloride concentrations ([Cl–]int) and glial cell swelling are often assumed to represent early events in ischemia, infections, or traumatic brain injury. Experimental data for glial [Cl–]int are lacking for most brain regions, under normal as well as under pathological conditions. We measured [Cl–]int in hippocampal and neocortical astrocytes and in hippocampal radial glia-like (RGL) cells in acute murine brain slices using fluorescence lifetime imaging microscopy with the chloride-sensitive dye MQAE at room temperature. We observed substantial heterogeneity in baseline [Cl–]int, ranging from 14.0 ± 2.0 mM in neocortical astrocytes to 28.4 ± 3.0 mM in dentate gyrus astrocytes. Chloride accumulation by the Na+-K+-2Cl– cotransporter (NKCC1) and chloride outward transport (efflux) through K+-Cl– cotransporters (KCC1 and KCC3) or excitatory amino acid transporter (EAAT) anion channels control [Cl–]int to variable extent in distinct brain regions. In hippocampal astrocytes, blocking NKCC1 decreased [Cl–]int, whereas KCC or EAAT anion channel inhibition had little effect. In contrast, neocortical astrocytic or RGL [Cl–]int was very sensitive to block of chloride outward transport, but not to NKCC1 inhibition. Mathematical modeling demonstrated that higher numbers of NKCC1 and KCC transporters can account for lower [Cl–]int in neocortical than in hippocampal astrocytes. Energy depletion mimicking ischemia for up to 10 min did not result in pronounced changes in [Cl–]int in any of the tested glial cell types. However, [Cl–]int changes occurred under ischemic conditions after blocking selected anion transporters. We conclude that stimulated chloride accumulation and chloride efflux compensate for each other and prevent glial swelling under transient energy deprivation.
Collapse
Affiliation(s)
- Miriam Engels
- Institute of Biological Information Processing, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| | - Manu Kalia
- Applied Analysis, Department of Applied Mathematics, University of Twente, Enschede, Netherlands.,Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sarah Rahmati
- Institute of Biological Information Processing, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| | - Laura Petersilie
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Peter Kovermann
- Institute of Biological Information Processing, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| | | | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hil G E Meijer
- Applied Analysis, Department of Applied Mathematics, University of Twente, Enschede, Netherlands
| | - Thomas Gensch
- Institute of Biological Information Processing, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| | - Christoph Fahlke
- Institute of Biological Information Processing, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
28
|
Control of membrane protein homeostasis by a chaperone-like glial cell adhesion molecule at multiple subcellular locations. Sci Rep 2021; 11:18435. [PMID: 34531445 PMCID: PMC8446001 DOI: 10.1038/s41598-021-97777-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/24/2021] [Indexed: 01/17/2023] Open
Abstract
The significance of crosstalks among constituents of plasma membrane protein clusters/complexes in cellular proteostasis and protein quality control (PQC) remains incompletely understood. Examining the glial (enriched) cell adhesion molecule (CAM), we demonstrate its chaperone-like role in the biosynthetic processing of the megalencephalic leukoencephalopathy with subcortical cyst 1 (MLC1)-heteromeric regulatory membrane protein complex, as well as the function of the GlialCAM/MLC1 signalling complex. We show that in the absence of GlialCAM, newly synthesized MLC1 molecules remain unfolded and are susceptible to polyubiquitination-dependent proteasomal degradation at the endoplasmic reticulum. At the plasma membrane, GlialCAM regulates the diffusional partitioning and endocytic dynamics of cluster members, including the ClC-2 chloride channel and MLC1. Impaired folding and/or expression of GlialCAM or MLC1 in the presence of diseases causing mutations, as well as plasma membrane tethering compromise the functional expression of the cluster, leading to compromised endo-lysosomal organellar identity. In addition, the enlarged endo-lysosomal compartments display accelerated acidification, ubiquitinated cargo-sorting and impaired endosomal recycling. Jointly, these observations indicate an essential and previously unrecognized role for CAM, where GliaCAM functions as a PQC factor for the MLC1 signalling complex biogenesis and possess a permissive role in the membrane dynamic and cargo sorting functions with implications in modulations of receptor signalling.
Collapse
|
29
|
Xie Q, Ma R, Li H, Wang J, Guo X, Chen H. Advancement in research on the role of the transient receptor potential vanilloid channel in cerebral ischemic injury (Review). Exp Ther Med 2021; 22:881. [PMID: 34194559 PMCID: PMC8237269 DOI: 10.3892/etm.2021.10313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 05/28/2021] [Indexed: 01/04/2023] Open
Abstract
Stroke is a common critical disease occurring in middle-aged and elderly individuals, and is characterized by high morbidity, lethality and mortality. As such, it is of great concern to medical professionals. The aim of the present review was to investigate the effects of transient receptor potential vanilloid (TRPV) subtypes during cerebral ischemia in ischemia-reperfusion animal models, oxygen glucose deprivation and in other administration cell models in vitro to explore new avenues for stroke research and clinical treatments. TRPV1, TRPV2 and TRPV4 employ different methodologies by which they confer protection against cerebral ischemic injury. TRPV1 and TRPV4 are likely related to the inhibition of inflammatory reactions, neurotoxicity and cell apoptosis, thus promoting nerve growth and regulation of intracellular calcium ions (Ca2+). The mechanisms of neuroprotection of TRPV1 are the JNK pathway, N-methyl-D-aspartate (NMDA) receptor and therapeutic hypothermia. The mechanisms of neuroprotection of TRPV4 are the PI3K/Akt pathways, NMDA receptor and p38 MAPK pathway, amongst others. The mechanisms by which TRPV2 confers its protective effects are predominantly connected with the regulation of nerve growth factor, MAPK and JNK pathways, as well as JNK-dependent pathways. Thus, TRPVs have the potential for improving outcomes associated with cerebral ischemic or reperfusion injuries. The protection conferred by TRPV1 and TRPV4 is closely related to cellular Ca2+ influx, while TRPV2 has a different target and mode of action, possibly due to its expression sites. However, in light of certain contradictory research conclusions, further experimentation is required to clarify the mechanisms and specific pathways by which TRPVs act to alleviate nerve injuries.
