1
|
Ray S, Stampf JL, Kudlacek O, Yang JW, Schicker KW, Graf Y, Losgott T, Boehm S, Salzer I. A triple cysteine motif as major determinant of the modulation of neuronal K V7 channels by the paracetamol metabolite N-acetyl-p-benzo quinone imine. Br J Pharmacol 2024; 181:2851-2868. [PMID: 38657956 DOI: 10.1111/bph.16380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/19/2024] [Accepted: 03/10/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND AND PURPOSE The analgesic action of paracetamol involves KV7 channels, and its metabolite N-acetyl-p-benzo quinone imine (NAPQI), a cysteine modifying reagent, was shown to increase currents through such channels in nociceptors. Modification of cysteine residues by N-ethylmaleimide, H2O2, or nitric oxide has been found to modulate currents through KV7 channels. The study aims to identify whether, and if so which, cysteine residues in neuronal KV7 channels might be responsible for the effects of NAPQI. EXPERIMENTAL APPROACH To address this question, we used a combination of perforated patch-clamp recordings, site-directed mutagenesis, and mass spectrometry applied to recombinant KV7.1 to KV7.5 channels. KEY RESULTS Currents through the cardiac subtype KV7.1 were reduced by NAPQI. Currents through all other subtypes were increased, either by an isolated shift of the channel voltage dependence to more negative values (KV7.3) or by such a shift combined with increased maximal current levels (KV7.2, KV7.4, KV7.5). A stretch of three cysteine residues in the S2-S3 linker region of KV7.2 was necessary and sufficient to mediate these effects. CONCLUSION AND IMPLICATION The paracetamol metabolite N-acetyl-p-benzo quinone imine (NAPQI) modifies cysteine residues of KV7 subunits and reinforces channel gating in homomeric and heteromeric KV7.2 to KV7.5, but not in KV7.1 channels. In KV7.2, a triple cysteine motif located within the S2-S3 linker region mediates this reinforcement that can be expected to reduce the excitability of nociceptors and to mediate antinociceptive actions of paracetamol.
Collapse
Affiliation(s)
- Sutirtha Ray
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jan-Luca Stampf
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Kudlacek
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jae-Won Yang
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Klaus W Schicker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Yvonne Graf
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Thomas Losgott
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Boehm
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Isabella Salzer
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Urrutia J, Arrizabalaga-Iriondo A, Sanchez-del-Rey A, Martinez-Ibargüen A, Gallego M, Casis O, Revuelta M. Therapeutic role of voltage-gated potassium channels in age-related neurodegenerative diseases. Front Cell Neurosci 2024; 18:1406709. [PMID: 38827782 PMCID: PMC11140135 DOI: 10.3389/fncel.2024.1406709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
Voltage-gated ion channels are essential for membrane potential maintenance, homeostasis, electrical signal production and controlling the Ca2+ flow through the membrane. Among all ion channels, the key regulators of neuronal excitability are the voltage-gated potassium channels (KV), the largest family of K+ channels. Due to the ROS high levels in the aging brain, K+ channels might be affected by oxidative agents and be key in aging and neurodegeneration processes. This review provides new insight about channelopathies in the most studied neurodegenerative disorders, such as Alzheimer Disease, Parkinson's Disease, Huntington Disease or Spinocerebellar Ataxia. The main affected KV channels in these neurodegenerative diseases are the KV1, KV2.1, KV3, KV4 and KV7. Moreover, in order to prevent or repair the development of these neurodegenerative diseases, previous KV channel modulators have been proposed as therapeutic targets.
Collapse
Affiliation(s)
- Janire Urrutia
- Department of Physiology, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Ane Arrizabalaga-Iriondo
- Department of Physiology, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Ana Sanchez-del-Rey
- Department of Otorhinolaryngology, Faculty of Medicine, University of the Basque Country, Bilbao, Spain
| | - Agustín Martinez-Ibargüen
- Department of Otorhinolaryngology, Faculty of Medicine, University of the Basque Country, Bilbao, Spain
| | - Mónica Gallego
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Oscar Casis
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Miren Revuelta
- Department of Physiology, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Bilbao, Spain
| |
Collapse
|
3
|
Katagiri A, Kishimoto S, Okamoto Y, Yamada M, Niwa H, Bereiter DA, Kato T. Effect of chronic intermittent hypoxia on ocular and intraoral mechanical allodynia mediated via the calcitonin gene-related peptide in a rat. Sleep 2024; 47:zsad332. [PMID: 38166171 PMCID: PMC10925949 DOI: 10.1093/sleep/zsad332] [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: 08/09/2023] [Revised: 12/15/2023] [Indexed: 01/04/2024] Open
Abstract
STUDY OBJECTIVES Obstructive sleep apnea, a significant hypoxic condition, may exacerbate several orofacial pain conditions. The study aims to define the involvement of calcitonin gene-related peptide (CGRP) in peripheral and central sensitization and in evoking orofacial mechanical allodynia under chronic intermittent hypoxia (CIH). METHODS Male rats were exposed to CIH. Orofacial mechanical allodynia was assessed using the eyeblink test and the two-bottle preference drinking test. The CGRP-immunoreactive neurons in the trigeminal ganglion (TG), CGRP-positive primary afferents projecting to laminae I-II of the trigeminal spinal subnucleus caudalis (Vc), and neural responses in the second-order neurons of the Vc were determined by immunohistochemistry. CGRP receptor antagonist was administrated in the TG. RESULTS CIH-induced ocular and intraoral mechanical allodynia. CGRP-immunoreactive neurons and activated satellite glial cells (SGCs) were significantly increased in the TG and the number of cFos-immunoreactive cells in laminae I-II of the Vc were significantly higher in CIH rats compared to normoxic rats. Local administration of the CGRP receptor antagonist in the TG of CIH rats attenuated orofacial mechanical allodynia; the number of CGRP-immunoreactive neurons and activated SGCs in the TG, and the density of CGRP-positive primary afferent terminals and the number of cFos-immunoreactive cells in laminae I-II of the Vc were significantly lower compared to vehicle-administrated CIH rats. CONCLUSIONS An increase in CGRP in the TG induced by CIH, as well as orofacial mechanical allodynia and central sensitization of second-order neurons in the Vc, supported the notion that CGRP plays a critical role in CIH-induced orofacial mechanical allodynia.
Collapse
Affiliation(s)
- Ayano Katagiri
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Saki Kishimoto
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Dental Anesthesiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yoshie Okamoto
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masaharu Yamada
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Dental Anesthesiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Hitoshi Niwa
- Department of Dental Anesthesiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - David A Bereiter
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, MN, USA
| | - Takafumi Kato
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
4
|
Steinert JR, Amal H. The contribution of an imbalanced redox signalling to neurological and neurodegenerative conditions. Free Radic Biol Med 2023; 194:71-83. [PMID: 36435368 DOI: 10.1016/j.freeradbiomed.2022.11.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
Nitric oxide and other redox active molecules such as oxygen free radicals provide essential signalling in diverse neuronal functions, but their excess production and insufficient scavenging induces cytotoxic redox stress which is associated with numerous neurodegenerative and neurological conditions. A further component of redox signalling is mediated by a homeostatic regulation of divalent metal ions, the imbalance of which contributes to neuronal dysfunction. Additional antioxidant molecules such as glutathione and enzymes such as super oxide dismutase are involved in maintaining a physiological redox status within neurons. When cellular processes are perturbed and generation of free radicals overwhelms the antioxidants capacity of the neurons, a resulting redox damage leads to neuronal dysfunction and cell death. Cellular sources for production of redox-active molecules may include NADPH oxidases, mitochondria, cytochrome P450 and nitric oxide (NO)-generating enzymes, such as endothelial, neuronal and inducible NO synthases. Several neurodegenerative and developmental neurological conditions are associated with an imbalanced redox state as a result of neuroinflammatory processes leading to nitrosative and oxidative stress. Ongoing research aims at understanding the causes and consequences of such imbalanced redox homeostasis and its role in neuronal dysfunction.
Collapse
Affiliation(s)
- Joern R Steinert
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham, NG7 2NR, UK.
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
5
|
Chen J, Ye H, Zhang J, Li A, Ni Y. Pathogenesis of seizures and epilepsy after stroke. ACTA EPILEPTOLOGICA 2022. [DOI: 10.1186/s42494-021-00068-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
AbstractStroke is the most frequent cause of secondary epilepsy in the elderly. The incidence of cerebral stroke is increasing with the extension of life expectancy, and the prevalence of post-stroke epilepsy (PSE) is rising. There are various seizure types after stroke, and the occurrence of epilepsy is closely related to the type and location of stroke. Moreover, the clinical treatment of post-stroke epilepsy is difficult, which increases the risk of disability and death, and affects the prognosis and quality of life of patients. Now seizure and epilepsy after stroke is more and more get the attention of the medical profession, has been more and more researchers have devoted to seizures after stroke and PSE clinical and basic research, and hope to get a scientific and unified guideline, to give timely and effective treatment, but the exact pathophysiologic mechanism has not yet formed a unified conclusion. It has been found that ion channels, neurotransmitters, proliferation of glial cells, genetics and other factors are involved in the occurrence and development of PSE. In this review, we discuss the pathogenesis of early-onset epileptic seizures and late-onset epilepsy after stroke, in order to provide a basis for clinicians to understand the disease, and expect to provide ideas for future exploration.
