1
|
Pryce G, Sisay S, Giovannoni G, Selwood DL, Baker D. Neuroprotection in an Experimental Model of Multiple Sclerosis via Opening of Big Conductance, Calcium-Activated Potassium Channels. Pharmaceuticals (Basel) 2023; 16:972. [PMID: 37513884 PMCID: PMC10383993 DOI: 10.3390/ph16070972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Big conductance calcium-activated (BK) channel openers can inhibit pathologically driven neural hyperactivity to control symptoms via hyperpolarizing signals to limit neural excitability. We hypothesized that BK channel openers would be neuroprotective during neuroinflammatory, autoimmune disease. The neurodegenerative disease was induced in a mouse experimental autoimmune encephalomyelitis model with translational value to detect neuroprotection in multiple sclerosis. Following the treatment with the BK channel openers, BMS-204253 and VSN16R, neuroprotection was assessed using subjective and objective clinical outcomes and by quantitating spinal nerve content. Treatment with BMS-204253 and VSN16R did not inhibit the development of relapsing autoimmunity, consistent with minimal channel expression via immune cells, nor did it change leukocyte levels in rodents or humans. However, it inhibited the accumulation of nerve loss and disability as a consequence of autoimmunity. Therefore, in addition to symptom control, BK channel openers have the potential to save nerves from excitotoxic damage and could be useful as either stand-alone neuroprotective agents or as add-ons to current disease-modifying treatments that block relapsing MS but do not have any direct neuroprotective activity.
Collapse
Affiliation(s)
- Gareth Pryce
- BartsMS, The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Sofia Sisay
- BartsMS, The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Gavin Giovannoni
- BartsMS, The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - David L Selwood
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - David Baker
- BartsMS, The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
2
|
Jiang L, Li J, Reilly S, Xin H, Guo N, Zhang X. Role of organellar Ca2+-activated K+ channels in disease development. Life Sci 2023; 316:121433. [PMID: 36708987 DOI: 10.1016/j.lfs.2023.121433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023]
Abstract
The organellar Ca2+-activated K+ channels share a similar ability to transfer the alteration of Ca2+ concentration to membrane conductance of potassium. Multiple effects of Ca2+-activated K+ channels on cell metabolism and complex signaling pathways during organ development have been explored. The organellar Ca2+-activated K+ channels are able to control the ionic equilibrium and are always associated with oxidative stress in different organelles and the whole cells. Some drugs targeting Ca2+-activated K+ channels have been tested for various diseases in clinical trials. In this review, the known roles of organellar Ca2+-activated K+ channels were described, and their effects on different diseases, particularly on diabetes, cardiovascular diseases, and neurological diseases were discussed. It was attempted to summarize the currently known operational modes with the involvement of organellar Ca2+-activated K+ channels. This review may assist scholars to more comprehensively understand organellar Ca2+-activated K+ channels and related diseases.
Collapse
Affiliation(s)
- Lan Jiang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiawei Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Nan Guo
- Department of Pharmacy, Minhang hospital, Fudan University, Shanghai, China.
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Selezneva A, Gibb AJ, Willis D. The contribution of ion channels to shaping macrophage behaviour. Front Pharmacol 2022; 13:970234. [PMID: 36160429 PMCID: PMC9490177 DOI: 10.3389/fphar.2022.970234] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022] Open
Abstract
The expanding roles of macrophages in physiological and pathophysiological mechanisms now include normal tissue homeostasis, tissue repair and regeneration, including neuronal tissue; initiation, progression, and resolution of the inflammatory response and a diverse array of anti-microbial activities. Two hallmarks of macrophage activity which appear to be fundamental to their diverse cellular functionalities are cellular plasticity and phenotypic heterogeneity. Macrophage plasticity allows these cells to take on a broad spectrum of differing cellular phenotypes in response to local and possibly previous encountered environmental signals. Cellular plasticity also contributes to tissue- and stimulus-dependent macrophage heterogeneity, which manifests itself as different macrophage phenotypes being found at different tissue locations and/or after different cell stimuli. Together, plasticity and heterogeneity align macrophage phenotypes to their required local cellular functions and prevent inappropriate activation of the cell, which could lead to pathology. To execute the appropriate function, which must be regulated at the qualitative, quantitative, spatial and temporal levels, macrophages constantly monitor intracellular and extracellular parameters to initiate and control the appropriate cell signaling cascades. The sensors and signaling mechanisms which control macrophages are the focus of a considerable amount of research. Ion channels regulate the flow of ions between cellular membranes and are critical to cell signaling mechanisms in a variety of cellular functions. It is therefore surprising that the role of ion channels in the macrophage biology has been relatively overlooked. In this review we provide a summary of ion channel research in macrophages. We begin by giving a narrative-based explanation of the membrane potential and its importance in cell biology. We then report on research implicating different ion channel families in macrophage functions. Finally, we highlight some areas of ion channel research in macrophages which need to be addressed, future possible developments in this field and therapeutic potential.