Collapse
Affiliation(s)
- Qian Xie
- School of Pharmacy and State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
| | - Rong Ma
- School of Pharmacy and State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
| | - Hongyan Li
- School of Pharmacy and State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
| | - Jian Wang
- School of Pharmacy and State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
| | - Xiaoqing Guo
- School of Pharmacy and State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
| | - Hai Chen
- School of Pharmacy and State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
| |
Collapse
|
30
|
Abstract
Our brains consist of 80% water, which is continuously shifted between different compartments and cell types during physiological and pathophysiological processes. Disturbances in brain water homeostasis occur with pathologies such as brain oedema and hydrocephalus, in which fluid accumulation leads to elevated intracranial pressure. Targeted pharmacological treatments do not exist for these conditions owing to our incomplete understanding of the molecular mechanisms governing brain water transport. Historically, the transmembrane movement of brain water was assumed to occur as passive movement of water along the osmotic gradient, greatly accelerated by water channels termed aquaporins. Although aquaporins govern the majority of fluid handling in the kidney, they do not suffice to explain the overall brain water movement: either they are not present in the membranes across which water flows or they appear not to be required for the observed flow of water. Notably, brain fluid can be secreted against an osmotic gradient, suggesting that conventional osmotic water flow may not describe all transmembrane fluid transport in the brain. The cotransport of water is an unconventional molecular mechanism that is introduced in this Review as a missing link to bridge the gap in our understanding of cellular and barrier brain water transport.
Collapse
Affiliation(s)
- Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
31
|
Kim KJ, Diaz JR, Presa JL, Muller PR, Brands MW, Khan MB, Hess DC, Althammer F, Stern JE, Filosa JA. Decreased parenchymal arteriolar tone uncouples vessel-to-neuronal communication in a mouse model of vascular cognitive impairment. GeroScience 2021; 43:1405-1422. [PMID: 33410092 PMCID: PMC8190257 DOI: 10.1007/s11357-020-00305-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/22/2020] [Indexed: 01/18/2023] Open
Abstract
Chronic hypoperfusion is a key contributor to cognitive decline and neurodegenerative conditions, but the cellular mechanisms remain ill-defined. Using a multidisciplinary approach, we sought to elucidate chronic hypoperfusion-evoked functional changes at the neurovascular unit. We used bilateral common carotid artery stenosis (BCAS), a well-established model of vascular cognitive impairment, combined with an ex vivo preparation that allows pressurization of parenchymal arterioles in a brain slice. Our results demonstrate that mild (~ 30%), chronic hypoperfusion significantly altered the functional integrity of the cortical neurovascular unit. Although pial cerebral perfusion recovered over time, parenchymal arterioles progressively lost tone, exhibiting significant reductions by day 28 post-surgery. We provide supportive evidence for reduced adenosine 1 receptor-mediated vasoconstriction as a potential mechanism in the adaptive response underlying the reduced baseline tone in parenchymal arterioles. In addition, we show that in response to the neuromodulator adenosine, the action potential frequency of cortical pyramidal neurons was significantly reduced in all groups. However, a significant decrease in adenosine-induced hyperpolarization was observed in BCAS 14 days. At the microvascular level, constriction-induced inhibition of pyramidal neurons was significantly compromised in BCAS mice. Collectively, these results suggest that BCAS uncouples vessel-to-neuron communication-vasculo-neuronal coupling-a potential early event in cognitive decline.
Collapse
Affiliation(s)
- Ki Jung Kim
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Juan Ramiro Diaz
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Jessica L Presa
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - P Robinson Muller
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Michael W Brands
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Mohammad B Khan
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | | | - Javier E Stern
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Jessica A Filosa
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
32
|
Zhao Y, Wang S, Song X, Yuan J, Qi D, Gu X, Yin MY, Han Z, Zhu Y, Liu Z, Zhang Y, Wei L, Wei ZZ. Glial Cell-Based Vascular Mechanisms and Transplantation Therapies in Brain Vessel and Neurodegenerative Diseases. Front Cell Neurosci 2021; 15:627682. [PMID: 33841101 PMCID: PMC8032950 DOI: 10.3389/fncel.2021.627682] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodevelopmental and neurodegenerative diseases (NDDs) with severe neurological/psychiatric symptoms, such as cerebrovascular pathology in AD, CAA, and chronic stroke, have brought greater attention with their incidence and prevalence having markedly increased over the past few years. Causes of the significant neuropathologies, especially those observed in neurological diseases in the CNS, are commonly believed to involve multiple factors such as an age, a total environment, genetics, and an immunity contributing to their progression, neuronal, and vascular injuries. We primarily focused on the studies of glial involvement/dysfunction in part with the blood-brain barrier (BBB) and the neurovascular unit (NVU) changes, and the vascular mechanisms, which have been both suggested as critical roles in chronic stroke and many other NDDs. It has been noted that glial cells including astrocytes (which outnumber other cell types in the CNS) essentially contribute more to the BBB integrity, extracellular homeostasis, neurotransmitter release, regulation of neurogenic niches in response to neuroinflammatory stimulus, and synaptic plasticity. In a recent study for NDDs utilizing cellular and molecular biology and genetic and pharmacological tools, the role of reactive astrocytes (RACs) and gliosis was demonstrated, able to trigger pathophysiological/psychopathological detrimental changes during the disease progression. We speculate, in particular, the BBB, the NVU, and changes of the astrocytes (potentially different populations from the RACs) not only interfere with neuronal development and synaptogenesis, but also generate oxidative damages, contribute to beta-amyloid clearances and disrupted vasculature, as well as lead to neuroinflammatory disorders. During the past several decades, stem cell therapy has been investigated with a research focus to target related neuro-/vascular pathologies (cell replacement and repair) and neurological/psychiatric symptoms (paracrine protection and homeostasis). Evidence shows that transplantation of neurogenic or vasculogenic cells could be achieved to pursue differentiation and maturation within the diseased brains as expected. It would be hoped that, via regulating functions of astrocytes, astrocytic involvement, and modulation of the BBB, the NVU and astrocytes should be among major targets for therapeutics against NDDs pathogenesis by drug and cell-based therapies. The non-invasive strategies in combination with stem cell transplantation such as the well-tested intranasal deliveries for drug and stem cells by our and many other groups show great translational potentials in NDDs. Neuroimaging and clinically relevant analyzing tools need to be evaluated in various NDDs brains.
