1
|
Zhuang W, Park M, Jeong J, Kim HR, Jang Y, Seo MS, An JR, Park H, Jung WK, Choi IW, Park WS. The sodium-glucose cotransporter 2 inhibitor tofogliflozin induces vasodilation of rabbit femoral artery by activating Kv channels, the SERCA pump, and the sGC/cGMP pathway. Eur J Pharmacol 2025; 996:177595. [PMID: 40189081 DOI: 10.1016/j.ejphar.2025.177595] [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: 12/17/2024] [Revised: 02/21/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025]
Abstract
Tofogliflozin is a sodium-glucose cotransporter 2 inhibitor widely used to treat type 2 diabetes mellitus, but it also exhibits cardio-protective effects. This study investigated the vasodilatory action of tofogliflozin using rabbit femoral artery rings pre-contracted with phenylephrine (1 μM). The results showed the concentration-dependent induction of vasodilation by tofogliflozin, a response that remained unchanged following endothelial removal, pretreatment with the nitric oxide synthase inhibitor L-NAME (100 μM), or the inhibition of low- and intermediate-conductance Ca2+-activated K+ channels using apamin (1 μM) in combination with TRAM-34 (1 μM). Furthermore, pretreatment with the voltage-dependent K+ (Kv) channel inhibitor 4-AP (3 mM) reduced the vasodilatory effects of tofogliflozin whereas pretreatment with the ATP-sensitive K+ channel inhibitor glibenclamide (10 μM) or the large-conductance Ca2+-activated K+ channel inhibitor paxilline (1 μM) did not. Notably, our findings indicated that Kv7.X, rather than Kv1.5 or Kv2.1, is the primary Kv subtype involved in tofogliflozin-induced vasodilation. The vasodilatory effects of tofogliflozin were also significantly inhibited in femoral arterial rings pretreated with the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump inhibitors thapsigargin (1 μM) and cyclopiazonic acid (10 μM). Tofogliflozin-induced vasodilation was unaltered in arterial rings exposed to the adenylyl cyclase inhibitor SQ 22536 (50 μM), the protein kinase A (PKA) inhibitor KT 5720 (1 μM), and the protein kinase G inhibitor KT 5823 (1 μM) whereas it was effectively reduced by the soluble guanylyl cyclase (sGC) inhibitor ODQ (10 μM). These findings suggest that tofogliflozin-induced vasodilation is mediated by the activation of the SERCA pump, the sGC/cGMP pathway, and Kv channels.
Collapse
Affiliation(s)
- Wenwen Zhuang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Minju Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Junsu Jeong
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Hye Ryung Kim
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - YeEun Jang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Mi Seon Seo
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, 27478, South Korea
| | - Jin Ryeol An
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, 27478, South Korea
| | - Hongzoo Park
- Institute of Medical Sciences, Department of Urology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Won-Kyo Jung
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan, 48513, South Korea
| | - Il-Whan Choi
- Department of Microbiology, College of Medicine, Inje University, Busan, 48516, South Korea
| | - Won Sun Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea.
| |
Collapse
|
2
|
Babur E, Saray H, Süer C, Dursun N. Inhibition of Rho-kinase by fasudil contributes to the modulation of the synaptic plasticity response in the rat hippocampus. Pflugers Arch 2025; 477:787-796. [PMID: 40216618 DOI: 10.1007/s00424-025-03078-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/06/2025] [Accepted: 03/12/2025] [Indexed: 05/21/2025]
Abstract
Metaplasticity refers to an activity-dependent change in the physiological or biochemical state of neurons that changes their ability to generate subsequently induced synaptic plasticity, such as long-term potentiation (LTP) or long-term depression (LTD). Rho-kinases (ROCK) are known to be important for stable changes in synaptic strength, especially LTP. In this study, we investigated whether LTP inhibition in synapses primed with 1-Hz stimulation was affected by ROCK inhibition in young adult male rats. The study also examined the pattern of tau phosphorylation that occurs during metaplastic regulation, applying into perspective the phosphorylation of tau protein by ROCK. Field potentials consisting of an excitatory postsynaptic potential (fEPSP) and population spike (PS) were recorded from the granule cell layer of the hippocampal dentate gyrus (DG). Metaplastic LTP was induced by strong tetanic stimulation (HFS) of the lateral perforant path after a low-frequency stimulation (LFS) protocol. A glass micropipette was inserted into the granule cell layer of the ipsilateral dentate gyrus to record fEPSP and drug infusion. Drug infusion (saline, n = 8; fasudil, n = 8, 10 µM) was started after the 15-min baseline recording and lasted for 60 min. Total and phosphorylated tau levels were measured in the stimulated hippocampus, which was immediately removed after the electrophysiological recording. LFS prevented the induction of LTP in response to HFS and even produced synaptic LTD in the saline-infused group (83.8 ± 2.6% of the baseline), but moderate potentiation of fEPSP (121.1 ± 7.7% of the baseline) occurred at the end of recording in the experiments where fasudil infusion was performed. LFS caused a comparable early depression, and HFS resulted in a comparable potentiation of the PS amplitude in both groups. Granular cells of the DG failed to exhibit synaptic LTP inhibition in the presence of fasudil, and levels of total and phosphorylated GSK-3β and levels of phosphorylated tau (Ser396 and Ser202-Thr205) were found to be lower than those of the control group. Based on these findings, it can be concluded that pharmacological inhibition of ROCK results in impaired ability of dentate gyrus neurons to inhibit synaptic LTP, and this result is accompanied by decreased phosphorylation of GSK-3β and tau proteins. The negative effect of fasudil on neuronal function should not be neglected when evaluating its effects as a therapeutic agent for diseases.