Collapse
|
4
|
Licitra R, Marchese M, Naef V, Ogi A, Martinelli M, Kiferle C, Fronte B, Santorelli FM. A Review on the Bioactivity of Cannabinoids on Zebrafish Models: Emphasis on Neurodevelopment. Biomedicines 2022; 10:biomedicines10081820. [PMID: 36009367 PMCID: PMC9404760 DOI: 10.3390/biomedicines10081820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022] Open
Abstract
For centuries, the cannabis plant has been used as a source of food, fiber, and medicine. Recently, scientific interest in cannabis has increased considerably, as its bioactive compounds have shown promising potential in the treatment of numerous musculoskeletal and neurological diseases in humans. However, the mechanisms that underlie its possible effects on neurodevelopment and nervous-system functioning remain poorly understood and need to be further investigated. Although the bulk of research on cannabis and cannabinoids is based on in vitro or rodent models, the zebrafish has now emerged as a powerful in vivo model for drug-screening studies and translational research. We here review the available literature on the use of cannabis/cannabinoids in zebrafish, and particularly in zebrafish models of neurological disorders. A critical analysis suggests that zebrafish could serve as an experimental tool for testing the bioactivity of cannabinoids, and they could thus provide important insights into the safety and efficacy of different cannabis-extract-based products. The review showed that zebrafish exhibit similar behaviors to rodents following cannabinoid exposure. The authors stress the importance of analyzing the full spectrum of naturally occurring cannabinoids, rather than just the main ones, THC and CBD, and they offer some pointers on performing behavioral analysis in zebrafish.
Collapse
Affiliation(s)
- Rosario Licitra
- Molecular Medicine and Neurobiology—ZebraLab, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy; (R.L.); (V.N.); (A.O.)
| | - Maria Marchese
- Molecular Medicine and Neurobiology—ZebraLab, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy; (R.L.); (V.N.); (A.O.)
- Correspondence: (M.M.); (F.M.S.)
| | - Valentina Naef
- Molecular Medicine and Neurobiology—ZebraLab, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy; (R.L.); (V.N.); (A.O.)
| | - Asahi Ogi
- Molecular Medicine and Neurobiology—ZebraLab, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy; (R.L.); (V.N.); (A.O.)
| | - Marco Martinelli
- PlantLab, Institute of Life Sciences, Scuola Superiore Sant’Anna, 56124 Pisa, Italy; (M.M.); (C.K.)
| | - Claudia Kiferle
- PlantLab, Institute of Life Sciences, Scuola Superiore Sant’Anna, 56124 Pisa, Italy; (M.M.); (C.K.)
| | - Baldassare Fronte
- Department of Veterinary Science, University of Pisa, 56124 Pisa, Italy;
| | - Filippo Maria Santorelli
- Molecular Medicine and Neurobiology—ZebraLab, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy; (R.L.); (V.N.); (A.O.)
- Correspondence: (M.M.); (F.M.S.)
| |
Collapse
|
5
|
Hurley MJ, Deacon RMJ, Chan AWE, Baker D, Selwood DL, Cogram P. Reversal of behavioural phenotype by the cannabinoid-like compound VSN16R in fragile X syndrome mice. Brain 2022; 145:76-82. [PMID: 34196695 PMCID: PMC8967084 DOI: 10.1093/brain/awab246] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/27/2021] [Accepted: 06/06/2021] [Indexed: 11/14/2022] Open
Abstract
Fragile X syndrome is the most common inherited intellectual disability and mono-genetic cause of autism spectrum disorder. It is a neurodevelopmental condition occurring due to a CGG trinucleotide expansion in the FMR1 gene. Polymorphisms and variants in large-conductance calcium-activated potassium channels are increasingly linked to intellectual disability and loss of FMR protein causes reduced large-conductance calcium-activated potassium channel activity leading to abnormalities in synapse function. Using the cannabinoid-like large-conductance calcium-activated potassium channel activator VSN16R we rescued behavioural deficits such as repetitive behaviour, hippocampal dependent tests of daily living, hyperactivity and memory in a mouse model of fragile X syndrome. VSN16R has been shown to be safe in a phase 1 study in healthy volunteers and in a phase 2 study in patients with multiple sclerosis with high oral bioavailability and no serious adverse effects reported. VSN16R could therefore be directly utilized in a fragile X syndrome clinical study. Moreover, VSN16R showed no evidence of tolerance, which strongly suggests that chronic VSN16R may have great therapeutic value for fragile X syndrome and autism spectrum disorder. This study provides new insight into the pathophysiology of fragile X syndrome and identifies a new pathway for drug intervention for this debilitating disorder.