Collapse
|
6
|
Spiers JG, Steinert JR. Nitrergic modulation of ion channel function in regulating neuronal excitability. Channels (Austin) 2021; 15:666-679. [PMID: 34802368 PMCID: PMC8632290 DOI: 10.1080/19336950.2021.2002594] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Nitric oxide (NO) signaling in the brain provides a wide range of functional properties in response to neuronal activity. NO exerts its effects through different signaling pathways, namely, through the canonical soluble guanylyl cyclase-mediated cGMP production route and via post-translational protein modifications. The latter pathways comprise cysteine S-nitrosylation and 3-nitrotyrosination of distinct tyrosine residues. Many ion channels are targeted by one or more of these signaling routes, which leads to their functional regulation under physiological conditions or facilities their dysfunction leading to channelopathies in many pathologies. The resulting alterations in ion channel function changes neuronal excitability, synaptic transmission, and action potential propagation. Transient and activity-dependent NO production mediates reversible ion channel modifications via cGMP and S-nitrosylation signaling, whereas more pronounced and longer-term NO production during conditions of elevated oxidative stress leads to increasingly cumulative and irreversible protein 3-nitrotyrosination. The complexity of this regulation and vast variety of target ion channels and their associated functional alterations presents a challenging task in assessing and understanding the role of NO signaling in physiology and disease.
Collapse
Affiliation(s)
- Jereme G Spiers
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Joern R Steinert
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
7
|
Bayasgalan T, Stupniki S, Kovács A, Csemer A, Szentesi P, Pocsai K, Dionisio L, Spitzmaul G, Pál B. Alteration of Mesopontine Cholinergic Function by the Lack of KCNQ4 Subunit. Front Cell Neurosci 2021; 15:707789. [PMID: 34381336 PMCID: PMC8352570 DOI: 10.3389/fncel.2021.707789] [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: 05/10/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022] Open
Abstract
The pedunculopontine nucleus (PPN), a structure known as a cholinergic member of the reticular activating system (RAS), is source and target of cholinergic neuromodulation and contributes to the regulation of the sleep–wakefulness cycle. The M-current is a voltage-gated potassium current modulated mainly by cholinergic signaling. KCNQ subunits ensemble into ion channels responsible for the M-current. In the central nervous system, KCNQ4 expression is restricted to certain brainstem structures such as the RAS nuclei. Here, we investigated the presence and functional significance of KCNQ4 in the PPN by behavioral studies and the gene and protein expressions and slice electrophysiology using a mouse model lacking KCNQ4 expression. We found that this mouse has alterations in the adaptation to changes in light–darkness cycles, representing the potential role of KCNQ4 in the regulation of the sleep–wakefulness cycle. As cholinergic neurons from the PPN participate in the regulation of this cycle, we investigated whether the cholinergic PPN might also possess functional KCNQ4 subunits. Although the M-current is an electrophysiological hallmark of cholinergic neurons, only a subpopulation of them had KCNQ4-dependent M-current. Interestingly, the absence of the KCNQ4 subunit altered the expression patterns of the other KCNQ subunits in the PPN. We also determined that, in wild-type animals, the cholinergic inputs of the PPN modulated the M-current, and these in turn can modulate the level of synchronization between neighboring PPN neurons. Taken together, the KCNQ4 subunit is present in a subpopulation of PPN cholinergic neurons, and it may contribute to the regulation of the sleep–wakefulness cycle.
Collapse
Affiliation(s)
- T Bayasgalan
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - S Stupniki
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional Del Sur (UNS), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - A Kovács
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - A Csemer
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - P Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - K Pocsai
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - L Dionisio
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional Del Sur (UNS), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - G Spitzmaul
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional Del Sur (UNS), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - B Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
8
|
Scheiblich H, Steinert JR. Nitrergic modulation of neuronal excitability in the mouse hippocampus is mediated via regulation of Kv2 and voltage-gated sodium channels. Hippocampus 2021; 31:1020-1038. [PMID: 34047430 DOI: 10.1002/hipo.23366] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/10/2021] [Accepted: 05/19/2021] [Indexed: 12/21/2022]
Abstract
Regulation of neuronal activity is a necessity for communication and information transmission. Many regulatory processes which have been studied provide a complex picture of how neurons can respond to permanently changing functional requirements. One such activity-dependent mechanism involves signaling mediated by nitric oxide (NO). Within the brain, NO is generated in response to neuronal NO synthase (nNOS) activation but NO-dependent pathways regulating neuronal excitability in the hippocampus remain to be fully elucidated. This study was set out to systematically assess the effects of NO on ion channel activities and intrinsic excitabilities of pyramidal neurons within the CA1 region of the mouse hippocampus. We characterized whole-cell potassium and sodium currents, both involved in action potential (AP) shaping and propagation and determined NO-mediated changes in excitabilities and AP waveforms. Our data describe a novel signaling by which NO, in a cGMP-independent manner, suppresses voltage-gated Kv2 potassium and voltage-gated sodium channel activities, thereby widening AP waveforms and reducing depolarization-induced AP firing rates. Our data show that glutathione, which possesses denitrosylating activity, is sufficient to prevent the observed nitrergic effects on potassium and sodium channels, whereas inhibition of cGMP signaling is also sufficient to abolish NO modulation of sodium currents. We propose that NO suppresses both ion channel activities via redox signaling and that an additional cGMP-mediated component is required to exert effects on sodium currents. Both mechanisms result in a dampened excitability and firing ability providing new data on nitrergic activities in the context of activity-dependent regulation of neuronal function following nNOS activation.
Collapse
Affiliation(s)
- Hannah Scheiblich
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Joern R Steinert
- Faculty of Medicine and Health Sciences, University of Nottingham, School of Life Sciences, Queen's Medical Centre, Nottingham, UK
| |
Collapse
|
9
|
Zhuang W, Yan Z. The S2-S3 Loop of Kv7.4 Channels Is Essential for Calmodulin Regulation of Channel Activation. Front Physiol 2021; 11:604134. [PMID: 33551832 PMCID: PMC7854705 DOI: 10.3389/fphys.2020.604134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/23/2020] [Indexed: 11/16/2022] Open
Abstract
Kv7.4 (KCNQ4) voltage-gated potassium channels control excitability in the inner ear and the central auditory pathway. Mutations in Kv7.4 channels result in inherited progressive deafness in humans. Calmodulin (CaM) is crucial for regulating Kv7 channels, but how CaM affects Kv7 activity has remained unclear. Here, based on electrophysiological recordings, we report that the third EF hand (EF3) of CaM controls the calcium-dependent regulation of Kv7.4 activation and that the S2–S3 loop of Kv7.4 is essential for the regulation mediated by CaM. Overexpression of the mutant CaM1234, which loses the calcium binding ability of all four EF hands, facilitates Kv7.4 activation by accelerating activation kinetics and shifting the voltage dependence of activation leftwards. The single mutant CaM3, which loses the calcium binding ability of the EF3, phenocopies facilitating effects of CaM1234 on Kv7.4 activation. Kv7.4 channels co-expressed with wild-type (WT) CaM show inhibited activation when intracellular calcium levels increase, while Kv7.4 channels co-expressed with CaM1234 or CaM3 are insensitive to calcium. Mutations C156A, C157A, C158V, R159, and R161A, which are located within the Kv7.4 S2–S3 loop, dramatically facilitate activation of Kv7.4 channels co-expressed with WT CaM but have no effect on activation of Kv7.4 channels co-expressed with CaM3, indicating that these five mutations decrease the inhibitory effect of Ca2+/CaM. The double mutation C156A/R159A decreases Ca2+/CaM binding and completely abolishes CaM-mediated calcium-dependent regulation of Kv7.4 activation. Taken together, our results provide mechanistic insights into CaM regulation of Kv7.4 activation and highlight the crucial role of the Kv7.4 S2–S3 loop in CaM regulation.
Collapse
Affiliation(s)
- Wenhui Zhuang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Physiology and Biophysics, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Zhiqiang Yan
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Physiology and Biophysics, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China.,Shenzhen Bay Laboratory, Institute of Molecular Physiology, Shenzhen, China
| |
Collapse
|
10
|
Visser EJ, Drummond PD, Lee-Visser JLA. Reduction in Migraine and Headache Frequency and Intensity With Combined Antioxidant Prophylaxis (N-acetylcysteine, Vitamin E, and Vitamin C): A Randomized Sham-Controlled Pilot Study. Pain Pract 2020; 20:737-747. [PMID: 32306462 DOI: 10.1111/papr.12902] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/02/2020] [Accepted: 04/14/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the preventive effects of a combined antioxidant drug (N-acetylcysteine, vitamin E, and vitamin C [NEC]) on migraine outcomes. Migraine is characterized by increased oxidative stress and neurogenic inflammation in the brain; therefore, antioxidants may have a migraine preventive effect. DESIGN Randomized, double-blind, sham-controlled pilot study. SETTING Australian community. SUBJECTS Adults reporting 2 to 8 migraines per month for at least a year. METHODS After a 1-month baseline period, 35 subjects completed 3 months of treatment with NEC (n = 19) or sham (n = 16) capsules. The primary outcome was the difference in mean number of headaches per month between baseline and final month of the trial for NEC and sham groups; secondary outcomes are listed below. RESULTS For NEC there was a significant decrease in mean number of headaches by 3.0 per month (P = 0.004) compared with 1.4 for sham (P = 0.073); there was no significant difference in these changes between the 2 groups (P = 0.052). Average monthly headache (P = 0.041) and migraine frequency (P = 0.018) were significantly less for NEC vs. sham. In NEC subjects, there was a significant decrease in average monthly migraine days (-3.1), moderate/severe headache days (-3.2), migraine duration, headache pain scores, and acute headache medication use. CONCLUSIONS This is the first randomized controlled trial to find that combined antioxidant therapy with NEC reduces headaches and migraines in adult migraineurs. Given the limitations of this pilot study, an adequately powered randomized controlled trial is planned to further investigate antioxidant prophylaxis in migraine.