Collapse
Affiliation(s)
- Ercan Babur
- Department of Physiology, Erciyes University Faculty of Medicine, Kayseri, 38000, Turkey
| | - Hatice Saray
- Department of Physiology, Erciyes University Faculty of Medicine, Kayseri, 38000, Turkey
| | - Cem Süer
- Department of Physiology, Erciyes University Faculty of Medicine, Kayseri, 38000, Turkey
| | - Nurcan Dursun
- Department of Physiology, Erciyes University Faculty of Medicine, Kayseri, 38000, Turkey.
| |
Collapse
|
3
|
Lemke J, Gollasch M, Tsvetkov D, Schulig L. Advances in the design and development of chemical modulators of the voltage-gated potassium channels K V7.4 and K V7.5. Expert Opin Drug Discov 2025; 20:47-62. [PMID: 39627683 DOI: 10.1080/17460441.2024.2438226] [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: 06/14/2024] [Revised: 10/20/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
INTRODUCTION Hypertension remains a major public health concern, with significant morbidity and mortality worldwide. Despite the availability of various antihypertensive medications, blood pressure control remains suboptimal in many individuals. During the last decades, KV7.4 and KV7.5, which were already known from the view of neuronal regulation, emerged as possible important players in the regulation of vascular tone and blood pressure. AREAS COVERED This review covers physiological functions and current advancements in the development of KV7.4 and KV7.5 channel modulators. The authors highlight the structural elements likely to be important for the future design of KV7 subtype-selective modulators, underscoring their potential as an innovative hypertension treatment. EXPERT OPINION Extensive research has been focused on targeting neuronal KV7.2 and KV7.3 channels, while KV7.4 and KV7.5 attracted less attention. Many of the developed compounds represent derivatives of flupirtine or retigabine, whereby subtype channel selectivity has only been demonstrated for a handful of individual compounds. Novel substances address additional sites within the binding pocket by incorporating new functional groups. A comprehensive and systematic evaluation of a compound set with significant subtype selectivity should be performed. The discovery of new highly active, less toxic, and selective compounds, therefore, remains the goal of further research in the coming years.
Collapse
Affiliation(s)
- Jana Lemke
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | - Maik Gollasch
- Department of Internal Medicine and Geriatrics, University Medicine, Greifswald, Germany
| | - Dmitry Tsvetkov
- Department of Internal Medicine and Geriatrics, University Medicine, Greifswald, Germany
| | - Lukas Schulig
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| |
Collapse
|
4
|
Liu E, Pang K, Liu M, Tan X, Hang Z, Mu S, Han W, Yue Q, Comai S, Sun J. Activation of Kv7 channels normalizes hyperactivity of the VTA-NAcLat circuit and attenuates methamphetamine-induced conditioned place preference and sensitization in mice. Mol Psychiatry 2023; 28:5183-5194. [PMID: 37604975 DOI: 10.1038/s41380-023-02218-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/23/2023]
Abstract
The brain circuit projecting from the ventral tegmental area (VTA) to the nucleus accumbens lateral shell (NAcLat) has a key role in methamphetamine (MA) addiction. As different dopamine (DA) neuron subpopulations in the VTA participate in different neuronal circuits, it is a challenge to isolate these DA neuron subtypes. Using retrograde tracing and Patch-seq, we isolated DA neurons in the VTA-NAcLat circuit in MA-treated mice and performed gene expression profiling. Among the differentially expressed genes, KCNQ genes were dramatically downregulated. KCNQ genes encode Kv7 channel proteins, which modulate neuronal excitability. Injection of both the Kv7.2/3 agonist ICA069673 and the Kv7.4 agonist fasudil into the VTA attenuated MA-induced conditioned place preference and locomotor sensitization and decreased neuronal excitability. Increasing Kv7.2/3 activity decreased neural oscillations, synaptic plasticity and DA release in the VTA-NacLat circuit in MA-treated mice. Furthermore, overexpression of only Kv7.3 channels in the VTA-NacLat circuit was sufficient to attenuate MA-induced reward behavior and decrease VTA neuron excitability. Activation of Kv7 channels in the VTA may become a novel treatment strategy for MA abuse.