Collapse
Affiliation(s)
- Michael J Hurley
- Neuroimmunology, Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO16 6YD, UK
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London NW3 2PF, UK
| | - Robert M J Deacon
- Biomedicine Division, Fraunhofer Research Institute, Center for System Biotechnology, Santiago, Chile
| | - A W Edith Chan
- Drug Discovery, UCL Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - David Baker
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - David L Selwood
- Drug Discovery, UCL Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Patricia Cogram
- Biomedicine Division, Fraunhofer Research Institute, Center for System Biotechnology, Santiago, Chile
- Department of Ecological Sciences, Institute of Ecology and Biodiversity, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| |
Collapse
|
6
|
Rupnik M, Baker D, Selwood DL. Oligodendrocytes, BK channels and remyelination. F1000Res 2021; 10:781. [PMID: 34909188 PMCID: PMC8596180 DOI: 10.12688/f1000research.53422.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 11/09/2023] Open
Abstract
Oligodendrocytes wrap multiple lamellae of their membrane, myelin, around axons of the central nervous system (CNS), to improve impulse conduction. Myelin synthesis is specialised and dynamic, responsive to local neuronal excitation. Subtle pathological insults are sufficient to cause significant neuronal metabolic impairment, so myelin preservation is necessary to safeguard neural networks. Multiple sclerosis (MS) is the most prevalent demyelinating disease of the CNS. In MS, inflammatory attacks against myelin, proposed to be autoimmune, cause myelin decay and oligodendrocyte loss, leaving neurons vulnerable. Current therapies target the prominent neuroinflammation but are mostly ineffective in protecting from neurodegeneration and the progressive neurological disability. People with MS have substantially higher levels of extracellular glutamate, the main excitatory neurotransmitter. This impairs cellular homeostasis to cause excitotoxic stress. Large conductance Ca2 +-activated K + channels (BK channels) could preserve myelin or allow its recovery by protecting cells from the resulting excessive excitability. This review evaluates the role of excitotoxic stress, myelination and BK channels in MS pathology, and explores the hypothesis that BK channel activation could be a therapeutic strategy to protect oligodendrocytes from excitotoxic stress in MS. This could reduce progression of neurological disability if used in parallel to immunomodulatory therapies.
Collapse
Affiliation(s)
- Maddalena Rupnik
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| | - David Baker
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, WC1E 6BT, UK
| | - David L. Selwood
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| |
Collapse
|
7
|
Rupnik M, Baker D, Selwood DL. Oligodendrocytes, BK channels and the preservation of myelin. F1000Res 2021; 10:781. [PMID: 34909188 PMCID: PMC8596180 DOI: 10.12688/f1000research.53422.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2021] [Indexed: 11/20/2022] Open
Abstract
Oligodendrocytes wrap multiple lamellae of their membrane, myelin, around axons of the central nervous system (CNS), to improve impulse conduction. Myelin synthesis is specialised and dynamic, responsive to local neuronal excitation. Subtle pathological insults are sufficient to cause significant neuronal metabolic impairment, so myelin preservation is necessary to safeguard neural networks. Multiple sclerosis (MS) is the most prevalent demyelinating disease of the CNS. In MS, inflammatory attacks against myelin, proposed to be autoimmune, cause myelin decay and oligodendrocyte loss, leaving neurons vulnerable. Current therapies target the prominent neuroinflammation but are mostly ineffective in protecting from neurodegeneration and the progressive neurological disability. People with MS have substantially higher levels of extracellular glutamate, the main excitatory neurotransmitter. This impairs cellular homeostasis to cause excitotoxic stress. Large conductance Ca2 +-activated K + channels (BK channels) could preserve myelin or allow its recovery by protecting cells from the resulting excessive excitability. This review evaluates the role of excitotoxic stress, myelination and BK channels in MS pathology, and explores the hypothesis that BK channel activation could be a therapeutic strategy to protect oligodendrocytes from excitotoxic stress in MS. This could reduce progression of neurological disability if used in parallel to immunomodulatory therapies.