Collapse
Affiliation(s)
- Eric John Visser
- School of Medicine, University of Notre Dame Australia, Fremantle, Western Australia, Australia
| | | | - Julia L A Lee-Visser
- School of Medicine, University of Notre Dame Australia, Fremantle, Western Australia, Australia
| |
Collapse
|
11
|
Su M, Li L, Wang J, Sun H, Zhang L, Zhao C, Xie Y, Gamper N, Du X, Zhang H. Kv7.4 Channel Contribute to Projection-Specific Auto-Inhibition of Dopamine Neurons in the Ventral Tegmental Area. Front Cell Neurosci 2019; 13:557. [PMID: 31920557 PMCID: PMC6930245 DOI: 10.3389/fncel.2019.00557] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/03/2019] [Indexed: 01/11/2023] Open
Abstract
Dopaminergic neurons in the ventral tegmental area (VTA) encode behavioral patterns important in reward and drug addiction as well as in emotional disorders. These functions of dopamine neurons are directly related to the release of dopamine in the targeted regions of the brain which are, thus, controlled by the excitability of dopamine neurons. One mechanism for modulation of dopamine neuronal excitability is mediated by the auto dopamine type 2 (D2) receptors, through activation of a Kir3/GIRK K+ channel which inhibits the firing of dopamine neurons. In this study, we provide evidence that Kv7.4, in addition to Kir3.2 channels, contributes to dopamine (DA)-mediated auto-inhibition of DA activity projecting to NAc and to basolateral amygdale (BLA). Furthermore, we demonstrate that D2 receptors enhance Kv7.4 currents through Gi/o protein and redox-dependent cellular pathway. Finally, we show this D2 mediated auto-inhibition is blunted in a social defeat mice model of depression, a phenomenon that may contribute to the altered excitability of VTA DA neurons in depressed animals. These results provide a new perspective for understanding the molecular mechanism of the excitability of VTA DA neurons and for potential new strategies against mental disorders involving altered excitability of DA neurons, such as major depression and drug addictions.
Collapse
Affiliation(s)
- Min Su
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Li Li
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Jing Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China.,Department of Pharmacochemistry, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hui Sun
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Ludi Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Chen Zhao
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Ying Xie
- Center for the Experimental Animal, Hebei Medical University, Shijiazhuang, China
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China.,Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| |
Collapse
|
12
|
Weise-Cross L, Resta TC, Jernigan NL. Redox Regulation of Ion Channels and Receptors in Pulmonary Hypertension. Antioxid Redox Signal 2019; 31:898-915. [PMID: 30569735 PMCID: PMC7061297 DOI: 10.1089/ars.2018.7699] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 12/11/2018] [Indexed: 02/06/2023]
Abstract
Significance: Pulmonary hypertension (PH) is characterized by elevated vascular resistance due to vasoconstriction and remodeling of the normally low-pressure pulmonary vasculature. Redox stress contributes to the pathophysiology of this disease by altering the regulation and activity of membrane receptors, K+ channels, and intracellular Ca2+ homeostasis. Recent Advances: Antioxidant therapies have had limited success in treating PH, leading to a growing appreciation that reductive stress, in addition to oxidative stress, plays a role in metabolic and cell signaling dysfunction in pulmonary vascular cells. Reactive oxygen species generation from mitochondria and NADPH oxidases has substantial effects on K+ conductance and membrane potential, and both receptor-operated and store-operated Ca2+ entry. Critical Issues: Some specific redox changes resulting from oxidation, S-nitrosylation, and S-glutathionylation are known to modulate membrane receptor and ion channel activity in PH. However, many sites of regulation that have been elucidated in nonpulmonary cell types have not been tested in the pulmonary vasculature, and context-specific molecular mechanisms are lacking. Future Directions: Here, we review what is known about redox regulation of membrane receptors and ion channels in PH. Further investigation of the mechanisms involved is needed to better understand the etiology of PH and develop better targeted treatment strategies.
Collapse
Affiliation(s)
- Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C. Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L. Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
13
|
Mondéjar-Parreño G, Moral-Sanz J, Barreira B, De la Cruz A, Gonzalez T, Callejo M, Esquivel-Ruiz S, Morales-Cano D, Moreno L, Valenzuela C, Perez-Vizcaino F, Cogolludo A. Activation of K v 7 channels as a novel mechanism for NO/cGMP-induced pulmonary vasodilation. Br J Pharmacol 2019; 176:2131-2145. [PMID: 30883701 DOI: 10.1111/bph.14662] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE The NO/cGMP pathway represents a major physiological signalling controlling tone in pulmonary arteries (PA), and drugs activating this pathway are used to treat pulmonary arterial hypertension. Kv channels expressed in PA smooth muscle cells (PASMCs) are key determinants of vascular tone. We aimed to analyse the contribution of Kv 1.5 and Kv 7 channels in the electrophysiological and vasodilating effects evoked by NO donors and the GC stimulator riociguat in PA. EXPERIMENTAL APPROACH Kv currents were recorded in isolated rat PASMCs using the patch-clamp technique. Vascular reactivity was assessed in a wire myograph. KEY RESULTS The NO donors diethylamine NONOate diethylammonium (DEA-NO) and sodium nitroprusside hyperpolarized the membrane potential and induced a bimodal effect on Kv currents (augmenting the current between -40 and -10 mV and decreasing it at more depolarized potentials). The hyperpolarization and the enhancement of the current were suppressed by Kv 7 channel inhibitors and by the GC inhibitor ODQ but preserved when Kv 1.5 channels were inhibited. Additionally, DEA-NO enhanced Kv 7.5 currents in COS7 cells expressing the KCNQ5 gene. Riociguat increased Kv currents at all potentials ≥-40 mV and induced membrane hyperpolarization. Both effects were prevented by Kv 7 inhibition. Likewise, PA relaxation induced by NO donors and riociguat was attenuated by Kv 7 inhibitors. CONCLUSIONS AND IMPLICATIONS NO donors and riociguat enhance Kv 7 currents, leading to PASMC hyperpolarization. This mechanism contributes to NO/cGMP-induced PA vasodilation. Our study identifies Kv 7 channels as a novel mechanism of action of vasodilator drugs used in the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Javier Moral-Sanz
- Centres for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Bianca Barreira
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Alicia De la Cruz
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Teresa Gonzalez
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Maria Callejo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Sergio Esquivel-Ruiz
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Daniel Morales-Cano
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Laura Moreno
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
14
|
Archer CR, Enslow BT, Taylor AB, De la Rosa V, Bhattacharya A, Shapiro MS. A mutually induced conformational fit underlies Ca 2+-directed interactions between calmodulin and the proximal C terminus of KCNQ4 K + channels. J Biol Chem 2019; 294:6094-6112. [PMID: 30808708 PMCID: PMC6463706 DOI: 10.1074/jbc.ra118.006857] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/24/2019] [Indexed: 11/06/2022] Open
Abstract
Calmodulin (CaM) conveys intracellular Ca2+ signals to KCNQ (Kv7, "M-type") K+ channels and many other ion channels. Whether this "calmodulation" involves a dramatic structural rearrangement or only slight perturbations of the CaM/KCNQ complex is as yet unclear. A consensus structural model of conformational shifts occurring between low nanomolar and physiologically high intracellular [Ca2+] is still under debate. Here, we used various techniques of biophysical chemical analyses to investigate the interactions between CaM and synthetic peptides corresponding to the A and B domains of the KCNQ4 subtype. We found that in the absence of CaM, the peptides are disordered, whereas Ca2+/CaM imposed helical structure on both KCNQ A and B domains. Isothermal titration calorimetry revealed that Ca2+/CaM has higher affinity for the B domain than for the A domain of KCNQ2-4 and much higher affinity for the B domain when prebound with the A domain. X-ray crystallography confirmed that these discrete peptides spontaneously form a complex with Ca2+/CaM, similar to previous reports of CaM binding KCNQ-AB domains that are linked together. Microscale thermophoresis and heteronuclear single-quantum coherence NMR spectroscopy indicated the C-lobe of Ca2+-free CaM to interact with the KCNQ4 B domain (Kd ∼10-20 μm), with increasing Ca2+ molar ratios shifting the CaM-B domain interactions via only the CaM C-lobe to also include the N-lobe. Our findings suggest that in response to increased Ca2+, CaM undergoes lobe switching that imposes a dramatic mutually induced conformational fit to both the proximal C terminus of KCNQ4 channels and CaM, likely underlying Ca2+-dependent regulation of KCNQ gating.
Collapse
Affiliation(s)
- Crystal R Archer
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229; Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Benjamin T Enslow
- the Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Alexander B Taylor
- Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Victor De la Rosa
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Akash Bhattacharya
- Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Mark S Shapiro
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229.
| |
Collapse
|
15
|
Diao Y, Yan W, Sun W, Luo Y, Li J, Yin Y. The dual role of KCNQ/M channels upon OGD or OGD/R insults in cultured cortical neurons of mice: Timing is crucial in targeting M-channels against ischemic injur ies. J Cell Physiol 2018; 234:12714-12726. [PMID: 30523632 DOI: 10.1002/jcp.27889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/13/2018] [Indexed: 01/09/2023]
Abstract
KCNQ/M potassium channels play a vital role in neuronal excitability; however, it is required to explore their pharmacological modulation on N-Methyl- d-aspartic acid receptors (NMDARs)-mediated glutamatergic transmission of neurons upon ischemic insults. In the current study, both presynaptic glutamatergic release and activities of NMDARs were measured by NMDAR-induced miniature excitatory postsynaptic currents (mEPSCs) in cultured cortical neurons of C57 mice undergoing oxygen and glucose deprivation (OGD) or OGD/reperfusion (OGD/R). The KCNQ/M-channel opener, retigabine (RTG), suppressed the overactivation of postsynaptic NMDARs induced by OGD and then NO transient; RTG also decreased OGD-induced neuronal death measured with MTT assay, suggesting the beneficial role of KCNQ/M-channels for the neurons exposed to ischemic insults. However, when the neurons exposed to the subsequent reperfusion, KCNQ/M-channels played a differential role from its protective effect. OGD/R increased presynaptic glutamatergic release, which was further augmented by RTG or decreased by KCNQ/M-channel blocker, XE991. Reactive oxygen species (ROS) were produced partly in a NO-dependent manner. In addition, XE991 decreased neuronal injuries upon reperfusion measured with DCF and PI staining. Meanwhile, the addition of RTG upon OGD or XE991 upon reperfusion can reverse OGD or OGD/R-reduced mitochondrial membrane potential. Our present study indicates the dual role of KCNQ/M-channels in OGD and OGD/R, which will decide the fate of neurons. Provided that activation of KCNQ/M-channels has differential effects on neuronal injuries during OGD or OGD/R, we propose that therapy targeting KCNQ/M-channels may be effective for ischemic injuries but the proper timing is so crucial for the corresponding treatment.