Collapse
Affiliation(s)
- E Liu
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Jinan, Shandong, China
| | - Kunkun Pang
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Jinan, Shandong, China
- Department of Ultrasound, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Min Liu
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Jinan, Shandong, China
| | - Xu Tan
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Jinan, Shandong, China
| | - Zhaofang Hang
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Jinan, Shandong, China
| | - Shouhong Mu
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Jinan, Shandong, China
| | - Weikai Han
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Jinan, Shandong, China
| | - Qingwei Yue
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Jinan, Shandong, China
| | - Stefano Comai
- Department of Psychiatry, McGill University, Montréal, QC, Canada
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Jinhao Sun
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Jinan, Shandong, China.
| |
Collapse
|
5
|
Sarkar T, Moinuddin SM, Isbatan A, Chen J, Mann D, Ahsan F. Intratracheally Administered Peptide-Modified Lipid Admixture Containing Fasudil and/or DETA NONOate Ameliorates Various Pathologies of Pulmonary Arterial Hypertension. Pharmaceuticals (Basel) 2023; 16:1656. [PMID: 38139783 PMCID: PMC10747237 DOI: 10.3390/ph16121656] [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] [Received: 11/03/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
This study examined the therapeutic potential of a combination therapy using fasudil, a Rho-kinase inhibitor, and DETA NONOate (DN), a nitric oxide donor, delivered as a lipid admixture modified with a cyclic homing peptide known as CAR (CAR-lipid mixture) for the treatment of pulmonary arterial hypertension (PAH). CAR-lipid mixtures were initially prepared via a thin-film hydration method and then combined with fasudil, DN, or a mixture of both. The therapeutic efficacy of this drug-laden lipid mixture was evaluated in a Sugen/Hypoxia (Su/Hx) rat model of PAH by measuring RV systolic pressure (RVSP), mean pulmonary arterial pressure (mPAP), Fulton indices, and assessing right ventricular (RV) functions, as well as evaluating pulmonary vascular morphology. Rats that received no treatment exhibited increases in RVSP, mPAP, Fulton indices, and changes in RV functional parameters. However, the treatment with the CAR-lipid mixture containing either fasudil or DN or a combination of both led to a decline in mPAP, RVSP, and Fulton indices compared to saline-treated rats. Similarly, rats that received these treatments showed concurrent improvement in various echocardiographic parameters such as pulmonary acceleration time (PAT), tricuspid annular plane systolic excursion (TAPSE), and ventricular free wall thickness (RVFWT). A significant decrease in the wall thickness of pulmonary arteries larger than 100 µm was observed with the combination therapy. The findings reveal that fasudil, DN, and their combination in a CAR-modified lipid mixture improved pulmonary hemodynamics, RV functions, and pathological alterations in the pulmonary vasculature. This study underscores the potential of combination therapy and targeted drug delivery in PAH treatment, laying the groundwork for future investigations into the optimization of these treatments, their long-term safety and efficacy, and the underlying mechanism of action of the proposed therapy.
Collapse
Affiliation(s)
- Tanoy Sarkar
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| | - Sakib M. Moinuddin
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| | - Ayman Isbatan
- Cardiovascular Research Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jiwang Chen
- Cardiovascular Research Center, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Medicine, Section of Pulmonary, Critical Care Medicine, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - David Mann
- Vascular BioSciences, Goleta, CA 93117, USA
| | - Fakhrul Ahsan
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| |
Collapse
|
6
|
Guan X, Wei D, Liang Z, Xie L, Wang Y, Huang Z, Wu J, Pang T. FDCA Attenuates Neuroinflammation and Brain Injury after Cerebral Ischemic Stroke. ACS Chem Neurosci 2023; 14:3839-3854. [PMID: 37768739 DOI: 10.1021/acschemneuro.3c00456] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023] Open
Abstract
Ischemic stroke is a deleterious cerebrovascular disease with few therapeutic options, and its functional recovery is highly associated with the integrity of the blood-brain barrier and neuroinflammation. The Rho-associated coiled-coil containing protein kinase (ROCK) inhibitor fasudil (F) and the pyruvate dehydrogenase kinase (PDK) inhibitor dichloroacetate (DCA) have been demonstrated to exhibit neuroprotection in a series of neurological disorders. Hence, we synthesized and biologically examined the new salt fasudil dichloroacetate (FDCA) and validated that FDCA was eligible for attenuating ischemic volume and neurological deficits in the rat transient middle cerebral artery occlusion (tMCAO) model. Additionally, FDCA exerted superior effects than fasudil and dichloroacetate alone or in combination in reducing cerebral ischemic injury. Particularly, FDCA could maintain the blood-brain barrier (BBB) integrity by inhibiting matrix metalloproteinase 9 (MMP-9) protein expression and the degradation of zonula occludens (ZO-1) and Occludin protein. Meanwhile, FDCA could mitigate the neuroinflammation induced by microglia. The in vivo and in vitro experiments further demonstrated that FDCA disrupted the phosphorylations of myosin phosphatase target subunit 1 (MYPT1), mitogen-activated protein kinase (MAPK) cascade, including p38 and c-Jun N-terminal kinase (JNK), and pyruvate dehydrogenase (PDH) and limited excessive lactic acid metabolites, resulting in inhibition of BBB disruption and neuroinflammation. In addition, FDCA potently mitigated inflammatory response in human monocytes isolated from ischemic stroke patients, which provides the possibilities of a clinical translation perspective. Overall, these findings provided a therapeutic potential for FDCA as a candidate agent for ischemic stroke and other neurological diseases associated with BBB disruption and neuroinflammation.