Collapse
Affiliation(s)
- Maddalena Rupnik
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| | - David Baker
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, WC1E 6BT, UK
| | - David L. Selwood
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| |
Collapse
|
8
|
Wen JY, Zhang J, Chen S, Chen Y, Zhang Y, Ma ZY, Zhang F, Xie WM, Fan YF, Duan JS, Chen ZW. Endothelium-derived hydrogen sulfide acts as a hyperpolarizing factor and exerts neuroprotective effects via activation of large-conductance Ca 2+ -activated K + channels. Br J Pharmacol 2021; 178:4155-4175. [PMID: 34216027 DOI: 10.1111/bph.15607] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Endothelium-derived hyperpolarizing factor (EDHF) has been suggested as a therapeutic target for vascular protection against ischaemic brain injury. However, the molecular entity of EDHF and its action on neurons remains unclear. This study was undertaken to demonstrate whether the hydrogen sulfide (H2 S) acts as EDHF and exerts neuroprotective effect via large-conductance Ca2+ -activated K+ (BKCa /KCa 1.1) channels. EXPERIMENTAL APPROACH The whole-cell patch-clamp technology was used to record the changes of BKCa currents in rat neurons induced by EDHF. The cerebral ischaemia/reperfusion model of mice and oxygen-glucose deprivation/reoxygenation (OGD/R) model of neurons were used to explore the neuroprotection of EDHF by activating BKCa channels in these neurons. KEY RESULTS Increases of BKCa currents and membrane hyperpolarization in hippocampal neurons induced by EDHF could be markedly inhibited by BKCa channel inhibitor iberiotoxin or endothelial H2 S synthase inhibitor propargylglycine. The H2 S donor, NaHS-induced BKCa current and membrane hyperpolarization in neurons were also inhibited by iberiotoxin, suggesting that H2 S acts as EDHF and activates the neuronal BKCa channels. Besides, we found that the protective effect of endothelium-derived H2 S against mice cerebral ischaemia/reperfusion injury was disrupted by iberiotoxin. Importantly, the inhibitory effect of NaHS or BKCa channel opener on OGD/R-induced neuron injury and the increment of intracellular Ca2+ level could be inhibited by iberiotoxin but enhanced by co-application with L-type but not T-type calcium channel inhibitor. CONCLUSION AND IMPLICATIONS Endothelium-derived H2 S acts as EDHF and exerts neuroprotective effects via activating the BKCa channels and then inhibiting the T-type calcium channels in hippocampal neurons.
Collapse
Affiliation(s)
- Ji-Yue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jie Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Shuo Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Ye Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Zi-Yao Ma
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Fang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Wei-Ming Xie
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yi-Fei Fan
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jing-Si Duan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhi-Wu Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
9
|
Al-Karagholi MAM, Ghanizada H, Nielsen CAW, Skandarioon C, Snellman J, Lopez Lopez C, Hansen JM, Ashina M. Opening of BKCa channels alters cerebral hemodynamic and causes headache in healthy volunteers. Cephalalgia 2020; 40:1145-1154. [DOI: 10.1177/0333102420940681] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction Preclinical data implicate large conductance calcium-activated potassium (BKCa) channels in the pathogenesis of headache and migraine, but the exact role of these channels is still unknown. Here, we investigated whether opening of BKCa channels would cause headache and vascular effects in healthy volunteers. Methods In a randomized, double-blind, placebo-controlled, cross-over study, 21 healthy volunteers aged 18–39 years were randomly allocated to receive an intravenous infusion of 0.05 mg/min BKCa channel opener MaxiPost and placebo on two different days. The primary endpoints were the difference in incidence of headache and the difference in area under the curve (AUC) for headache intensity scores (0–12 hours) and for middle cerebral artery blood flow velocity (VMCA) (0–2 hours) between MaxiPost and placebo. The secondary endpoints were the differences in area under the curve for superficial temporal artery and radial artery diameter (0–2 hours) between MaxiPost and placebo. Results Twenty participants completed the study. Eighteen participants (90%) developed headache after MaxiPost compared with six (30%) after placebo ( p = 0.0005); the difference of incidence is 60% (95% confidence interval 36–84%). The area under the curve for headache intensity (AUC0–12 hours, p = 0.0003), for mean VMCA (AUC0–2 hours, p = 0.0001), for superficial temporal artery diameter (AUC0–2 hours, p = 0.003), and for radial artery diameter (AUC0–2 hours, p = 0.03) were significantly larger after MaxiPost compared to placebo. Conclusion MaxiPost caused headache and dilation in extra- and intracerebral arteries. Our findings suggest a possible role of BKCa channels in headache pathophysiology in humans. ClinicalTrials.gov, ID: NCT03887325.