Collapse
Affiliation(s)
- Yingying Zhao
- Beijing Clinical Research Institute, Beijing, China.,Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States.,Department of Critical Care Medicine, Airport Hospital of Tianjin Medical University General Hospital, Tianjin, China
| | - Shuanglin Wang
- Department of Critical Care Medicine, Airport Hospital of Tianjin Medical University General Hospital, Tianjin, China.,Department of Cardiovascular Thoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China.,Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaopeng Song
- Mclean Imaging Center, Harvard Medical School, McLean Hospital, Belmont, MA, United States
| | - Junliang Yuan
- Mclean Imaging Center, Harvard Medical School, McLean Hospital, Belmont, MA, United States.,Department of Neurology, Institute of Mental Health, Peking University Sixth Hospital, Beijing, China
| | - Dong Qi
- Beijing Clinical Research Institute, Beijing, China
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Michael Yaoyao Yin
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States.,Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Zhou Han
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, United States
| | - Yanbing Zhu
- Beijing Clinical Research Institute, Beijing, China
| | - Zhandong Liu
- Beijing Clinical Research Institute, Beijing, China
| | - Yongbo Zhang
- Beijing Clinical Research Institute, Beijing, China
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States.,Emory Specialized Center of Sex Differences, Emory University, Atlanta, GA, United States
| |
Collapse
|
33
|
Neuropathy-causing TRPV4 mutations disrupt TRPV4-RhoA interactions and impair neurite extension. Nat Commun 2021; 12:1444. [PMID: 33664271 PMCID: PMC7933254 DOI: 10.1038/s41467-021-21699-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 02/02/2021] [Indexed: 12/19/2022] Open
Abstract
TRPV4 is a cell surface-expressed calcium-permeable cation channel that mediates cell-specific effects on cellular morphology and function. Dominant missense mutations of TRPV4 cause distinct, tissue-specific diseases, but the pathogenic mechanisms are unknown. Mutations causing peripheral neuropathy localize to the intracellular N-terminal domain whereas skeletal dysplasia mutations are in multiple domains. Using an unbiased screen, we identified the cytoskeletal remodeling GTPase RhoA as a TRPV4 interactor. TRPV4-RhoA binding occurs via the TRPV4 N-terminal domain, resulting in suppression of TRPV4 channel activity, inhibition of RhoA activation, and extension of neurites in vitro. Neuropathy but not skeletal dysplasia mutations disrupt TRPV4-RhoA binding and cytoskeletal outgrowth. However, inhibition of RhoA restores neurite length in vitro and in a fly model of TRPV4 neuropathy. Together these results identify RhoA as a critical mediator of TRPV4-induced cell structure changes and suggest that disruption of TRPV4-RhoA binding may contribute to tissue-specific toxicity of TRPV4 neuropathy mutations. TRPV4 dominant mutations cause neuropathy. Here, the authors show that TRPV4 binds and interacts with RhoA, modulating the actin cytoskeleton. Neuropathy-causing mutations of TRPV4 disrupt this complex, leading to RhoA activation and impairment of neurite extension in cultured cells and flies.
Collapse
|
34
|
Transient Receptor Potential Vanilloid in the Brain Gliovascular Unit: Prospective Targets in Therapy. Pharmaceutics 2021; 13:pharmaceutics13030334. [PMID: 33806707 PMCID: PMC7999963 DOI: 10.3390/pharmaceutics13030334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 12/25/2022] Open
Abstract
The gliovascular unit (GVU) is composed of the brain microvascular endothelial cells forming blood–brain barrier and the neighboring surrounding “mural” cells (e.g., pericytes) and astrocytes. Modulation of the GVU/BBB features could be observed in a variety of vascular, immunologic, neuro-psychiatric diseases, and cancers, which can disrupt the brain homeostasis. Ca2+ dynamics have been regarded as a major factor in determining BBB/GVU properties, and previous studies have demonstrated the role of transient receptor potential vanilloid (TRPV) channels in modulating Ca2+ and BBB/GVU properties. The physiological role of thermosensitive TRPV channels in the BBB/GVU, as well as their possible therapeutic potential as targets in treating brain diseases via preserving the BBB are reviewed. TRPV2 and TRPV4 are the most abundant isoforms in the human BBB, and TRPV2 was evidenced to play a main role in regulating human BBB integrity. Interspecies differences in TRPV2 and TRPV4 BBB expression complicate further preclinical validation. More studies are still needed to better establish the physiopathological TRPV roles such as in astrocytes, vascular smooth muscle cells, and pericytes. The effect of the chronic TRPV modulation should also deserve further studies to evaluate their benefit and innocuity in vivo.