Collapse
Affiliation(s)
- Xin Guan
- State Key Laboratory of Natural Medicines, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Dasha Wei
- State Key Laboratory of Natural Medicines, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Zhuangzhuang Liang
- State Key Laboratory of Natural Medicines, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Luyang Xie
- State Key Laboratory of Natural Medicines, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yifang Wang
- Department of Neurology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, P. R. China
| | - Zhangjian Huang
- State Key Laboratory of Natural Medicines, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Jin Wu
- Department of Neurology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, P. R. China
| | - Tao Pang
- State Key Laboratory of Natural Medicines, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, P. R. China
| |
Collapse
|
7
|
Zhang H, Li H, Lu M, Wang S, Ma X, Wang F, Liu J, Li X, Yang H, Zhang F, Shen H, Buckley NJ, Gamper N, Yamoah EN, Lv P. Repressor element 1-silencing transcription factor deficiency yields profound hearing loss through K v7.4 channel upsurge in auditory neurons and hair cells. eLife 2022; 11:76754. [PMID: 36125121 PMCID: PMC9525063 DOI: 10.7554/elife.76754] [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: 01/04/2022] [Accepted: 09/16/2022] [Indexed: 11/23/2022] Open
Abstract
Repressor element 1-silencing transcription factor (REST) is a transcriptional repressor that recognizes neuron-restrictive silencer elements in the mammalian genomes in a tissue- and cell-specific manner. The identity of REST target genes and molecular details of how REST regulates them are emerging. We performed conditional null deletion of Rest (cKO), mainly restricted to murine hair cells (HCs) and auditory neurons (aka spiral ganglion neurons [SGNs]). Null inactivation of full-length REST did not affect the development of normal HCs and SGNs but manifested as progressive hearing loss in adult mice. We found that the inactivation of REST resulted in an increased abundance of Kv7.4 channels at the transcript, protein, and functional levels. Specifically, we found that SGNs and HCs from Rest cKO mice displayed increased Kv7.4 expression and augmented Kv7 currents; SGN’s excitability was also significantly reduced. Administration of a compound with Kv7.4 channel activator activity, fasudil, recapitulated progressive hearing loss in mice. In contrast, inhibition of the Kv7 channels by XE991 rescued the auditory phenotype of Rest cKO mice. Previous studies identified some loss-of-function mutations within the Kv7.4-coding gene, Kcnq4, as a causative factor for progressive hearing loss in mice and humans. Thus, the findings reveal that a critical homeostatic Kv7.4 channel level is required for proper auditory functions.