Collapse
Affiliation(s)
- Mohammad Al-Mahdi Al-Karagholi
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Glostrup, Denmark
| | - Hashmat Ghanizada
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Glostrup, Denmark
| | - Cherie Amalie Waldorff Nielsen
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Glostrup, Denmark
| | - Camilla Skandarioon
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Glostrup, Denmark
| | | | | | - Jakob Møller Hansen
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Glostrup, Denmark
- Danish Headache Knowledge Center, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Messoud Ashina
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Glostrup, Denmark
- Danish Headache Knowledge Center, Rigshospitalet Glostrup, Glostrup, Denmark
| |
Collapse
|
10
|
Dyhrfjeld-Johnsen J, Cederroth CR. Current Clinical Trials for Tinnitus. Otolaryngol Clin North Am 2020; 53:651-666. [DOI: 10.1016/j.otc.2020.03.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
11
|
Bertrand JA, Schicht M, Stamer WD, Baker D, Sherwood JM, Lütjen-Drecoll E, Selwood DL, Overby DR. The β4-Subunit of the Large-Conductance Potassium Ion Channel KCa1.1 Regulates Outflow Facility in Mice. Invest Ophthalmol Vis Sci 2020; 61:41. [PMID: 32203982 PMCID: PMC7401454 DOI: 10.1167/iovs.61.3.41] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Purpose The large-conductance calcium-activated potassium channel KCa1.1 (BKCa, maxi-K) influences aqueous humor outflow facility, but the contribution of auxiliary β-subunits to KCa1.1 activity in the outflow pathway is unknown. Methods Using quantitative polymerase chain reaction, we measured expression of β-subunit genes in anterior segments of C57BL/6J mice (Kcnmb1-4) and in cultured human trabecular meshwork (TM) and Schlemm's canal (SC) cells (KCNMB1-4). We also measured expression of Kcnma1/KCNMA1 that encodes the pore-forming α-subunit. Using confocal immunofluorescence, we visualized the distribution of β4 in the conventional outflow pathway of mice. Using iPerfusion, we measured outflow facility in enucleated mouse eyes in response to 100 or 500 nM iberiotoxin (IbTX; N = 9) or 100 nM martentoxin (MarTX; N = 12). MarTX selectively blocks β4-containing KCa1.1 channels, whereas IbTX blocks KCa1.1 channels that lack β4. Results Kcnmb4 was the most highly expressed β-subunit in mouse conventional outflow tissues, expressed at a level comparable to Kcnma1. β4 was present within the juxtacanalicular TM, appearing to label cellular processes connecting to SC cells. Accordingly, KCNMB4 was the most highly expressed β-subunit in human TM cells, and the sole β-subunit in human SC cells. To dissect functional contribution, MarTX decreased outflow facility by 35% (27%, 42%; mean, 95% confidence interval) relative to vehicle-treated contralateral eyes, whereas IbTX reduced outflow facility by 16% (6%, 25%). Conclusions The β4-subunit regulates KCa1.1 activity in the conventional outflow pathway, significantly influencing outflow function. Targeting β4-containing KCa1.1 channels may be a promising approach to lower intraocular pressure to treat glaucoma.
Collapse
|
12
|
Zhu JJ, Wu SH, Chen X, Jiang TT, Li XQ, Li JM, Yan Y, Wu XJ, Liu YY, Dong P. Tanshinone IIA Suppresses Hypoxia-induced Apoptosis in Medial Vestibular Nucleus Cells Via a Skp2/BKCa Axis. Curr Pharm Des 2020; 26:4185-4194. [PMID: 32484767 DOI: 10.2174/1381612826666200602144405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/13/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND The aim of the present study was to investigate the protective effects of Tanshinone IIA (Tan IIA) on hypoxia-induced injury in the medial vestibular nucleus (MVN) cells. METHODS An in vitro hypoxia model was established using MVN cells exposed to hypoxia. The hypoxia-induced cell damage was confirmed by assessing cell viability, apoptosis and expression of apoptosis-associated proteins. Oxidative stress and related indicators were also measured following hypoxia modeling and Tan IIA treatment, and the genes potentially involved in the response were predicted using multiple GEO datasets. RESULTS The results of the present study showed that Tan IIA significantly increased cell viability, decreased cell apoptosis and decreased the ratio of Bax/Bcl-2 in hypoxia treated cells. In addition, hypoxia treatment increased oxidative stress in MVN cells, and treatment with Tan IIA reduced the oxidative stress. The expression of SPhase Kinase Associated Protein 2 (SKP2) was upregulated in hypoxia treated cells, and Tan IIA treatment reduced the expression of SKP2. Mechanistically, SKP2 interacted with large-conductance Ca2+-activated K+ channels (BKCa), regulating its expression, and BKCa knockdown alleviated the protective effects of Tan IIA on hypoxia induced cell apoptosis. CONCLUSION The results of the present study suggested that Tan IIA had a protective effect on hypoxia-induced cell damage through its anti-apoptotic and anti-oxidative activity via an SKP2/BKCa axis. These findings suggest that Tan IIA may be a potential therapeutic for the treatment of hypoxia-induced vertigo.