Collapse
|
35
|
Petzold GC, Dreier JP. Spreading depolarization evoked by endothelin-1 is inhibited by octanol but not by carbenoxolone. BRAIN HEMORRHAGES 2021. [DOI: 10.1016/j.hest.2020.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
36
|
Brymer KJ, Barnes JR, Parsons MP. Entering a new era of quantifying glutamate clearance in health and disease. J Neurosci Res 2021; 99:1598-1617. [PMID: 33618436 DOI: 10.1002/jnr.24810] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/22/2021] [Accepted: 02/01/2021] [Indexed: 12/21/2022]
Abstract
Glutamate transporter proteins, expressed on both neurons and glia, serve as the main gatekeepers that dictate the spatial and temporal actions of extracellular glutamate. Glutamate is essential to the function of the healthy brain yet paradoxically contributes to the toxicity associated with many neurodegenerative diseases. Rapid transporter-mediated glutamate uptake, primarily occurring at astrocytic processes, tightens the efficiency of excitatory network activity and prevents toxic glutamate build-up in the extracellular space. Glutamate transporter dysfunction is thought to underlie myriad central nervous system (CNS) diseases including Alzheimer and Huntington disease. Over the past few decades, techniques such as biochemical uptake assays and electrophysiological recordings of transporter currents from individual astrocytes have revealed the remarkable ability of the CNS to efficiently clear extracellular glutamate. In more recent years, the rapidly evolving glutamate-sensing "sniffers" now allow researchers to visualize real-time glutamate transients on a millisecond time scale with single synapse spatial resolution in defined cell populations. As we transition to an increased reliance on optical-based methods of glutamate visualization and quantification, it is of utmost importance to understand not only the advantages that glutamate biosensors bring to the table but also the associated caveats and their implications for data interpretation. In this review, we summarize the strengths and limitations of the commonly used methods to quantify glutamate uptake. We then discuss what these techniques, when viewed as a complementary whole, have told us about the brain's ability to regulate glutamate levels, in both health and in the context of neurodegenerative disease.
Collapse
Affiliation(s)
- Kyle J Brymer
- Faculty of Medicine, Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Jocelyn R Barnes
- Faculty of Medicine, Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Matthew P Parsons
- Faculty of Medicine, Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
37
|
Tirosh A, Tuncman G, Calay ES, Rathaus M, Ron I, Tirosh A, Yalcin A, Lee YG, Livne R, Ron S, Minsky N, Arruda AP, Hotamisligil GS. Intercellular Transmission of Hepatic ER Stress in Obesity Disrupts Systemic Metabolism. Cell Metab 2021; 33:319-333.e6. [PMID: 33340456 PMCID: PMC7858244 DOI: 10.1016/j.cmet.2020.11.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 07/30/2020] [Accepted: 11/12/2020] [Indexed: 12/22/2022]
Abstract
Endoplasmic reticulum stress (ERS) has a pathophysiological role in obesity-associated insulin resistance. Yet, the coordinated tissue response to ERS remains unclear. Increased connexin 43 (Cx43)-mediated intercellular communication has been implicated in tissue-adaptive and -maladaptive response to various chronic stresses. Here, we demonstrate that in hepatocytes, ERS results in increased Cx43 expression and cell-cell coupling. Co-culture of ER-stressed "donor" cells resulted in intercellular transmission of ERS and dysfunction to ERS-naive "recipient" cells ("bystander response"), which could be prevented by genetic or pharmacologic suppression of Cx43. Hepatocytes from obese mice were able to transmit ERS to hepatocytes from lean mice, and mice lacking liver Cx43 were protected from diet-induced ERS, insulin resistance, and hepatosteatosis. Taken together, our results indicate that in obesity, the increased Cx43-mediated cell-cell coupling allows intercellular propagation of ERS. This novel maladaptive response to over-nutrition exacerbates the tissue ERS burden, promoting hepatosteatosis and impairing whole-body glucose metabolism.
Collapse
Affiliation(s)
- Amir Tirosh
- Sabri Ülker Center for Metabolic Research, Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, 52621 Tel-HaShomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Harvard Medical School, Boston, MA 02115, USA.
| | - Gurol Tuncman
- Sabri Ülker Center for Metabolic Research, Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ediz S Calay
- Sabri Ülker Center for Metabolic Research, Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Moran Rathaus
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, 52621 Tel-HaShomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Idit Ron
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, 52621 Tel-HaShomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amit Tirosh
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, 52621 Tel-HaShomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Abdullah Yalcin
- Sabri Ülker Center for Metabolic Research, Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Adnan Menderes Üniversitesi Medical School, Department of Medical Biology, 09100 Aydin, Turkey
| | - Yankun G Lee
- Sabri Ülker Center for Metabolic Research, Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Rinat Livne
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, 52621 Tel-HaShomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sophie Ron
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, 52621 Tel-HaShomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Neri Minsky
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, 52621 Tel-HaShomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ana Paula Arruda
- Sabri Ülker Center for Metabolic Research, Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
38
|
Hwang SM, Lee JY, Park CK, Kim YH. The Role of TRP Channels and PMCA in Brain Disorders: Intracellular Calcium and pH Homeostasis. Front Cell Dev Biol 2021; 9:584388. [PMID: 33585474 PMCID: PMC7876282 DOI: 10.3389/fcell.2021.584388] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Brain disorders include neurodegenerative diseases (NDs) with different conditions that primarily affect the neurons and glia in the brain. However, the risk factors and pathophysiological mechanisms of NDs have not been fully elucidated. Homeostasis of intracellular Ca2+ concentration and intracellular pH (pHi) is crucial for cell function. The regulatory processes of these ionic mechanisms may be absent or excessive in pathological conditions, leading to a loss of cell death in distinct regions of ND patients. Herein, we review the potential involvement of transient receptor potential (TRP) channels in NDs, where disrupted Ca2+ homeostasis leads to cell death. The capability of TRP channels to restore or excite the cell through Ca2+ regulation depending on the level of plasma membrane Ca2+ ATPase (PMCA) activity is discussed in detail. As PMCA simultaneously affects intracellular Ca2+ regulation as well as pHi, TRP channels and PMCA thus play vital roles in modulating ionic homeostasis in various cell types or specific regions of the brain where the TRP channels and PMCA are expressed. For this reason, the dysfunction of TRP channels and/or PMCA under pathological conditions disrupts neuronal homeostasis due to abnormal Ca2+ and pH levels in the brain, resulting in various NDs. This review addresses the function of TRP channels and PMCA in controlling intracellular Ca2+ and pH, which may provide novel targets for treating NDs.