Collapse
Affiliation(s)
- Haiwei Zhang
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Hongchen Li
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Mingshun Lu
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Shengnan Wang
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Xueya Ma
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Fei Wang
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Jiaxi Liu
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Xinyu Li
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Haichao Yang
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Fan Zhang
- Department of Pharmacology, Hebei Medical University, Hebei, China
| | - Haitao Shen
- Laboratory of Pathology, Hebei Medical University, Hebei, China
| | - Noel J Buckley
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Nikita Gamper
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, University of Nevada Reno, Reno, United States
| | - Ping Lv
- Department of Pharmacology, Hebei Medical University, Hebei, China
| |
Collapse
|
8
|
Singh SP, William M, Malavia M, Chu XP. Behavior of KCNQ Channels in Neural Plasticity and Motor Disorders. MEMBRANES 2022; 12:membranes12050499. [PMID: 35629827 PMCID: PMC9143857 DOI: 10.3390/membranes12050499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 02/01/2023]
Abstract
The broad distribution of voltage-gated potassium channels (VGKCs) in the human body makes them a critical component for the study of physiological and pathological function. Within the KCNQ family of VGKCs, these aqueous conduits serve an array of critical roles in homeostasis, especially in neural tissue. Moreover, the greater emphasis on genomic identification in the past century has led to a growth in literature on the role of the ion channels in pathological disease as well. Despite this, there is a need to consolidate the updated findings regarding both the pharmacotherapeutic and pathological roles of KCNQ channels, especially regarding neural plasticity and motor disorders which have the largest body of literature on this channel. Specifically, KCNQ channels serve a remarkable role in modulating the synaptic efficiency required to create appropriate plasticity in the brain. This role can serve as a foundation for clinical approaches to chronic pain. Additionally, KCNQ channels in motor disorders have been utilized as a direction for contemporary pharmacotherapeutic developments due to the muscarinic properties of this channel. The aim of this study is to provide a contemporary review of the behavior of these channels in neural plasticity and motor disorders. Upon review, the behavior of these channels is largely dependent on the physiological role that KCNQ modulatory factors (i.e., pharmacotherapeutic options) serve in pathological diseases.
Collapse
|
9
|
Lin H, Bai H, Yang Z, Shen Q, Li M, Huang Y, Lv F, Wang S. Conjugated Polymers for Biomedical Applications. Chem Commun (Camb) 2022; 58:7232-7244. [DOI: 10.1039/d2cc02177c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Conjugated polymers (CPs) are a series of organic semiconductor materials with large π-conjugated backbones and delocalized electronic structures. Due to their specific photophysical properties and photoelectric effects, plenty of CPs...
Collapse
|
10
|
Progression of KCNQ4 related genetic hearing loss: a narrative review. JOURNAL OF BIO-X RESEARCH 2021. [DOI: 10.1097/jbr.0000000000000112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
11
|
Zheng CB, Gao WC, Pang PP, Ma X, Peng LC, Yang L, Li X. Synthesis and vasorelaxant evaluation of novel 7-methoxyl-2,3-disubstituted-quinoxaline derivatives. Bioorg Med Chem Lett 2021; 36:127785. [PMID: 33444740 DOI: 10.1016/j.bmcl.2021.127785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/11/2020] [Accepted: 01/05/2021] [Indexed: 01/02/2023]
Abstract
An array of novel 7-methoxyl-2,3-disubstituted quinoxaline derivatives was designed, synthesized and their potential antihypertensive activities were examined, in an attempt to discover potent small molecules with vasorelaxant effects. The vasoactivities of these compounds on vascular tone, as well as underlying mechanisms were hereby explored. Results showed that five compounds (7s, 7t, 7v, 7w, 7γ) could induce endothelium-independent relaxation in high extracellular K+- and phenylephrine-precontracted C57 mice aortic rings. These five compounds, unlike other commonly used vasodilators, could slowly but effectively inhibit vasoconstriction.
Collapse
Affiliation(s)
- Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China
| | - Wen-Cong Gao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China
| | - Pan-Pan Pang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China
| | - Xin Ma
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China
| | - Li-Chun Peng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China
| | - Liang Yang
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Science, Shandong University, Ji'nan, Shandong 250012, China
| | - Xun Li
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong 250002, China.
| |
Collapse
|
12
|
Activation of KCNQ4 as a Therapeutic Strategy to Treat Hearing Loss. Int J Mol Sci 2021; 22:ijms22052510. [PMID: 33801540 PMCID: PMC7958948 DOI: 10.3390/ijms22052510] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/16/2021] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
Potassium voltage-gated channel subfamily q member 4 (KCNQ4) is a voltage-gated potassium channel that plays essential roles in maintaining ion homeostasis and regulating hair cell membrane potential. Reduction of the activity of the KCNQ4 channel owing to genetic mutations is responsible for nonsyndromic hearing loss, a typically late-onset, initially high-frequency loss progressing over time. In addition, variants of KCNQ4 have also been associated with noise-induced hearing loss and age-related hearing loss. Therefore, the discovery of small compounds activating or potentiating KCNQ4 is an important strategy for the curative treatment of hearing loss. In this review, we updated the current concept of the physiological role of KCNQ4 in the inner ear and the pathologic mechanism underlying the role of KCNQ4 variants with regard to hearing loss. Finally, we focused on currently developed KCNQ4 activators and their pros and cons, paving the way for the future development of specific KCNQ4 activators as a remedy for hearing loss.