Collapse
Affiliation(s)
- Jing-Jing Zhu
- Department of Otolaryngology, Head and Neck Surgery, Shanghai General Hospital of Nanjing Medical University, Shanghai 200080, China
| | - Shu-Hui Wu
- Department of Otolaryngology, Baoshan Branch, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Xiang Chen
- Department of General Surgery, Baoshan Branch, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Ting-Ting Jiang
- Department of Otolaryngology, Baoshan Branch, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Xin-Qian Li
- Department of Otolaryngology, Baoshan Branch, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Jing-Min Li
- Department of Otolaryngology, Baoshan Branch, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Yong Yan
- Department of Otolaryngology, Baoshan Branch, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Xue-Jun Wu
- Department of Otolaryngology, Baoshan Branch, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Yu-Ying Liu
- Department of Otolaryngology, Head and Neck Surgery, Shanghai General Hospital of Nanjing Medical University, Shanghai 200080, China
| | - Pin Dong
- Department of Otolaryngology, Head and Neck Surgery, Shanghai General Hospital of Nanjing Medical University, Shanghai 200080, China
| |
Collapse
|
13
|
Tabatabaee S, Baker D, Selwood DL, Whalley BJ, Stephens GJ. The Cannabinoid-Like Compound, VSN16R, Acts on Large Conductance, Ca 2+-Activated K + Channels to Modulate Hippocampal CA1 Pyramidal Neuron Firing. Pharmaceuticals (Basel) 2019; 12:E104. [PMID: 31277369 PMCID: PMC6789497 DOI: 10.3390/ph12030104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 12/15/2022] Open
Abstract
Large conductance, Ca2+-activated K+ (BKCa) channels are widely expressed in the central nervous system, where they regulate action potential duration, firing frequency and consequential neurotransmitter release. Moreover, drug action on, mutations to, or changes in expression levels of BKCa can modulate neuronal hyperexcitability. Amongst other potential mechanisms of action, cannabinoid compounds have recently been reported to activate BKCa channels. Here, we examined the effects of the cannabinoid-like compound (R,Z)-3-(6-(dimethylamino)-6-oxohex-1-en-1-yl)-N-(1-hydroxypropan-2-yl) benzamide (VSN16R) at CA1 pyramidal neurons in hippocampal ex vivo brain slices using current clamp electrophysiology. We also investigated effects of the BKCa channel blockers iberiotoxin (IBTX) and the novel 7-pra-martentoxin (7-Pra-MarTx) on VSN16R action. VSN16R (100 μM) increased first and second fast after-hyperpolarization (fAHP) amplitude, decreased first and second inter spike interval (ISI) and shortened first action potential (AP) width under high frequency stimulation protocols in mouse hippocampal pyramidal neurons. IBTX (100 nM) decreased first fAHP amplitude, increased second ISI and broadened first and second AP width under high frequency stimulation protocols; IBTX also broadened first and second AP width under low frequency stimulation protocols. IBTX blocked effects of VSN16R on fAHP amplitude and ISI. 7-Pra-MarTx (100 nM) had no significant effects on fAHP amplitude and ISI but, unlike IBTX, shortened first and second AP width under high frequency stimulation protocols; 7-Pra-MarTx also shortened second AP width under low frequency stimulation protocols. However, in the presence of 7-Pra-MarTx, VSN16R retained some effects on AP waveform under high frequency stimulation protocols; moreover, VSN16R effects were revealed under low frequency stimulation protocols. These findings demonstrate that VSN16R has effects in native hippocampal neurons consistent with its causing an increase in initial firing frequency via activation of IBTX-sensitive BKCa channels. The differential pharmacological effects described suggest that VSN16R may differentially target BKCa channel subtypes.
Collapse
Affiliation(s)
| | - David Baker
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London E1 4AT, UK
| | - David L Selwood
- Department of Medicinal Chemistry, UCL Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | | | - Gary J Stephens
- Reading School of Pharmacy, University of Reading, Reading RG6 6AP, UK.
| |
Collapse
|
14
|
Gupta S, Manchanda R. A computational model of large conductance voltage and calcium activated potassium channels: implications for calcium dynamics and electrophysiology in detrusor smooth muscle cells. J Comput Neurosci 2019; 46:233-256. [PMID: 31025235 DOI: 10.1007/s10827-019-00713-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 11/25/2022]
Abstract
The large conductance voltage and calcium activated potassium (BK) channels play a crucial role in regulating the excitability of detrusor smooth muscle, which lines the wall of the urinary bladder. These channels have been widely characterized in terms of their molecular structure, pharmacology and electrophysiology. They control the repolarising and hyperpolarising phases of the action potential, thereby regulating the firing frequency and contraction profiles of the smooth muscle. Several groups have reported varied profiles of BK currents and I-V curves under similar experimental conditions. However, no single computational model has been able to reconcile these apparent discrepancies. In view of the channels' physiological importance, it is imperative to understand their mechanistic underpinnings so that a realistic model can be created. This paper presents a computational model of the BK channel, based on the Hodgkin-Huxley formalism, constructed by utilising three activation processes - membrane potential, calcium inflow from voltage-gated calcium channels on the membrane and calcium released from the ryanodine receptors present on the sarcoplasmic reticulum. In our model, we attribute the discrepant profiles to the underlying cytosolic calcium received by the channel during its activation. The model enables us to make heuristic predictions regarding the nature of the sub-membrane calcium dynamics underlying the BK channel's activation. We have employed the model to reproduce various physiological characteristics of the channel and found the simulated responses to be in accordance with the experimental findings. Additionally, we have used the model to investigate the role of this channel in electrophysiological signals, such as the action potential and spontaneous transient hyperpolarisations. Furthermore, the clinical effects of BK channel openers, mallotoxin and NS19504, were simulated for the detrusor smooth muscle cells. Our findings support the proposed application of these drugs for amelioration of the condition of overactive bladder. We thus propose a physiologically realistic BK channel model which can be integrated with other biophysical mechanisms such as ion channels, pumps and exchangers to further elucidate its micro-domain interaction with the intracellular calcium environment.