Collapse
Affiliation(s)
- Sung-Min Hwang
- Gachon Pain Center, Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| | - Ji Yeon Lee
- Gil Medical Center, Department of Anesthesiology and Pain Medicine, Gachon University, Incheon, South Korea
| | - Chul-Kyu Park
- Gachon Pain Center, Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| | - Yong Ho Kim
- Gachon Pain Center, Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| |
Collapse
|
39
|
Liu Y, Fan H, Li X, Liu J, Qu X, Wu X, Liu M, Liu Z, Yao R. Trpv4 regulates Nlrp3 inflammasome via SIRT1/PGC-1α pathway in a cuprizone-induced mouse model of demyelination. Exp Neurol 2020; 337:113593. [PMID: 33387462 DOI: 10.1016/j.expneurol.2020.113593] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 11/28/2020] [Accepted: 12/27/2020] [Indexed: 12/29/2022]
Abstract
Increasing evidence has demonstrated that the Nod-like receptor pyrin domain containing 3 (Nlrp3) inflammasome overactivated during demyelinating disorders. It has been implicated that transient receptor potential type 4 (Trpv4) is regarded as a polymodal ionotropic receptor that plays an important role in a multitude of pathological conditions, including inflammation. The aim of this study was to investigate whether the Trpv4 channel regulates Nlrp3 inflammasome in the corpus callosum of mice with demyelination. Our results showed that CPZ treatment significantly increased the expression of Trpv4, activated Nlrp3 inflammasome, reduced peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) and decreased mitochondrial function. siRNA-mediated Nlrp3 knockdown inhibited glial activation and alleviated demyelination. Whereas knockdown of Trpv4 by siRNA markedly ameliorated Nlrp3 inflammasome activation and restored mitochondrial function as well as reducing the level of reactive oxygen species (ROS). Meanwhile, glial activation, demyelination and behavioral impairment induced by CPZ were also alleviated by siRNA-mediated Trpv4 knockdown. Furthermore, immunoprecipitation and use of a lysine acetylation assay showed that Sirtuin1 (SIRT1) mediated the PGC-1α deacetylation, which is involved in Nlrp3 inflammasome activation. These findings suggest that Trpv4 regulates mitochondrial function through the SIRT1/PGC-1α pathway, which further trigger Nlrp3 inflammasome activation in the CPZ-induced demyelination in mice.
Collapse
Affiliation(s)
- Yanan Liu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China; Department of Human Anatomy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Hongbin Fan
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China; Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, PR China
| | - Xinyu Li
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China; Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, PR China
| | - Jing Liu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China; Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, PR China
| | - Xuebin Qu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Xiuxiang Wu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Meiying Liu
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Zhian Liu
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China.
| |
Collapse
|
40
|
Cho J, Huh Y. Astrocytic Calcium Dynamics Along the Pain Pathway. Front Cell Neurosci 2020; 14:594216. [PMID: 33192331 PMCID: PMC7596274 DOI: 10.3389/fncel.2020.594216] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/14/2020] [Indexed: 12/30/2022] Open
Abstract
Astrocytes, once thought to be passive cells merely filling the space between neurons in the nervous system, are receiving attention as active modulators of the brain and spinal cord physiology by providing nutrients, maintaining homeostasis, and modulating synaptic transmission. Accumulating evidence indicates that astrocytes are critically involved in chronic pain regulation. Injury induces astrocytes to become reactive, and recent studies suggest that reactive astrocytes can have either neuroprotective or neurodegenerative effects. While the exact mechanisms underlying the transition from resting astrocytes to reactive astrocytes remain unknown, astrocytic calcium increase, coordinated by inflammatory molecules, has been suggested to trigger this transition. In this mini review article, we will discuss the roles of astrocytic calcium, channels contributing to calcium dynamics in astrocytes, astrocyte activations along the pain pathway, and possible relationships between astrocytic calcium dynamics and chronic pain.