Collapse
|
13
|
Mondejar-Parreño G, Perez-Vizcaino F, Cogolludo A. Kv7 Channels in Lung Diseases. Front Physiol 2020; 11:634. [PMID: 32676036 PMCID: PMC7333540 DOI: 10.3389/fphys.2020.00634] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/18/2020] [Indexed: 12/23/2022] Open
Abstract
Lung diseases constitute a global health concern causing disability. According to WHO in 2016, respiratory diseases accounted for 24% of world population mortality, the second cause of death after cardiovascular diseases. The Kv7 channels family is a group of voltage-dependent K+ channels (Kv) encoded by KCNQ genes that are involved in various physiological functions in numerous cell types, especially, cardiac myocytes, smooth muscle cells, neurons, and epithelial cells. Kv7 channel α-subunits are regulated by KCNE1–5 ancillary β-subunits, which modulate several characteristics of Kv7 channels such as biophysical properties, cell-location, channel trafficking, and pharmacological sensitivity. Kv7 channels are mainly expressed in two large groups of lung tissues: pulmonary arteries (PAs) and bronchial tubes. In PA, Kv7 channels are expressed in pulmonary artery smooth muscle cells (PASMCs); while in the airway (trachea, bronchus, and bronchioles), Kv7 channels are expressed in airway smooth muscle cells (ASMCs), airway epithelial cells (AEPs), and vagal airway C-fibers (VACFs). The functional role of Kv7 channels may vary depending on the cell type. Several studies have demonstrated that the impairment of Kv7 channel has a strong impact on pulmonary physiology contributing to the pathophysiology of different respiratory diseases such as cystic fibrosis, asthma, chronic obstructive pulmonary disease, chronic coughing, lung cancer, and pulmonary hypertension. Kv7 channels are now recognized as playing relevant physiological roles in many tissues, which have encouraged the search for Kv7 channel modulators with potential therapeutic use in many diseases including those affecting the lung. Modulation of Kv7 channels has been proposed to provide beneficial effects in a number of lung conditions. Therefore, Kv7 channel openers/enhancers or drugs acting partly through these channels have been proposed as bronchodilators, expectorants, antitussives, chemotherapeutics and pulmonary vasodilators.
Collapse
Affiliation(s)
- Gema Mondejar-Parreño
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Francisco Perez-Vizcaino
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Angel Cogolludo
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| |
Collapse
|
14
|
Wang L, Qiao GH, Hu HN, Gao ZB, Nan FJ. Discovery of Novel Retigabine Derivatives as Potent KCNQ4 and KCNQ5 Channel Agonists with Improved Specificity. ACS Med Chem Lett 2019; 10:27-33. [PMID: 30655942 DOI: 10.1021/acsmedchemlett.8b00315] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 12/19/2018] [Indexed: 12/22/2022] Open
Abstract
Recent research suggests that KCNQ isoforms, particularly the KCNQ4 and KCNQ5 subtypes expressed in smooth muscle cells, are involved in both establishing and maintaining resting membrane potentials and regulating smooth muscle contractility. Retigabine (RTG) is a first-in-class antiepileptic drug that potentiates neuronal KCNQ potassium channels, but poor subtype selectivity limits its further application as a pharmacological tool. In this study, we improved the subtype specificity of retigabine by altering the N-1/3 substituents and discovered several compounds that show better selectivity for KCNQ4 and KCNQ5 channels. Among these compounds, 10g is highly selective for KCNQ4 and KCNQ5 channels without potentiating KCNQ1 and KCNQ2 channels. These results are an advance in the exploration of small molecule modifiers that selectively activate different KCNQ isoforms. The developed compounds could also serve as new pharmacological tools for elucidating the function of KCNQ channels natively expressed in various tissues.
Collapse
Affiliation(s)
- Lei Wang
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China
| | - Guan-Hua Qiao
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China
| | - Hai-Ning Hu
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhao-Bing Gao
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fa-Jun Nan
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
15
|
Du X, Gao H, Jaffe D, Zhang H, Gamper N. M-type K + channels in peripheral nociceptive pathways. Br J Pharmacol 2018; 175:2158-2172. [PMID: 28800673 PMCID: PMC5980636 DOI: 10.1111/bph.13978] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/17/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022] Open
Abstract
Pathological pain is a hyperexcitability disorder. Since the excitability of a neuron is set and controlled by a complement of ion channels it expresses, in order to understand and treat pain, we need to develop a mechanistic insight into the key ion channels controlling excitability within the mammalian pain pathways and how these ion channels are regulated and modulated in various physiological and pathophysiological settings. In this review, we will discuss the emerging data on the expression in pain pathways, functional role and modulation of a family of voltage-gated K+ channels called 'M channels' (KCNQ, Kv 7). M channels are increasingly recognized as important players in controlling pain signalling, especially within the peripheral somatosensory system. We will also discuss the therapeutic potential of M channels as analgesic drug targets. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc/.