Collapse
Affiliation(s)
- Yu Diao
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Weijie Yan
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Sun
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanlin Luo
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Junfa Li
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yanling Yin
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Zhang YP, Brown RE, Zhang PC, Zhao YT, Ju XH, Song C. DHA, EPA and their combination at various ratios differently modulated Aβ 25-35-induced neurotoxicity in SH-SY5Y cells. Prostaglandins Leukot Essent Fatty Acids 2018; 136:85-94. [PMID: 28778752 DOI: 10.1016/j.plefa.2017.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/01/2017] [Accepted: 07/13/2017] [Indexed: 10/19/2022]
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs), docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), have been reported to prevent neurodegenerative diseases such as Alzheimer's disease (AD) in both experimental and clinical/epidemiological studies. However, whether DHA and EPA from natural products exert similar or different neuroprotective effects and how these n-3 PUFAs target cellular and molecular mechanisms associated with neurodegenerative disease pathogenesis are unknown. In the present study, we used amyloid-β (Aβ)25-35-treated differentiated SH-SY5Y cells as a model of AD to compare the neuroprotective effect of DHA, EPA and their combination at various ratios. Administration of 20μM Aβ25-35 significantly decreased SH-SY5Y cell viability, the expression of nerve growth factor (NGF), its TrkA receptor, and the level of glutathione (GSH) and increased reactive oxygen species (ROS), nitric oxide, tumor necrosis factor (TNF)-α, brain derived neurotrophic factor (BDNF) and its TrkB receptor. Aβ25-35 also increased the Bax/Bcl-2 ratio and the expression of Caspase-3 in these cells. Compared with the Aβ group, pretreatment with DHA/EPA significantly reduced cell death, especially at ratio of 1:1 and 2:1 DHA/EPA or pure DHA. However, the most efficient ratio for reducing changes in ROS and GSH and for decreasing TNF-α appeared at ratio of 1:2 and 1:1, respectively. The ratio of 1:1, 2:1 and pure DHA resulted in significant increase in the level of NGF. Furthermore, pure DHA was the most efficient for reducing Bax/Bcl ratio and Caspase-3 expression. In conclusion, DHA, EPA and their combination differently modulated Aβ25-35-induced neurotoxicity in SH-SY5Y cells by exerting anti-oxidative, anti-inflammatory and neurotrophic effects.
Collapse
Affiliation(s)
- Yong-Ping Zhang
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Canada; Marine Medicine Research and Development Center, Shenzhen Institute of Guangdong Ocean University, Shenzhen 518120, China
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Canada
| | - Ping-Cheng Zhang
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Canada
| | - Yun-Tao Zhao
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; Marine Medicine Research and Development Center, Shenzhen Institute of Guangdong Ocean University, Shenzhen 518120, China
| | - Xiang-Hong Ju
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; Marine Medicine Research and Development Center, Shenzhen Institute of Guangdong Ocean University, Shenzhen 518120, China
| | - Cai Song
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Canada; Marine Medicine Research and Development Center, Shenzhen Institute of Guangdong Ocean University, Shenzhen 518120, China; Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 40402, Taiwan; Departments of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan.
| |
Collapse
|
17
|
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
|
18
|
De Logu F, Nassini R, Landini L, Geppetti P. Pathways of CGRP Release from Primary Sensory Neurons. Handb Exp Pharmacol 2018; 255:65-84. [PMID: 29980913 DOI: 10.1007/164_2018_145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The benefit reported in a variety of clinical trials by a series of small molecule antagonists for the calcitonin gene-related peptide (CGRP) receptor, or four monoclonal antibodies against the neuropeptide or its receptor, has underscored the release of CGRP from terminals of primary sensory neurons, including trigeminal neurons, as one of the major mechanisms of migraine headaches. A large variety of excitatory ion channels and receptors have been reported to elicit CGRP release, thus proposing these agonists as migraine-provoking agents. On the other side, activators of inhibitory channels and receptors may be regarded as potential antimigraine agents. The knowledge of the intracellular pathways underlying the exocytotic process that results in CGRP secretion or its inhibition is, therefore, of importance for understanding how migraine pain originates and how to treat the disease.
Collapse
Affiliation(s)
- Francesco De Logu
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, Headache Center, University of Florence, Florence, Italy
| | - Romina Nassini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, Headache Center, University of Florence, Florence, Italy
| | - Lorenzo Landini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, Headache Center, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, Headache Center, University of Florence, Florence, Italy.
| |
Collapse
|
19
|
The Kv7/KCNQ channel blocker XE991 protects nigral dopaminergic neurons in the 6-hydroxydopamine rat model of Parkinson's disease. Brain Res Bull 2017; 137:132-139. [PMID: 29174294 DOI: 10.1016/j.brainresbull.2017.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/11/2017] [Accepted: 11/18/2017] [Indexed: 12/21/2022]
Abstract
The excitability of dopaminergic neurons in the substantia nigra pars compacta (SNc) that supply the striatum with dopamine (DA) determines the function of the nigrostriatal system for motor coordination. We previously showed that 4-pyridinylmethyl-9(10H)-anthracenone (XE991), a specific blocker of Kv7/KCNQ channels, enhanced the excitability of nigral DA neurons and resulted in attenuation of haloperidol-induced catalepsy in a Parkinson's disease (PD) rat model. However, whether XE991 exhibits neuroprotective effects towards DA neuron degeneration remains unknown. The aim of this study was to investigate the effects of Kv7/KCNQ channel blocker, XE991, on 6-hydroxydopamine (6-OHDA)-induced nigral DA neuron degeneration and motor dysfunction. Using immunofluorescence staining and western blotting, we showed that intracerebroventricular administration of XE991 prevented the 6-OHDA-induced decrease in tyrosine hydroxylase (TH)-positive neurons and TH protein expression in the SNc. High-performance liquid chromatography with electrochemical detection (HPLC-ECD) also revealed that XE991 partly restored the levels of DA and its metabolites in the striatum. Moreover, XE991 decreased apomorphine (APO)-induced contralateral rotations, enhanced balance and coordination, and attenuated muscle rigidity in 6-OHDA-treated rats. Importantly, all neuroprotective effects by XE991 were abolished by co-application of Kv7/KCNQ channel opener retigabine and XE991. Thus, Kv7/KCNQ channel inhibition by XE991 can exert neuroprotective effects against 6-OHDA-induced degeneration of the nigrostriatal DA system and motor dysfunction.
Collapse
|
20
|
Tse JKY. Gut Microbiota, Nitric Oxide, and Microglia as Prerequisites for Neurodegenerative Disorders. ACS Chem Neurosci 2017. [PMID: 28640632 DOI: 10.1021/acschemneuro.7b00176] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Regulating fluctuating endogenous nitric oxide (NO) levels is necessary for proper physiological functions. Aberrant NO pathways are implicated in a number of neurological disorders, including Alzheimer's disease (AD) and Parkinson's disease. The mechanism of NO in oxidative and nitrosative stress with pathological consequences involves reactions with reactive oxygen species (e.g., superoxide) to form the highly reactive peroxynitrite, hydrogen peroxide, hypochloride ions and hydroxyl radical. NO levels are typically regulated by endogenous nitric oxide synthases (NOS), and inflammatory iNOS is implicated in the pathogenesis of neurodegenerative diseases, in which elevated NO mediates axonal degeneration and activates cyclooxygenases to provoke neuroinflammation. NO also instigates a down-regulated secretion of brain-derived neurotrophic factor, which is essential for neuronal survival, development and differentiation, synaptogenesis, and learning and memory. The gut-brain axis denotes communication between the enteric nervous system (ENS) of the GI tract and the central nervous system (CNS) of the brain, and the modes of communication include the vagus nerve, passive diffusion and carrier by oxyhemoglobin. Amyloid precursor protein that forms amyloid beta plaques in AD is normally expressed in the ENS by gut bacteria, but when amyloid beta accumulates, it compromises CNS functions. Escherichia coli and Salmonella enterica are among the many bacterial strains that express and secrete amyloid proteins and contribute to AD pathogenesis. Gut microbiota is essential for regulating microglia maturation and activation, and activated microglia secrete significant amounts of iNOS. Pharmacological interventions and lifestyle modifications to rectify aberrant NO signaling in AD include NOS inhibitors, NMDA receptor antagonists, potassium channel modulators, probiotics, diet, and exercise.
Collapse
Affiliation(s)
- Joyce K. Y. Tse
- University Research Facility in Chemical
and Environmental Analysis, and Department of Civil and Environmental
Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| |
Collapse
|
21
|
Intracellular zinc activates KCNQ channels by reducing their dependence on phosphatidylinositol 4,5-bisphosphate. Proc Natl Acad Sci U S A 2017; 114:E6410-E6419. [PMID: 28716904 DOI: 10.1073/pnas.1620598114] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
M-type (Kv7, KCNQ) potassium channels are proteins that control the excitability of neurons and muscle cells. Many physiological and pathological mechanisms of excitation operate via the suppression of M channel activity or expression. Conversely, pharmacological augmentation of M channel activity is a recognized strategy for the treatment of hyperexcitability disorders such as pain and epilepsy. However, physiological mechanisms resulting in M channel potentiation are rare. Here we report that intracellular free zinc directly and reversibly augments the activity of recombinant and native M channels. This effect is mechanistically distinct from the known redox-dependent KCNQ channel potentiation. Interestingly, the effect of zinc cannot be attributed to a single histidine- or cysteine-containing zinc-binding site within KCNQ channels. Instead, zinc dramatically reduces KCNQ channel dependence on its obligatory physiological activator, phosphatidylinositol 4,5-bisphosphate (PIP2). We hypothesize that zinc facilitates interactions of the lipid-facing interface of a KCNQ protein with the inner leaflet of the plasma membrane in a way similar to that promoted by PIP2 Because zinc is increasingly recognized as a ubiquitous intracellular second messenger, this discovery might represent a hitherto unknown native pathway of M channel modulation and provide a fresh strategy for the design of M channel activators for therapeutic purposes.