Collapse
Affiliation(s)
- Jeiwon Cho
- Brain and Cognitive Science, Scranton College, Ewha Womans University, Seoul, South Korea
| | - Yeowool Huh
- Department of Medical Science, College of Medicine, Catholic Kwandong University, Incheon, South Korea.,Translational Brain Research Center, Catholic Kwandong University, International St. Mary's Hospital, Incheon, South Korea
| |
Collapse
|
41
|
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: 4.4] [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
|
42
|
Liu N, Yan F, Ma Q, Zhao J. Modulation of TRPV4 and BKCa for treatment of brain diseases. Bioorg Med Chem 2020; 28:115609. [PMID: 32690264 DOI: 10.1016/j.bmc.2020.115609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/22/2022]
Abstract
As a member of transient receptor potential family, the transient receptor potential vanilloid 4 (TRPV4) is a kind of nonselective calcium-permeable cation channel, which belongs to non-voltage gated Ca2+ channel. Large-conductance Ca2+-activated K+ channel (BKCa) represents a unique superfamily of Ca2+-activated K+ channel (KCa) that is both voltage and intracellular Ca2+ dependent. Not surprisingly, aberrant function of either TRPV4 or BKCa in neurons has been associated with brain disorders, such as Alzheimer's disease, cerebral ischemia, brain tumor, epilepsy, as well as headache. In these diseases, vascular dysfunction is a common characteristic. Notably, endothelial and smooth muscle TRPV4 can mediate BKCa to regulate cerebral blood flow and pressure. Therefore, in this review, we not only discuss the diverse functions of TRPV4 and BKCa in neurons to integrate relative signaling pathways in the context of cerebral physiological and pathological situations respectively, but also reveal the relationship between TRPV4 and BKCa in regulation of cerebral vascular tone as an etiologic factor. Based on these analyses, this review demonstrates the effective mechanisms of compounds targeting these two channels, which may be potential therapeutic strategies for diseases in the brain.
Collapse
Affiliation(s)
- Na Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China; Department of Anesthesiology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, PR China
| | - Fang Yan
- Medical School, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Qingjie Ma
- Department of Anesthesiology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, PR China
| | - Jianhua Zhao
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, PR China.
| |
Collapse
|
43
|
Zhao Y, Yang J, Li C, Zhou G, Wan H, Ding Z, Wan H, Zhou H. Role of the neurovascular unit in the process of cerebral ischemic injury. Pharmacol Res 2020; 160:105103. [PMID: 32739425 DOI: 10.1016/j.phrs.2020.105103] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022]
Abstract
Cerebral ischemic injury exhibits both high morbidity and mortality worldwide. Traditional research of the pathogenesis of cerebral ischemic injury has focused on separate analyses of the involved cell types. In recent years, the neurovascular unit (NVU) mechanism of cerebral ischemic injury has been proposed in modern medicine. Hence, more effective strategies for the treatment of cerebral ischemic injury may be provided through comprehensive analysis of brain cells and the extracellular matrix. However, recent studies that have investigated the function of the NVU in cerebral ischemic injury have been insufficient. In addition, the metabolism and energy conversion of the NVU depend on interactions among multiple cell types, which make it difficult to identify the unique contribution of each cell type. Therefore, in the present review, we comprehensively summarize the regulatory effects and recovery mechanisms of four major cell types (i.e., astrocytes, microglia, brain-microvascular endothelial cells, and neurons) in the NVU under cerebral ischemic injury, as well as discuss the interactions among these cell types in the NVU. Furthermore, we discuss the common signaling pathways and signaling factors that mediate cerebral ischemic injury in the NVU, which may help to provide a theoretical basis for the comprehensive elucidation of cerebral ischemic injury.
Collapse
Affiliation(s)
- Yu Zhao
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Chang Li
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Guoying Zhou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Haofang Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Zhishan Ding
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| |
Collapse
|
44
|
Vagus nerve stimulation reduces spreading depolarization burden and cortical infarct volume in a rat model of stroke. PLoS One 2020; 15:e0236444. [PMID: 32702055 PMCID: PMC7377493 DOI: 10.1371/journal.pone.0236444] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/06/2020] [Indexed: 01/30/2023] Open
Abstract
Cortical spreading depolarization (SD) waves negatively affect neuronal survival and outcome after ischemic stroke. We here aimed to investigate the effects of vagus nerve stimulation (VNS) on SDs in a rat model of focal ischemia. To this end, we delivered non-invasive VNS (nVNS) or invasive VNS (iVNS) during permanent middle cerebral artery occlusion (MCAO), and found that both interventions significantly reduced the frequency of SDs in the cortical peri-infarct area compared to sham VNS, without affecting relative blood flow changes, blood pressure, heart rate or breathing rate. In separate groups of rats subjected to transient MCAO, we found that cortical stroke volume was reduced 72 h after transient MCAO, whereas stroke volume in the basal ganglia remained unchanged. In rats treated with nVNS, motor outcome was improved 2 days after transient MCAO, but was similar to sham VNS animals 3 days after ischemia. We postulate that VNS may be a safe and efficient intervention to reduce the clinical burden of SD waves in stroke and other conditions.
Collapse
|
45
|
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: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 05/14/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction Numerous studies have shown the ability of low-energy acoustic waves such as focused ultrasound or shockwave to transiently open blood-brain barrier (BBB) and facilitate drug delivery to the brain. Preclinical and clinical evidences have well demonstrated the efficacy and safety in treating various brain disorders. However, the molecular mechanisms of acoustic waves on the BBB are still not fully understood. Objectives The present study aimed at exploring the possible molecular mechanisms of acoustic wave stimulation on brains. Methods: Briefly describe the experimental design The left hemisphere of the rat‘s brain was treated with pulsed ultrasound from a commercial focused shockwave or a planar ultrasound device, and the right hemisphere served as a control. One hour after the mechanical wave stimulation or overnight, the rats were sacrificed and the brains were harvested for protein or histological analysis. Agonists and antagonists related to the signal transduction pathways of tight junction proteins were used to investigate the possible intracellular mechanisms. Results Intracellular signal transduction analysis shows calcium influx through transient receptor potential vanilloid 4 (TRPV4) channels, and the activation of PKC-δ pathway to mediate dissociation of ZO-1 and occludin after acoustic wave stimulation. The activation of TRPV4 or PKC-δ signaling further increased the expression level of TRPV4, suggesting a feedback loop to regulate BBB permeability. Moreover, the tight junction proteins dissociation can be reversed by administration of PKC-δ inhibitor and TRPV4 antagonist. Conclusion The present study shows the crucial role of TRPV4 in acoustic wave-mediated BBB permeability, specifically its effect on compromising tight junction proteins, ZO-1 and occludin. Our findings provide a new molecular perspective to explain acoustic wave-mediated BBB opening. Moreover, activation of TRPV4 by agonists may reduce the threshold intensity level of acoustic waves for BBB opening, which may prevent undesirable mechanical damages while maintaining efficient BBB opening.