Collapse
Affiliation(s)
- Xiaona Du
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
| | - Haixia Gao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
- School of Biomedical Sciences, Faculty of Biological SciencesUniversity of LeedsLeedsUK
| | - David Jaffe
- Department of Biology, UTSA Neurosciences InstituteUniversity of Texas at San AntonioSan AntonioTXUSA
| | - Hailin Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
| | - Nikita Gamper
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
- School of Biomedical Sciences, Faculty of Biological SciencesUniversity of LeedsLeedsUK
| |
Collapse
|
16
|
Zhao C, Su M, Wang Y, Li X, Zhang Y, Du X, Zhang H. Selective Modulation of K + Channel Kv7.4 Significantly Affects the Excitability of DRN 5-HT Neurons. Front Cell Neurosci 2017; 11:405. [PMID: 29311835 PMCID: PMC5735115 DOI: 10.3389/fncel.2017.00405] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/04/2017] [Indexed: 11/17/2022] Open
Abstract
The serotonin (5-HT) system originating in the dorsal raphe nucleus (DRN) is implicated in various mood- and emotion-related disorders, such as anxiety, fear and stress. Abnormal activity of DRN 5-HT neurons is the key factor in the development of these disorders. Here, we describe a crucial role for the Kv7.4 potassium channel in modulating DRN 5-HT neuronal excitability. We demonstrate that Kv7.4 is selectively expressed in 5-HT neurons of the DRN. Using selective Kv7.4 opener fasudil and Kv7.4 knock-out mice, we demonstrate that Kv7.4 is a potent modulator of DRN 5-HT neuronal excitability. Furthermore, we demonstrate that the cellular redox signaling mechanism is involved in this 5-HT activation of Kv7.4. The current study suggests a new strategy for treating psychiatric disorders related to altered activity of DRN 5-HT neurons using K+ channel modulators.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Min Su
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Yingzi Wang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Xinmeng Li
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Yongxue Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| |
Collapse
|
17
|
Li L, Sun H, Ding J, Niu C, Su M, Zhang L, Li Y, Wang C, Gamper N, Du X, Zhang H. Selective targeting of M-type potassium K v 7.4 channels demonstrates their key role in the regulation of dopaminergic neuronal excitability and depression-like behaviour. Br J Pharmacol 2017; 174:4277-4294. [PMID: 28885682 DOI: 10.1111/bph.14026] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 08/20/2017] [Accepted: 08/21/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The mesolimbic dopamine system originating in the ventral tegmental area (VTA) is involved in the development of depression, and firing patterns of VTA dopaminergic neurons are key determinants in this process. Here, we describe a crucial role for the M-type Kv 7.4 channels in modulating excitability of VTA dopaminergic neurons and in the development of depressive behaviour in mice. EXPERIMENTAL APPROACH We used Kv 7.4 channel knockout mice and a social defeat model of depression in combination with electrophysiological techniques (patch clamp recording and in vivo single-unit recordings), immunohistochemistry, single-cell PCR and behavioural analyses (social interaction time and glucose preference tests) to investigate VTA circuits involved in the development of depression-like behaviour. KEY RESULTS Among the Kv 7 channels, Kv 7.4 channels are selectively expressed in dopaminergic neurons of the VTA. Using a newly identified selective Kv 7.4 channel activator, fasudil, and Kv 7.4 channel knockout mice, we demonstrate that these channels are a dominant modulator of excitability of VTA dopaminergic neurons, in vitro and in vivo. Down-regulation of Kv 7.4 channels could be a causal factor of the altered excitability of VTA dopaminergic neurons and depression-like behaviour. The selective Kv 7.4 channel activator, fasudil, strongly alleviated depression-like behaviour in the social defeat mouse model of depression. CONCLUSION AND IMPLICATIONS Because expression of Kv 7.4 channels in the CNS is limited, selectively targeting this M channel subunit is likely to produce less on-target side effects than non-selective M channel modulators. Thus, Kv 7.4 channels may offer alternative targets in treatment of depression.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hui Sun
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jie Ding
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chenxu Niu
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Min Su
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ludi Zhang
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yingmin Li
- Department of Forensic Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chuan Wang
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Nikita Gamper
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China.,Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Xiaona Du
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hailin Zhang
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
18
|
Townes-Anderson E, Wang J, Halász É, Sugino I, Pitler A, Whitehead I, Zarbin M. Fasudil, a Clinically Used ROCK Inhibitor, Stabilizes Rod Photoreceptor Synapses after Retinal Detachment. Transl Vis Sci Technol 2017; 6:22. [PMID: 28660097 PMCID: PMC5482187 DOI: 10.1167/tvst.6.3.22] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 05/08/2017] [Indexed: 12/20/2022] Open