Collapse
Affiliation(s)
- Suranjana Gupta
- Computational NeuroPhysiology Lab, Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Rohit Manchanda
- Computational NeuroPhysiology Lab, Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India.
| |
Collapse
|
15
|
Abstract
Cannabinoids influence cardiovascular variables in health and disease via multiple mechanisms. The chapter covers the impact of cannabinoids on cardiovascular function in physiology and pathology and presents a critical analysis of the proposed signalling pathways governing regulation of cardiovascular function by endogenously produced and exogenous cannabinoids. We know that endocannabinoid system is overactivated under pathological conditions and plays both a protective compensatory role, such as in some forms of hypertension, atherosclerosis and other inflammatory conditions, and a pathophysiological role, such as in disease states associated with excessive hypotension. This chapter focuses on the mechanisms affecting hemodynamics and vasomotor effects of cannabinoids in health and disease states, highlighting mismatches between some studies. The chapter will first review the effects of marijuana smoking on cardiovascular system and then describe the impact of exogenous cannabinoids on cardiovascular parameters in humans and experimental animals. This will be followed by analysis of the impact of cannabinoids on reactivity of isolated vessels. The article critically reviews current knowledge on cannabinoid induction of vascular relaxation by cannabinoid receptor-dependent and -independent mechanisms and dysregulation of vascular endocannabinoid signaling in disease states.
Collapse
Affiliation(s)
- Alexander I Bondarenko
- Circulatory Physiology Department, Bogomoletz Institute of Physiology National Academy of Sciences of Ukraine, Kiev, Ukraine.
| |
Collapse
|
16
|
Arroyo González R. A review of the effects of baclofen and of THC:CBD oromucosal spray on spasticity-related walking impairment in multiple sclerosis. Expert Rev Neurother 2018; 18:785-791. [PMID: 30235965 DOI: 10.1080/14737175.2018.1510772] [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] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a complex disease with a heterogeneous and unpredictable clinical course. Mobility impairment after progressive paralyses and muscle tone spasticity is common. Areas covered: The prevalence, assessment, and pharmacological management of gait impairment and spasticity in MS and their effects on health-related quality of life (HRQoL) are discussed. The roles of oral and intrathecal baclofen and of delta-9-tetrahydrocannabinol/cannabidiol (THC:CBD) oromucosal spray in treating MS spasticity-related gait impairment are reviewed. Expert commentary: Mobility impairment and spasticity are experienced by approximately 90% and 80% of MS patients, respectively, during the disease course. Prevalence and severity of gait impairment and spasticity increase as disease progresses. The symptoms are related and both impact negatively on HRQoL. Oral baclofen and tizanidine are generally used for first-line treatment of MS spasticity but are ineffective in approximately 40% of cases. Second-line therapy includes add-on THC:CBD spray for patients with resistant MS spasticity. Results of studies evaluating baclofen for treating MS spasticity gait impairment are equivocal. In studies of patients with resistant MS spasticity, THC:CBD spray consistently improved the timed 10-meter walk test and significantly improved multiple spatial-temporal and kinematic gait parameters. THC:CBD oromucosal spray warrants further investigation as a treatment for MS spasticity-related gait impairment.
Collapse
Affiliation(s)
- Rafael Arroyo González
- a Neurology Department , Hospital Universitario Quironsalud Madrid , Pozuelo de Alarcón, Madrid , Spain
| |
Collapse
|
17
|
Bondarenko AI, Panasiuk O, Drachuk K, Montecucco F, Brandt KJ, Mach F. The quest for endothelial atypical cannabinoid receptor: BK Ca channels act as cellular sensors for cannabinoids in in vitro and in situ endothelial cells. Vascul Pharmacol 2018; 102:44-55. [PMID: 29355732 PMCID: PMC6481560 DOI: 10.1016/j.vph.2018.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/20/2022]
Abstract
Endothelium-dependent component of cannabinoid-induced vasodilation has been postulated to require G-protein-coupled non-CB1/CB2 endothelial cannabinoid (eCB) receptor. GPR18 was proposed as a candidate for eCBR. To address the hypothesis that the effects attributed to eCBR are mediated by G-protein-coupled receptor (GPCR)-independent targets, we studied the electrical responses in endothelial cells, focusing on BKCa channels. In patches excised from endothelial-derived EA.hy926 cells, N-arachidonoyl glycine (NAGly) and abnormal cannabidiol (abn-cbd), prototypical agonists for eCB receptor, stimulate single BKCa activity in a concentration- and Ca2+-dependent manner. The postulated eCB receptor inhibitors rimonabant and AM251 were found to inhibit basal and stimulated by NAGly- and abn-cbd BKCa activity in cell-free patches. In isolated mice aortas, abn-cbd and NAGly produced endothelial cell hyperpolarization that was sensitive to paxilline, a selective BKCa inhibitor, but not to GPR18 antibody, and mimicked by NS1619, a direct BKCa opener. In excised patches from mice aortic endothelium, single channel activity with characteristics similar to BKCa was established by the addition of abn-cbd and NAGly. We conclude that the two cannabinoids abn-cbd and NAGly initiate a GPR18-independent activation of BKCa channels in mice aortic endothelial cells that might contribute to vasodilation to cannabinoids.