Collapse
|
22
|
Deyama S, Sugano Y, Mori S, Amano T, Yoshioka M, Kaneda K, Minami M. Activation of the NMDA receptor–neuronal nitric oxide synthase pathway within the ventral bed nucleus of the stria terminalis mediates the negative affective component of pain. Neuropharmacology 2017; 118:59-68. [DOI: 10.1016/j.neuropharm.2017.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/05/2017] [Accepted: 03/06/2017] [Indexed: 12/23/2022]
|
23
|
Zhang Y, Jiao G, Song C, Gu S, Brown RE, Zhang J, Zhang P, Gagnon J, Locke S, Stefanova R, Pelletier C, Zhang Y, Lu H. An Extract from Shrimp Processing By-Products Protects SH-SY5Y Cells from Neurotoxicity Induced by Aβ 25-35. Mar Drugs 2017; 15:md15030083. [PMID: 28327516 PMCID: PMC5367040 DOI: 10.3390/md15030083] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/07/2017] [Accepted: 03/15/2017] [Indexed: 12/14/2022] Open
Abstract
Increased evidence suggests that marine unsaturated fatty acids (FAs) can protect neurons from amyloid-β (Aβ)-induced neurodegeneration. Nuclear magnetic resonance (NMR), high performance liquid chromatography (HPLC) and gas chromatography (GC) assays showed that the acetone extract 4-2A obtained from shrimp Pandalus borealis industry processing wastes contained 67.19% monounsaturated FAs and 16.84% polyunsaturated FAs. The present study evaluated the anti-oxidative and anti-inflammatory effects of 4-2A in Aβ25–35-insulted differentiated SH-SY5Y cells. Cell viability and cytotoxicity were measured by using 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) assays. Quantitative PCR and Western blotting were used to study the expression of neurotrophins, pro-inflammatory cytokines and apoptosis-related genes. Administration of 20 μM Aβ25–35 significantly reduced SH-SY5Y cell viability, the expression of nerve growth factor (NGF) and its tyrosine kinase TrkA receptor, as well as the level of glutathione, while increased reactive oxygen species (ROS), nitric oxide, tumor necrosis factor (TNF)-α, brain derived neurotrophic factor (BDNF) and its TrkB receptor. Aβ25–35 also increased the Bax/Bcl-2 ratio and Caspase-3 expression. Treatment with 4-2A significantly attenuated the Aβ25–35-induced changes in cell viability, ROS, GSH, NGF, TrkA, TNF-α, the Bax/Bcl-2 ratio and Caspase-3, except for nitric oxide, BDNF and TrKB. In conclusion, 4-2A effectively protected SH-SY5Y cells against Aβ-induced neuronal apoptosis/death by suppressing inflammation and oxidative stress and up-regulating NGF and TrKA expression.
Collapse
Affiliation(s)
- Yongping Zhang
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China.
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Guangling Jiao
- Coastal Zones Research Institute Inc., 232B, avenue de l'Église, Shippagan, NB E8S 1J2, Canada.
- Aquatic and Crop Resource Development, National Research Council of Canada, 1411 Oxford Street, Halifax, NS B3H 3Z1, Canada.
| | - Cai Song
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China.
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
- Graduate Institute of Neural and Cognitive Sciences, China Medical University Hospital, Taichung 40402, Taiwan.
| | - Shelly Gu
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Junzeng Zhang
- Aquatic and Crop Resource Development, National Research Council of Canada, 1411 Oxford Street, Halifax, NS B3H 3Z1, Canada.
| | - Pingcheng Zhang
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Jacques Gagnon
- Coastal Zones Research Institute Inc., 232B, avenue de l'Église, Shippagan, NB E8S 1J2, Canada.
| | - Steven Locke
- Aquatic and Crop Resource Development, National Research Council of Canada, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada.
| | - Roumiana Stefanova
- Aquatic and Crop Resource Development, National Research Council of Canada, 1411 Oxford Street, Halifax, NS B3H 3Z1, Canada.
| | - Claude Pelletier
- Coastal Zones Research Institute Inc., 232B, avenue de l'Église, Shippagan, NB E8S 1J2, Canada.
| | - Yi Zhang
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China.
| | - Hongyu Lu
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Poor management of chronic pain remains a significant cause of misery with huge socioeconomic costs. Accumulating research in potassium (K+) channel physiology has uncovered several promising leads for the development of novel analgesics. RECENT FINDINGS We now recognize that certain K+ channel subunits are directly gated to pain-relevant stimuli (Kv1.1, K2P) whereas others are specifically modulated by inflammatory processes (Kv7, BKCA, K2P). Genetic analyses illustrate that K+ channel gene variation can predict pain sensitivity (KCNS1, GIRKs), risk for persistent pain (KCNS1, GIRKs, TRESK) and analgesic effectiveness (GIRK2). Importantly, preclinical studies confirm that K+ channel dysfunction can be a pain trigger in traumatic neuropathies (Kv9.1/Kv2.1, Kv7, Kv1.2) and migraine (TRESK). Finally, emerging data suggest that even pain in diabetes, bone cancer and autoimmune neuropathies may have K+ channel dysfunction constituents. SUMMARY There is a long-sought need for superior pharmacotherapy of pain syndromes. Although universal enhancement of K+ channel function in the periphery can decrease nociceptive excitability irrespective of the underlying cause, a more refined targeting of subunits with dominant nociceptive roles could yield highly efficacious treatments with fewer side-effects. The ongoing characterization of molecular interactions linking K+ channel dysfunction to pain is instrumental for identifying candidates with the most therapeutic potential.
Collapse
|
25
|
Greenberg HZE, Shi J, Jahan KS, Martinucci MC, Gilbert SJ, Vanessa Ho WS, Albert AP. Stimulation of calcium-sensing receptors induces endothelium-dependent vasorelaxations via nitric oxide production and activation of IKCa channels. Vascul Pharmacol 2016; 80:75-84. [PMID: 26772767 PMCID: PMC4830458 DOI: 10.1016/j.vph.2016.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/09/2015] [Accepted: 01/02/2016] [Indexed: 11/25/2022]
Abstract
Stimulation of vascular calcium-sensing receptors (CaSRs) is reported to induce both constrictions and relaxations. However, cellular mechanisms involved in these responses remain unclear. The present study investigates the effect of stimulating CaSRs on vascular contractility and focuses on the role of the endothelium, nitric oxide (NO) and K(+) channels in these responses. In wire myography studies, increasing [Ca(2+)]o from 1mM to 6mM induced concentration-dependent relaxations of methoxamine pre-contracted rabbit mesenteric arteries. [Ca(2+)]o-induced relaxations were dependent on a functional endothelium, and were inhibited by the negative allosteric CaSR modulator Calhex-231. [Ca(2+)]o-induced relaxations were reduced by inhibitors of endothelial NO synthase, guanylate cyclase, and protein kinase G. CaSR activation also induced NO production in freshly isolated endothelial cells (ECs) in experiments using the fluorescent NO indicator DAF-FM. Pre-treatment with inhibitors of large (BKCa) and intermediate (IKCa) Ca(2+)-activated K(+) channels (iberiotoxin and charybdotoxin), and Kv7 channels (linopirdine) also reduced [Ca(2+)]o-induced vasorelaxations. Increasing [Ca(2+)]o also activated IKCa currents in perforated-patch recordings of isolated mesenteric artery ECs. These findings indicate that stimulation of CaSRs induces endothelium-dependent vasorelaxations which are mediated by two separate pathways involving production of NO and activation of IKCa channels. NO stimulates PKG leading to BKCa activation in vascular smooth muscle cells, whereas IKCa activity contributes to endothelium-derived hyperpolarisations.