Collapse
|
46
|
Toft-Bertelsen TL, Larsen BR, Christensen SK, Khandelia H, Waagepetersen HS, MacAulay N. Clearance of activity-evoked K + transients and associated glia cell swelling occur independently of AQP4: A study with an isoform-selective AQP4 inhibitor. Glia 2020; 69:28-41. [PMID: 32506554 DOI: 10.1002/glia.23851] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 12/29/2022]
Abstract
The mammalian brain consists of 80% water, which is continuously shifted between different compartments and cellular structures by mechanisms that are, to a large extent, unresolved. Aquaporin 4 (AQP4) is abundantly expressed in glia and ependymal cells of the mammalian brain and has been proposed to act as a gatekeeper for brain water dynamics, predominantly based on studies utilizing AQP4-deficient mice. However, these mice have a range of secondary effects due to the gene deletion. An efficient and selective AQP4 inhibitor has thus been sorely needed to validate the results obtained in the AQP4-/- mice to quantify the contribution of AQP4 to brain fluid dynamics. In AQP4-expressing Xenopus laevis oocytes monitored by a high-resolution volume recording system, we here demonstrate that the compound TGN-020 is such a selective AQP4 inhibitor. TGN-020 targets the tested species of AQP4 with an IC50 of ~3.5 μM, but displays no inhibitory effect on the other AQPs (AQP1-AQP9). With this tool, we employed rat hippocampal slices and ion-sensitive microelectrodes to determine the role of AQP4 in glia cell swelling following neuronal activity. TGN-020-mediated inhibition of AQP4 did not prevent stimulus-induced extracellular space shrinkage, nor did it slow clearance of the activity-evoked K+ transient. These data, obtained with a verified isoform-selective AQP4 inhibitor, indicate that AQP4 is not required for the astrocytic contribution to the K+ clearance or the associated extracellular space shrinkage.
Collapse
Affiliation(s)
- Trine Lisberg Toft-Bertelsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Brian Roland Larsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sofie Kjellerup Christensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Himanshu Khandelia
- Department of Physics, Chemistry and Pharmacy, Faculty of Science, University of Southern Denmark, Odense, Denmark
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Tanaka K, Matsumoto S, Yamada T, Yamasaki R, Suzuki M, Kido MA, Kira JI. Reduced Post-ischemic Brain Injury in Transient Receptor Potential Vanilloid 4 Knockout Mice. Front Neurosci 2020; 14:453. [PMID: 32477057 PMCID: PMC7235376 DOI: 10.3389/fnins.2020.00453] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 04/14/2020] [Indexed: 12/25/2022] Open
Abstract
Background and Purpose In the acute phase of ischemia-reperfusion, hypoperfusion associated with ischemia and reperfusion in microvascular regions and disruption of the blood-brain barrier (BBB) contribute to post-ischemic brain injury. We aimed to clarify whether brain injury following transient middle cerebral artery occlusion (tMCAO) is ameliorated in Transient receptor potential vanilloid 4 knockout (Trpv4-/- ) mice. Methods tMCAO was induced in wild-type (WT) and Trpv4-/- mice aged 8-10 weeks. Ischemia-induced lesion volume was evaluated by 2,3,5-triphenyltetrazolium chloride staining at 24 h post-tMCAO. Tissue water content and Evans blue leakage in the ipsilateral hemisphere and a neurological score were evaluated at 48 h post-tMCAO. Transmission electron microscopy (TEM) was performed to assess the morphological changes in microvasculature in the ischemic lesions at 6 h post-tMCAO. Results Compared with WT mice, Trpv4-/- mice showed reduced ischemia-induced lesion volume and reduced water content and Evans blue leakage in the ipsilateral hemisphere alongside milder neurological symptoms. The loss of zonula occludens-1 and occludin proteins in the ipsilateral hemisphere was attenuated in Trpv4-/- mice. TEM revealed that parenchymal microvessels in the ischemic lesion were compressed and narrowed by the swollen endfeet of astrocytes in WT mice, but these effects were markedly ameliorated in Trpv4-/- mice. Conclusion The present results demonstrate that TRPV4 contributes to post-ischemic brain injury. The preserved microcirculation and BBB function shortly after reperfusion are the key neuroprotective roles of TRPV4 inhibition, which represents a promising target for the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Koji Tanaka
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shoji Matsumoto
- Department of Comprehensive Strokology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Takeshi Yamada
- Department of Neurology, Saiseikai Fukuoka General Hospital, Fukuoka, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Makoto Suzuki
- Department of Pharmacology, Division of Molecular Pharmacology, Jichi Medical University, Shimotsuke, Japan
| | - Mizuho A Kido
- Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
48
|
MacAulay N. Molecular mechanisms of K + clearance and extracellular space shrinkage-Glia cells as the stars. Glia 2020; 68:2192-2211. [PMID: 32181522 DOI: 10.1002/glia.23824] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 12/17/2022]
Abstract
Neuronal signaling in the central nervous system (CNS) associates with release of K+ into the extracellular space resulting in transient increases in [K+ ]o . This elevated K+ is swiftly removed, in part, via uptake by neighboring glia cells. This process occurs in parallel to the [K+ ]o elevation and glia cells thus act as K+ sinks during the neuronal activity, while releasing it at the termination of the pulse. The molecular transport mechanisms governing this glial K+ absorption remain a point of debate. Passive distribution of K+ via Kir4.1-mediated spatial buffering of K+ has become a favorite within the glial field, although evidence for a quantitatively significant contribution from this ion channel to K+ clearance from the extracellular space is sparse. The Na+ /K+ -ATPase, but not the Na+ /K+ /Cl- cotransporter, NKCC1, shapes the activity-evoked K+ transient. The different isoform combinations of the Na+ /K+ -ATPase expressed in glia cells and neurons display different kinetic characteristics and are thereby distinctly geared toward their temporal and quantitative contribution to K+ clearance. The glia cell swelling occurring with the K+ transient was long assumed to be directly associated with K+ uptake and/or AQP4, although accumulating evidence suggests that they are not. Rather, activation of bicarbonate- and lactate transporters appear to lead to glial cell swelling via the activity-evoked alkaline transient, K+ -mediated glial depolarization, and metabolic demand. This review covers evidence, or lack thereof, accumulated over the last half century on the molecular mechanisms supporting activity-evoked K+ and extracellular space dynamics.