Abstract
Purpose Retinal detachment disrupts the rod-bipolar synapse in the outer plexiform layer by retraction of rod axons. We showed that breakage is due to RhoA activation whereas inhibition of Rho kinase (ROCK), using Y27632, reduces synaptic damage. We test whether the ROCK inhibitor fasudil, used for other clinical applications, can prevent synaptic injury after detachment. Methods Detachments were made in pigs by subretinal injection of balanced salt solution (BSS) or fasudil (1, 10 mM). In some animals, fasudil was injected intravitreally after BSS-induced detachment. After 2 to 4 hours, retinae were fixed for immunocytochemistry and confocal microscopy. Axon retraction was quantified by imaging synaptic vesicle label in the outer nuclear layer. Apoptosis was analyzed using propidium iodide staining. For biochemical analysis by Western blotting, retinal explants, detached from retinal pigmented epithelium, were cultured for 2 hours. Results Subretinal injection of fasudil (10 mM) reduced retraction of rod spherules by 51.3% compared to control detachments (n = 3 pigs, P = 0.002). Intravitreal injection of 10 mM fasudil, a more clinically feasible route of administration, also reduced retraction (28.7%, n = 5, P < 0.05). Controls had no photoreceptor degeneration at 2 hours, but by 4 hours apoptosis was evident. Fasudil 10 mM reduced pyknotic nuclei by 55.7% (n = 4, P < 0.001). Phosphorylation of cofilin and myosin light chain, downstream effectors of ROCK, was decreased with 30 μM fasudil (n = 8–10 explants, P < 0.05). Conclusions Inhibition of ROCK signaling with fasudil reduced photoreceptor degeneration and preserved the rod-bipolar synapse after retinal detachment. Translational Relevance These results support the possibility, previously tested with Y27632, that ROCK inhibition may attenuate synaptic damage in iatrogenic detachments.
Collapse
Affiliation(s)
- Ellen Townes-Anderson
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jianfeng Wang
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Éva Halász
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Ilene Sugino
- Institute of Ophthalmology and Visual Science, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Amy Pitler
- Department of Microbiology, Biochemistry, and Medical Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Ian Whitehead
- Department of Microbiology, Biochemistry, and Medical Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Marco Zarbin
- Institute of Ophthalmology and Visual Science, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
19
|
Zhang X, An H, Li J, Zhang Y, Liu Y, Jia Z, Zhang W, Chu L, Zhang H. Selective activation of vascular K v 7.4/K v 7.5 K + channels by fasudil contributes to its vasorelaxant effect. Br J Pharmacol 2016; 173:3480-3491. [PMID: 27677924 DOI: 10.1111/bph.13639] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 09/13/2016] [Accepted: 09/15/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Kv 7 (Kv 7.1-7.5) channels play an important role in the regulation of neuronal excitability and the cardiac action potential. Growing evidence suggests Kv 7.4/Kv 7.5 channels play a crucial role in regulating vascular smooth muscle contractility. Most of the reported Kv 7 openers have shown poor selectivity across these five subtypes. In this study, fasudil - a drug used for cerebral vasospasm - has been found to be a selective opener of Kv 7.4/Kv 7.5 channels. EXPERIMENTAL APPROACH A perforated whole-cell patch technique was used to record the currents and membrane potential. Homology modelling and a docking technique were used to investigate the interaction between fasudil and the Kv 7.4 channel. An isometric tension recording technique was used to assess the vascular tension. KEY RESULTS Fasudil selectively and potently enhanced Kv 7.4 and Kv 7.4/Kv 7.5 currents expressed in HEK293 cells, and shifted the voltage-dependent activation curve in a more negative direction. Fasudil did not affect either Kv 7.2 and Kv 7.2/Kv 7.3 currents expressed in HEK293 cells, the native neuronal M-type K+ currents, or the resting membrane potential in small rat dorsal root ganglia neurons. The Val248 in S5 and Ile308 in S6 segment of Kv 7.4 were critical for this activating effect of fasudil. Fasudil relaxed precontracted rat small arteries in a concentration-dependent fashion; this effect was antagonized by the Kv 7 channel blocker XE991. CONCLUSIONS AND IMPLICATIONS These results suggest that fasudil is a selective Kv 7.4/Kv 7.5 channel opener and provide a new dimension for developing selective Kv 7 modulators and a new prospective for the use, action and mechanism of fasudil.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, China.,Department of Pharmacology, Institution of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Hebei Province; Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Junwei Li
- Key Laboratory of Molecular Biophysics, Hebei Province; Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Yuanyuan Zhang
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yang Liu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zhanfeng Jia
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Wei Zhang
- Department of Pharmacology, Institution of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
| | - Li Chu
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|