Collapse
Affiliation(s)
- Harry Z E Greenberg
- Vascular Biology Research Centre, Institute of Cardiovascular & Cell Sciences, St. George's, University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Jian Shi
- Vascular Biology Research Centre, Institute of Cardiovascular & Cell Sciences, St. George's, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Kazi S Jahan
- Vascular Biology Research Centre, Institute of Cardiovascular & Cell Sciences, St. George's, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Matthew C Martinucci
- Vascular Biology Research Centre, Institute of Cardiovascular & Cell Sciences, St. George's, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Steven J Gilbert
- Vascular Biology Research Centre, Institute of Cardiovascular & Cell Sciences, St. George's, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - W-S Vanessa Ho
- Vascular Biology Research Centre, Institute of Cardiovascular & Cell Sciences, St. George's, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Anthony P Albert
- Vascular Biology Research Centre, Institute of Cardiovascular & Cell Sciences, St. George's, University of London, Cranmer Terrace, London SW17 0RE, UK
| |
Collapse
|
26
|
Martin CA, Myers KM, Chen A, Martin NT, Barajas A, Schweizer FE, Krantz DE. Ziram, a pesticide associated with increased risk for Parkinson's disease, differentially affects the presynaptic function of aminergic and glutamatergic nerve terminals at the Drosophila neuromuscular junction. Exp Neurol 2016; 275 Pt 1:232-41. [PMID: 26439313 PMCID: PMC4688233 DOI: 10.1016/j.expneurol.2015.09.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 09/04/2015] [Accepted: 09/26/2015] [Indexed: 12/29/2022]
Abstract
Multiple populations of aminergic neurons are affected in Parkinson's disease (PD), with serotonergic and noradrenergic loci responsible for some non-motor symptoms. Environmental toxins, such as the dithiocarbamate fungicide ziram, significantly increase the risk of developing PD and the attendant spectrum of both motor and non-motor symptoms. The mechanisms by which ziram and other environmental toxins increase the risk of PD, and the potential effects of these toxins on aminergic neurons, remain unclear. To determine the relative effects of ziram on the synaptic function of aminergic versus non-aminergic neurons, we used live-imaging at the Drosophila melanogaster larval neuromuscular junction (NMJ). In contrast to nearly all other studies of this model synapse, we imaged presynaptic function at both glutamatergic Type Ib and aminergic Type II boutons, the latter responsible for storage and release of octopamine, the invertebrate equivalent of noradrenalin. To quantify the kinetics of exo- and endo-cytosis, we employed an acid-sensitive form of GFP fused to the Drosophila vesicular monoamine transporter (DVMAT-pHluorin). Additional genetic probes were used to visualize intracellular calcium flux (GCaMP) and voltage changes (ArcLight). We find that at glutamatergic Type Ib terminals, exposure to ziram increases exocytosis and inhibits endocytosis. By contrast, at octopaminergic Type II terminals, ziram has no detectable effect on exocytosis and dramatically inhibits endocytosis. In contrast to other reports on the neuronal effects of ziram, these effects do not appear to result from perturbation of the Ubiquitin Proteasome System (UPS) or calcium homeostasis. Unexpectedly, ziram also caused spontaneous and synchronized bursts of calcium influx (measured by GCaMP) and electrical activity (measured by ArcLight) at aminergic Type II, but not glutamatergic Type Ib, nerve terminals. These events are sensitive to both tetrodotoxin and cadmium chloride, and thus appear to represent spontaneous depolarizations followed by calcium influx into Type II terminals. We speculate that the differential effects of ziram on Type II versus Type Ib terminals may be relevant to the specific sensitivity of aminergic neurons in PD, and suggest that changes in neuronal excitability could contribute to the increased risk for PD caused by exposure to ziram. We also suggest that the fly NMJ will be useful to explore the synaptic effects of other pesticides associated with an increased risk of PD.
Collapse
Affiliation(s)
- Ciara A Martin
- Department of Psychiatry and Biobehavioral Sciences, Hatos Center for Neuropharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, United States; UCLA Interdepartmental Program in Molecular Toxicology, Los Angeles, CA 90095, United States.
| | - Katherine M Myers
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, United States; UCLA Interdepartmental Program for Neuroscience, Los Angeles, CA 90095, United States.
| | - Audrey Chen
- Department of Psychiatry and Biobehavioral Sciences, Hatos Center for Neuropharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, United States; Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, United States.
| | - Nathan T Martin
- UCLA Biomedical Physics Interdepartmental Graduate Program, Los Angeles, CA 90095, United States.
| | - Angel Barajas
- Department of Psychiatry and Biobehavioral Sciences, Hatos Center for Neuropharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, United States.
| | - Felix E Schweizer
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, United States; UCLA Interdepartmental Program for Neuroscience, Los Angeles, CA 90095, United States.
| | - David E Krantz
- Department of Psychiatry and Biobehavioral Sciences, Hatos Center for Neuropharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, United States; UCLA Interdepartmental Program in Molecular Toxicology, Los Angeles, CA 90095, United States; UCLA Interdepartmental Program for Neuroscience, Los Angeles, CA 90095, United States.
| |
Collapse
|
27
|
Getting to NO Alzheimer's Disease: Neuroprotection versus Neurotoxicity Mediated by Nitric Oxide. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3806157. [PMID: 26697132 PMCID: PMC4677236 DOI: 10.1155/2016/3806157] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/16/2015] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder involving the loss of neurons in the brain which leads to progressive memory loss and behavioral changes. To date, there are only limited medications for AD and no known cure. Nitric oxide (NO) has long been considered part of the neurotoxic insult caused by neuroinflammation in the Alzheimer's brain. However, focusing on early developments, prior to the appearance of cognitive symptoms, is changing that perception. This has highlighted a compensatory, neuroprotective role for NO that protects synapses by increasing neuronal excitability. A potential mechanism for augmentation of excitability by NO is via modulation of voltage-gated potassium channel activity (Kv7 and Kv2). Identification of the ionic mechanisms and signaling pathways that mediate this protection is an important next step for the field. Harnessing the protective role of NO and related signaling pathways could provide a therapeutic avenue that prevents synapse loss early in disease.
Collapse
|
28
|
Nitric Oxide-Mediated Posttranslational Modifications: Impacts at the Synapse. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:5681036. [PMID: 26635909 PMCID: PMC4655263 DOI: 10.1155/2016/5681036] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/28/2015] [Indexed: 11/18/2022]
Abstract
Nitric oxide (NO) is an important gasotransmitter molecule that is involved in numerous physiological processes throughout the nervous system. In addition to its involvement in physiological plasticity processes (long-term potentiation, LTP; long-term depression, LTD) which can include NMDAR-mediated calcium-dependent activation of neuronal nitric oxide synthase (nNOS), new insights into physiological and pathological consequences of nitrergic signalling have recently emerged. In addition to the canonical cGMP-mediated signalling, NO is also implicated in numerous pathways involving posttranslational modifications. In this review we discuss the multiple effects of S-nitrosylation and 3-nitrotyrosination on proteins with potential modulation of function but limit the analyses to signalling involved in synaptic transmission and vesicular release. Here, crucial proteins which mediate synaptic transmission can undergo posttranslational modifications with either pre- or postsynaptic origin. During normal brain function, both pathways serve as important cellular signalling cascades that modulate a diverse array of physiological processes, including synaptic plasticity, transcriptional activity, and neuronal survival. In contrast, evidence suggests that aging and disease can induce nitrosative stress via excessive NO production. Consequently, uncontrolled S-nitrosylation/3-nitrotyrosination can occur and represent pathological features that contribute to the onset and progression of various neurodegenerative diseases, including Parkinson's, Alzheimer's, and Huntington's.
Collapse
|
29
|
Neuroprotection of Neuro2a cells and the cytokine suppressive and anti-inflammatory mode of action of resveratrol in activated RAW264.7 macrophages and C8-B4 microglia. Neurochem Int 2015; 95:46-54. [PMID: 26522689 DOI: 10.1016/j.neuint.2015.10.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 09/08/2015] [Accepted: 10/24/2015] [Indexed: 02/07/2023]
Abstract
Chronic inflammation is a hallmark of neurodegenerative disease and cytotoxic levels of nitric oxide (NO) and pro-inflammatory cytokines can initiate neuronal death pathways. A range of cellular assays were used to assess the anti-inflammatory and neuroprotective action of resveratrol using murine microglial (C8-B4), macrophage (RAW264.7) and neuronal-like (Neuro2a) cell lines. We examined the release of NO by Griess assay and used a Bioplex array to measure a panel of pro- and anti-inflammatory cytokines and chemokines, in response to the inflammatory stimuli lipopolysaccharide (LPS) and interferon-γ (IFN-γ). Resveratrol was a potent inhibitor of NO and cytokine release in activated macrophages and microglia. The activity of resveratrol increased marginally in potency with longer pre-incubation times in cell culture that was not due to cytotoxicity. Using an NO donor we show that resveratrol can protect Neuro2a cells from cytotoxic concentrations of NO. The protective effect of resveratrol from pro-inflammatory signalling in RAW264.7 cells was confirmed in co-culture experiments leading to increased survival of Neuro2a cells. Together our data are indicative of the potential neuroprotective effect of resveratrol during nitrosative stress and neuroinflammation.
Collapse
|
30
|
Hermann A, Sitdikova GF, Weiger TM. Oxidative Stress and Maxi Calcium-Activated Potassium (BK) Channels. Biomolecules 2015; 5:1870-911. [PMID: 26287261 PMCID: PMC4598779 DOI: 10.3390/biom5031870] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 01/13/2023] Open
Abstract
All cells contain ion channels in their outer (plasma) and inner (organelle) membranes. Ion channels, similar to other proteins, are targets of oxidative impact, which modulates ion fluxes across membranes. Subsequently, these ion currents affect electrical excitability, such as action potential discharge (in neurons, muscle, and receptor cells), alteration of the membrane resting potential, synaptic transmission, hormone secretion, muscle contraction or coordination of the cell cycle. In this chapter we summarize effects of oxidative stress and redox mechanisms on some ion channels, in particular on maxi calcium-activated potassium (BK) channels which play an outstanding role in a plethora of physiological and pathophysiological functions in almost all cells and tissues. We first elaborate on some general features of ion channel structure and function and then summarize effects of oxidative alterations of ion channels and their functional consequences.
Collapse
Affiliation(s)
- Anton Hermann
- Department of Cell Biology, Division of Cellular and Molecular Neurobiology, University of Salzburg, Salzburg 5020, Austria.
| | - Guzel F Sitdikova
- Department of Physiology of Man and Animals, Kazan Federal University, Kazan 420008, Russia.
| | - Thomas M Weiger
- Department of Cell Biology, Division of Cellular and Molecular Neurobiology, University of Salzburg, Salzburg 5020, Austria.
| |
Collapse
|
31
|
Huang D, Huang S, Peers C, Du X, Zhang H, Gamper N. GABAB receptors inhibit low-voltage activated and high-voltage activated Ca(2+) channels in sensory neurons via distinct mechanisms. Biochem Biophys Res Commun 2015; 465:188-93. [PMID: 26239659 DOI: 10.1016/j.bbrc.2015.07.137] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 07/28/2015] [Indexed: 12/16/2022]
Abstract
Growing evidence suggests that mammalian peripheral somatosensory neurons express functional receptors for gamma-aminobutyric acid, GABAA and GABAB. Moreover, local release of GABA by pain-sensing (nociceptive) nerve fibres has also been suggested. Yet, the functional significance of GABA receptor triggering in nociceptive neurons is not fully understood. Here we used patch-clamp recordings from small-diameter cultured DRG neurons to investigate effects of GABAB receptor agonist baclofen on voltage-gated Ca(2+) currents. We found that baclofen inhibited both low-voltage activated (LVA, T-type) and high-voltage activated (HVA) Ca(2+) currents in a proportion of DRG neurons by 22% and 32% respectively; both effects were sensitive to Gi/o inhibitor pertussis toxin. Inhibitory effect of baclofen on both current types was about twice less efficacious as compared to that of the μ-opioid receptor agonist DAMGO. Surprisingly, only HVA but not LVA current modulation by baclofen was partially prevented by G protein inhibitor GDP-β-S. In contrast, only LVA but not HVA current modulation was reversed by the application of a reducing agent dithiothreitol (DTT). Inhibition of T-type Ca(2+) current by baclofen and the recovery of such inhibition by DTT were successfully reconstituted in the expression system. Our data suggest that inhibition of LVA current in DRG neurons by baclofen is partially mediated by an unconventional signaling pathway that involves a redox mechanism. These findings reinforce the idea of targeting peripheral GABA receptors for pain relief.