Collapse
Affiliation(s)
- Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
49
|
Gerkau NJ, Rakers C, Durry S, Petzold GC, Rose CR. Reverse NCX Attenuates Cellular Sodium Loading in Metabolically Compromised Cortex. Cereb Cortex 2019; 28:4264-4280. [PMID: 29136153 DOI: 10.1093/cercor/bhx280] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/04/2017] [Indexed: 01/05/2023] Open
Abstract
In core regions of ischemic stroke, disruption of blood flow causes breakdown of ionic gradients and, ultimately, calcium overload and cell death. In the surrounding penumbra, cells may recover upon reperfusion, but recovery is hampered by additional metabolic demands imposed by peri-infarct depolarizations (PIDs). There is evidence that sodium influx drives PIDs, but no data exist on PID-related sodium accumulations in vivo. Here, we found that PIDs in mouse neocortex are associated with propagating sodium elevations in neurons and astrocytes. Similar transient sodium elevations were induced in acute tissue slices by brief chemical ischemia. Blocking NMDA-receptors dampened sodium and accompanying calcium loads of neurons in tissue slices, while inhibiting glutamate transport diminished sodium influx into astrocytes, but amplified neuronal sodium loads. In both cell types, inhibition of sodium/calcium exchange (NCX) increased sodium transients. Blocking NCX also significantly reduced calcium transients, a result confirmed in vivo. Our study provides the first quantitative data on sodium elevations in peri-infarct regions in vivo. They suggest that sodium influx drives reversal of NCX, triggering a massive secondary calcium elevation while promoting export of sodium. Reported neuroprotective effects of NCX activity in stroke models might thus be related to its dampening of ischemia-induced sodium loading.
Collapse
Affiliation(s)
- Niklas J Gerkau
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Universitaetsstrasse 1, Duesseldorf, Germany
| | - Cordula Rakers
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, Bonn, Germany
| | - Simone Durry
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Universitaetsstrasse 1, Duesseldorf, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, Bonn, Germany.,Department of Neurology, University Hospital Bonn, Sigmund-Freud-Str. 25, Bonn, Germany
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Universitaetsstrasse 1, Duesseldorf, Germany
| |
Collapse
|
50
|
Beiersdorfer A, Lohr C. AMPA Receptor-Mediated Ca 2+ Transients in Mouse Olfactory Ensheathing Cells. Front Cell Neurosci 2019; 13:451. [PMID: 31636544 PMCID: PMC6788192 DOI: 10.3389/fncel.2019.00451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/20/2019] [Indexed: 11/13/2022] Open
Abstract
Ca2+ signaling in glial cells is primarily triggered by metabotropic pathways and the subsequent Ca2+ release from internal Ca2+ stores. However, there is upcoming evidence that various ion channels might also initiate Ca2+ rises in glial cells by Ca2+ influx. We investigated AMPA receptor-mediated inward currents and Ca2+ transients in olfactory ensheathing cells (OECs), a specialized glial cell population in the olfactory bulb (OB), using whole-cell voltage-clamp recordings and confocal Ca2+ imaging. By immunohistochemistry we showed immunoreactivity to the AMPA receptor subunits GluA1, GluA2 and GluA4 in OECs, suggesting the presence of AMPA receptors in OECs. Kainate-induced inward currents were mediated exclusively by AMPA receptors, as they were sensitive to the specific AMPA receptor antagonist, GYKI53655. Moreover, kainate-induced inward currents were reduced by the selective Ca2+-permeable AMPA receptor inhibitor, NASPM, suggesting the presence of functional Ca2+-permeable AMPA receptors in OECs. Additionally, kainate application evoked Ca2+ transients in OECs which were abolished in the absence of extracellular Ca2+, indicating that Ca2+ influx via Ca2+-permeable AMPA receptors contribute to kainate-induced Ca2+ transients. However, kainate-induced Ca2+ transients were partly reduced upon Ca2+ store depletion, leading to the conclusion that Ca2+ influx via AMPA receptor channels is essential to trigger Ca2+ transients in OECs, whereas Ca2+ release from internal stores contributes in part to the kainate-evoked Ca2+ response. Endogenous glutamate release by OSN axons initiated Ca2+ transients in OECs, equally mediated by metabotropic receptors (glutamatergic and purinergic) and AMPA receptors, suggesting a prominent role for AMPA receptor mediated Ca2+ signaling in axon-OEC communication.
Collapse
Affiliation(s)
| | - Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| |
Collapse
|