Collapse
Affiliation(s)
- Dongyang Huang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, 050011, PR China
| | - Sha Huang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, 050011, PR China
| | - Chris Peers
- Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, 050011, PR China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, 050011, PR China.
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, 050011, PR China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
32
|
Augmentation of M-type (KCNQ) potassium channels as a novel strategy to reduce stroke-induced brain injury. J Neurosci 2015; 35:2101-11. [PMID: 25653366 DOI: 10.1523/jneurosci.3805-14.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cerebral ischemic stroke is a worldwide cause of mortality/morbidity and thus an important focus of research to decrease the severity of brain injury. Therapeutic options for acute stroke are still limited. In neurons throughout the brain, "M-type" K(+) currents, underlain by KCNQ subunits 2-5, play dominant roles in control over excitability, and are thus implicated in myriad neurological and psychiatric disorders. Although KCNQ channel openers, such as retigabine, have emerged as anti-epilepsy drugs, their effects on ischemic injury remain unknown. Here, we investigated the protective effects of M-channel openers on stroke-induced brain injury in mouse photothrombotic and middle cerebral artery occlusion (MCAo) models. Both photothrombosis and MCAo led to rapid, predictable, and consistently sized necrotic brain lesions, inflammatory responses, and behavioral deficits. Administration of three distinct M-channel openers at 0-6 h after ischemic injury significantly decreased brain infarct size and inflammation, and prevented neurological dysfunction, although they were more effective when administered 0-3 h poststroke. Thus, we show beneficial effects against stroke-induced brain injury and neuronal death through pharmacological regulation of ion channels that control neuronal excitability.
Collapse
|
33
|
Sforna L, D'Adamo MC, Servettini I, Guglielmi L, Pessia M, Franciolini F, Catacuzzeno L. Expression and function of a CP339,818-sensitive K⁺ current in a subpopulation of putative nociceptive neurons from adult mouse trigeminal ganglia. J Neurophysiol 2015; 113:2653-65. [PMID: 25652918 PMCID: PMC4416569 DOI: 10.1152/jn.00379.2014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 02/02/2015] [Indexed: 01/15/2023] Open
Abstract
Trigeminal ganglion (TG) neurons are functionally and morphologically heterogeneous, and the molecular basis of this heterogeneity is still not fully understood. Here we describe experiments showing that a subpopulation of neurons expresses a delayed-rectifying K(+) current (IDRK) with a characteristically high (nanomolar) sensitivity to the dihydroquinoline CP339,818 (CP). Although submicromolar CP has previously been shown to selectively block Kv1.3 and Kv1.4 channels, the CP-sensitive IDRK found in TG neurons could not be associated with either of these two K(+) channels. It could neither be associated with Kv2.1 channels homomeric or heteromerically associated with the Kv9.2, Kv9.3, or Kv6.4 subunits, whose block by CP, tested using two-electrode voltage-clamp recordings from Xenopus oocytes, resulted in the low micromolar range, nor to the Kv7 subfamily, given the lack of blocking efficacy of 3 μM XE991. Within the group of multiple-firing neurons considered in this study, the CP-sensitive IDRK was preferentially expressed in a subpopulation showing several nociceptive markers, such as small membrane capacitance, sensitivity to capsaicin, and slow afterhyperpolarization (AHP); in these neurons the CP-sensitive IDRK controls the membrane resting potential, the firing frequency, and the AHP duration. A biophysical study of the CP-sensitive IDRK indicated the presence of two kinetically distinct components: a fast deactivating component having a relatively depolarized steady-state inactivation (IDRKf) and a slow deactivating component with a more hyperpolarized V1/2 for steady-state inactivation (IDRKs).
Collapse
Affiliation(s)
- Luigi Sforna
- Dipartimento di Chimica, Biologia e Biotecnologie, Universitá di Perugia, Perugia, Italy; and
| | - Maria Cristina D'Adamo
- Dipartimento di Medicina Sperimentale, Facoltá di Medicina e Chirurgia, Universitá di Perugia, Perugia, Italy
| | - Ilenio Servettini
- Dipartimento di Medicina Sperimentale, Facoltá di Medicina e Chirurgia, Universitá di Perugia, Perugia, Italy
| | - Luca Guglielmi
- Dipartimento di Medicina Sperimentale, Facoltá di Medicina e Chirurgia, Universitá di Perugia, Perugia, Italy
| | - Mauro Pessia
- Dipartimento di Medicina Sperimentale, Facoltá di Medicina e Chirurgia, Universitá di Perugia, Perugia, Italy
| | - Fabio Franciolini
- Dipartimento di Chimica, Biologia e Biotecnologie, Universitá di Perugia, Perugia, Italy; and
| | - Luigi Catacuzzeno
- Dipartimento di Chimica, Biologia e Biotecnologie, Universitá di Perugia, Perugia, Italy; and
| |
Collapse
|
34
|
Bradley SA, Steinert JR. Characterisation and comparison of temporal release profiles of nitric oxide generating donors. J Neurosci Methods 2015; 245:116-24. [PMID: 25749567 PMCID: PMC4401449 DOI: 10.1016/j.jneumeth.2015.02.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/19/2015] [Accepted: 02/20/2015] [Indexed: 01/11/2023]
Abstract
Nitric oxide release profiles were characterised for commonly used donors. Released NO differs greatly between donors and depends on storage conditions. High release donors (NOC-5, PAPA NONOate) decay quickly. SNP and GSNO show greater stability releasing consistent lower NO levels. This comprehensive characterisation provides knowledge to define NO concentrations released in vitro.
Background Nitric oxide (NO) is a vital signalling molecule in a variety of tissues including the neuronal, vascular and reproductive system. However, its high diffusibility and inactivation make characterisation of nitrergic signalling difficult. The use of NO donors is essential to characterise downstream signalling pathways but knowledge of donor release capacities is lacking, thus making comparisons of donor responses difficult. New method This study characterises NO profiles of commonly used NO donors. Donors were stored under defined conditions and temporal release profiles detected to allow determination of released NO concentrations. Results Using NO-sensitive microsensors we assessed release profiles of NO donors following different storage times and conditions. We found that donors such as NOC-5 and PAPA-NONOate decayed substantially within days, whereas SNP and GSNO showed greater stability releasing consistent levels of NO over days. In all donors tested, the amount of released NO differs between frozen and unfrozen stocks. Comparison with existing method(s) Fluorescent and amperometric approaches to measure NO concentrations yield a wide range of levels. However, due to a lack of characterisation of the release profiles, inconsistent effects on NO signalling have been widely documented. Our systematic assessment of release profiles of a range of NO donors therefore provides new essential data allowing for improved and defined investigations of nitrergic signalling. Conclusions This is the first systematic comparison of temporal release profiles of different NO donors allowing researchers to compare conditions across different studies and the use of defined NO levels by choosing specific donors and concentrations.
Collapse
Affiliation(s)
- Sophie A Bradley
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Leicester LE1 9HN, UK
| | - Joern R Steinert
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Leicester LE1 9HN, UK.
| |
Collapse
|
35
|
Abstract
SIGNIFICANCE The pulmonary circulation is a low-pressure, low-resistance, highly compliant vasculature. In contrast to the systemic circulation, it is not primarily regulated by a central nervous control mechanism. The regulation of resting membrane potential due to ion channels is of integral importance in the physiology and pathophysiology of the pulmonary vasculature. RECENT ADVANCES Redox-driven ion conductance changes initiated by direct oxidation, nitration, and S-nitrosylation of the cysteine thiols and indirect phosphorylation of the threonine and serine residues directly affect pulmonary vascular tone. CRITICAL ISSUES Molecular mechanisms of changes in ion channel conductance, especially the identification of the sites of action, are still not fully elucidated. FUTURE DIRECTIONS Further investigation of the interaction between redox status and ion channel gating, especially the physiological significance of S-glutathionylation and S-nitrosylation, could result in a better understanding of the physiological and pathophysiological importance of these mediators in general and the implications of such modifications in cellular functions and related diseases and their importance for targeted treatment strategies.
Collapse
Affiliation(s)
- Andrea Olschewski
- 1 Ludwig Boltzmann Institute for Lung Vascular Research , Graz, Austria
| | | |
Collapse
|
36
|
Gamper N, Ooi L. Redox and nitric oxide-mediated regulation of sensory neuron ion channel function. Antioxid Redox Signal 2015; 22:486-504. [PMID: 24735331 PMCID: PMC4323017 DOI: 10.1089/ars.2014.5884] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
SIGNIFICANCE Reactive oxygen and nitrogen species (ROS and RNS, respectively) can intimately control neuronal excitability and synaptic strength by regulating the function of many ion channels. In peripheral sensory neurons, such regulation contributes towards the control of somatosensory processing; therefore, understanding the mechanisms of such regulation is necessary for the development of new therapeutic strategies and for the treatment of sensory dysfunctions, such as chronic pain. RECENT ADVANCES Tremendous progress in deciphering nitric oxide (NO) and ROS signaling in the nervous system has been made in recent decades. This includes the recognition of these molecules as important second messengers and the elucidation of their metabolic pathways and cellular targets. Mounting evidence suggests that these targets include many ion channels which can be directly or indirectly modulated by ROS and NO. However, the mechanisms specific to sensory neurons are still poorly understood. This review will therefore summarize recent findings that highlight the complex nature of the signaling pathways involved in redox/NO regulation of sensory neuron ion channels and excitability; references to redox mechanisms described in other neuron types will be made where necessary. CRITICAL ISSUES The complexity and interplay within the redox, NO, and other gasotransmitter modulation of protein function are still largely unresolved. Issues of specificity and intracellular localization of these signaling cascades will also be addressed. FUTURE DIRECTIONS Since our understanding of ROS and RNS signaling in sensory neurons is limited, there is a multitude of future directions; one of the most important issues for further study is the establishment of the exact roles that these signaling pathways play in pain processing and the translation of this understanding into new therapeutics.
Collapse
Affiliation(s)
- Nikita Gamper
- 1 Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds , Leeds, United Kingdom
| | | |
Collapse
|
37
|
|
38
|
Pottabathini R, Kumar A, Bhatnagar A, Garg S. Possible involvement of nitric oxide modulatory mechanism in the protective effect of retigabine against spinal nerve ligation-induced neuropathic pain. Cell Mol Neurobiol 2015; 35:137-46. [PMID: 25182225 PMCID: PMC11486261 DOI: 10.1007/s10571-014-0105-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 08/23/2014] [Indexed: 01/02/2023]
Abstract
Decreasing the hyperexcitability of neurons through opening of voltage-gated potassium (Kv7) channels has been suggested as one of the protective mechanisms in the effective management of neuropathic pain. Reactive oxygen/nitrogen species are well implicated in the pathophysiology of neuropathic pain. Further, M current generated by opening of voltage-gated potassium channels (Kv7) has been modulated by reactive oxygen/nitrogen species. The present study has been designed to elucidate the nitric oxide modulatory mechanism in the protective effect of retigabine against spinal nerve ligation-induced neuropathic pain in rats. Ligation of L5/L6 spinal nerves resulted in alterations in various behavioral (as evident from marked increase in thermal and mechanical hyperalgesia, and allodynia) and biochemical (raised lipid peroxidation, nitrite, and depletion of GSH, SOD, and catalase) cascades as compared to sham treatment. Administration of retigabine (10 mg/kg) for 28 days attenuated these behavioral and biochemical cascades as compared to control rats. Further, L-arginine (100 mg/kg) pretreatment with retigabine (5 mg/kg) significantly reversed the protective effect of retigabine in spinal nerve-ligated rats. However, L-NAME (10 mg/kg) pretreatment with retigabine (5 mg/kg) significantly potentiated their protective effects which were significant as compared to their effect per se, respectively. The present study highlights the possible involvement of nitric oxide modulatory mechanism in the protective effect of retigabine against L5/L6 spinal nerve ligation-induced behavioral and biochemical alterations in rats.
Collapse
Affiliation(s)
- Raghavender Pottabathini
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, India
| | - Archana Bhatnagar
- Department of Biochemistry, Basic Medical Science Building, Panjab University, Chandigarh, 160014 India
| | - Sukant Garg
- Department of Pathology, Dr. Harvansh Singh Judge Institute of Dental Sciences and Hospital, Panjab University, Chandigarh, India
| |
Collapse
|
39
|
Abstract
SIGNIFICANCE Voltage-gated K+ channels are a large family of K+-selective ion channel protein complexes that open on membrane depolarization. These K+ channels are expressed in diverse tissues and their function is vital for numerous physiological processes, in particular of neurons and muscle cells. Potentially reversible oxidative regulation of voltage-gated K+ channels by reactive species such as reactive oxygen species (ROS) represents a contributing mechanism of normal cellular plasticity and may play important roles in diverse pathologies including neurodegenerative diseases. RECENT ADVANCES Studies using various protocols of oxidative modification, site-directed mutagenesis, and structural and kinetic modeling provide a broader phenomenology and emerging mechanistic insights. CRITICAL ISSUES Physicochemical mechanisms of the functional consequences of oxidative modifications of voltage-gated K+ channels are only beginning to be revealed. In vivo documentation of oxidative modifications of specific amino-acid residues of various voltage-gated K+ channel proteins, including the target specificity issue, is largely absent. FUTURE DIRECTIONS High-resolution chemical and proteomic analysis of ion channel proteins with respect to oxidative modification combined with ongoing studies on channel structure and function will provide a better understanding of how the function of voltage-gated K+ channels is tuned by ROS and the corresponding reducing enzymes to meet cellular needs.
Collapse
Affiliation(s)
- Nirakar Sahoo
- 1 Department of Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena and Jena University Hospital , Jena, Germany
| | | | | |
Collapse
|
40
|
Optogenetic patterning of whisker-barrel cortical system in transgenic rat expressing channelrhodopsin-2. PLoS One 2014; 9:e93706. [PMID: 24695456 PMCID: PMC3973546 DOI: 10.1371/journal.pone.0093706] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 03/05/2014] [Indexed: 01/03/2023] Open
Abstract
The rodent whisker-barrel system has been an ideal model for studying somatosensory representations in the cortex. However, it remains a challenge to experimentally stimulate whiskers with a given pattern under spatiotemporal precision. Recently the optogenetic manipulation of neuronal activity has made possible the analysis of the neuronal network with precise spatiotemporal resolution. Here we identified the selective expression of channelrhodopsin-2 (ChR2), an algal light-driven cation channel, in the large mechanoreceptive neurons in the trigeminal ganglion (TG) as well as their peripheral nerve endings innervating the whisker follicles of a transgenic rat. The spatiotemporal pattern of whisker irradiation thus produced a barrel-cortical response with a specific spatiotemporal pattern as evidenced by electrophysiological and functional MRI (fMRI) studies. Our methods of generating an optogenetic tactile pattern (OTP) can be expected to facilitate studies on how the spatiotemporal pattern of touch is represented in the somatosensory cortex, as Hubel and Wiesel did in the visual cortex.
Collapse
|
41
|
Raddant AC, Russo AF. Reactive oxygen species induce procalcitonin expression in trigeminal ganglia glia. Headache 2014; 54:472-84. [PMID: 24512072 DOI: 10.1111/head.12301] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2013] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To examine calcitonin gene-related peptide (CGRP) gene expression under inflammatory conditions using trigeminal ganglia organ cultures as an experimental system. These cultures have increased proinflammatory signaling that may mimic neurogenic inflammation in the migraine state. BACKGROUND The trigeminal nerve sends peripheral pain signals to the central nervous system during migraine. Understanding the dynamic processes that occur within the trigeminal nerve and ganglion may provide insights into events that contribute to migraine pain. A neuropeptide of particular interest is CGRP, which can be elevated and play a causal role in migraine. However, most studies have overlooked a second splice product of the Calca gene that encodes calcitonin (CT), a peptide hormone involved in calcium homeostasis. Importantly, a precursor form of CT called procalcitonin (proCT) can act as a partial agonist at the CGRP receptor and elevated proCT has recently been reported during migraine. METHODS We used a trigeminal ganglion whole organ explant model, which has previously been demonstrated to induce pro-inflammatory agents in vitro. Quantitative polymerase chain reaction and immunohistochemistry were used to evaluate changes in messenger ribonucleic acid (mRNA) and protein levels of CGRP and proCT. RESULTS Whole mouse trigeminal ganglia cultured for 24 hours showed a 10-fold increase in CT mRNA, with no change in CGRP mRNA. A similar effect was observed in ganglia from adult rats. ProCT immunoreactivity was localized in glial cells. Cutting the tissue blunted the increase in CT, suggesting that induction required the close environment of the intact ganglia. Consistent with this prediction, there were increased reactive oxygen species in the ganglia, and the elevated CT mRNA was reduced by antioxidant treatment. Surprisingly, reactive oxygen species were increased in neurons, not glia. CONCLUSIONS These results demonstrate that reactive oxygen species can activate proCT expression from the CGRP gene in trigeminal glia by a paracrine regulatory mechanism. We propose that this glial recruitment pathway may occur following cortical spreading depression and neurogenic inflammation to increase CGRP nociceptive actions in migraine.
Collapse
Affiliation(s)
- Ann C Raddant
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | | |
Collapse
|
42
|
Du X, Gamper N. Potassium channels in peripheral pain pathways: expression, function and therapeutic potential. Curr Neuropharmacol 2013; 11:621-40. [PMID: 24396338 PMCID: PMC3849788 DOI: 10.2174/1570159x113119990042] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Electrical excitation of peripheral somatosensory nerves is a first step in generation of most pain signals in mammalian nervous system. Such excitation is controlled by an intricate set of ion channels that are coordinated to produce a degree of excitation that is proportional to the strength of the external stimulation. However, in many disease states this coordination is disrupted resulting in deregulated peripheral excitability which, in turn, may underpin pathological pain states (i.e. migraine, neuralgia, neuropathic and inflammatory pains). One of the major groups of ion channels that are essential for controlling neuronal excitability is potassium channel family and, hereby, the focus of this review is on the K+ channels in peripheral pain pathways. The aim of the review is threefold. First, we will discuss current evidence for the expression and functional role of various K+ channels in peripheral nociceptive fibres. Second, we will consider a hypothesis suggesting that reduced functional activity of K+ channels within peripheral nociceptive pathways is a general feature of many types of pain. Third, we will evaluate the perspectives of pharmacological enhancement of K+ channels in nociceptive pathways as a strategy for new analgesic drug design.
Collapse
Affiliation(s)
- Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
- Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|