Collapse
Affiliation(s)
- Alexander I Bondarenko
- Circulatory Physiology Department, Bogomoletz Institute of Physiology NAS of Ukraine, Bogomoletz Str 4, Kiev 01024, Ukraine; Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21/III, Graz, 8010, Austria.
| | - Olga Panasiuk
- Circulatory Physiology Department, Bogomoletz Institute of Physiology NAS of Ukraine, Bogomoletz Str 4, Kiev 01024, Ukraine
| | - Konstantin Drachuk
- Circulatory Physiology Department, Bogomoletz Institute of Physiology NAS of Ukraine, Bogomoletz Str 4, Kiev 01024, Ukraine
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, largo Benzi 10, 16132 Genoa, Italy; Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| | - Karim J Brandt
- Division of Cardiology, Foundation for Medical Researches, Department of Internal Medicine, University of Geneva, Av. de la Roseraie 64, CH 1211, Geneva 4, Switzerland
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Internal Medicine, University of Geneva, Av. de la Roseraie 64, CH 1211, Geneva 4, Switzerland
| |
Collapse
|
18
|
Baker D, Pryce G, Visintin C, Sisay S, Bondarenko AI, Vanessa Ho WS, Jackson SJ, Williams TE, Al-Izki S, Sevastou I, Okuyama M, Graier WF, Stevenson LA, Tanner C, Ross R, Pertwee RG, Henstridge CM, Irving AJ, Schulman J, Powell K, Baker MD, Giovannoni G, Selwood DL. Big conductance calcium-activated potassium channel openers control spasticity without sedation. Br J Pharmacol 2017; 174:2662-2681. [PMID: 28677901 PMCID: PMC5522996 DOI: 10.1111/bph.13889] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/27/2017] [Accepted: 05/17/2017] [Indexed: 12/19/2022] Open
Abstract
Background and Purpose Our initial aim was to generate cannabinoid agents that control spasticity, occurring as a consequence of multiple sclerosis (MS), whilst avoiding the sedative side effects associated with cannabis. VSN16R was synthesized as an anandamide (endocannabinoid) analogue in an anti‐metabolite approach to identify drugs that target spasticity. Experimental Approach Following the initial chemistry, a variety of biochemical, pharmacological and electrophysiological approaches, using isolated cells, tissue‐based assays and in vivo animal models, were used to demonstrate the activity, efficacy, pharmacokinetics and mechanism of action of VSN16R. Toxicological and safety studies were performed in animals and humans. Key Results VSN16R had nanomolar activity in tissue‐based, functional assays and dose‐dependently inhibited spasticity in a mouse experimental encephalomyelitis model of MS. This effect occurred with over 1000‐fold therapeutic window, without affecting normal muscle tone. Efficacy was achieved at plasma levels that are feasible and safe in humans. VSN16R did not bind to known CB1/CB2/GPPR55 cannabinoid‐related receptors in receptor‐based assays but acted on a vascular cannabinoid target. This was identified as the major neuronal form of the big conductance, calcium‐activated potassium (BKCa) channel. Drug‐induced opening of neuronal BKCa channels induced membrane hyperpolarization, limiting excessive neural‐excitability and controlling spasticity. Conclusions and Implications We identified the neuronal form of the BKCa channel as the target for VSN16R and demonstrated that its activation alleviates neuronal excitability and spasticity in an experimental model of MS, revealing a novel mechanism to control spasticity. VSN16R is a potential, safe and selective ligand for controlling neural hyper‐excitability in spasticity.
Collapse
Affiliation(s)
- David Baker
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Department of Neuroinflammation, UCL Institute of Neurology, University College London, London, UK
| | - Gareth Pryce
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Department of Neuroinflammation, UCL Institute of Neurology, University College London, London, UK
| | - Cristina Visintin
- Department of Neuroinflammation, UCL Institute of Neurology, University College London, London, UK.,Department of Medicinal Chemistry, UCL Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Sofia Sisay
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alexander I Bondarenko
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria.,A.A. Bogomoletz Institute of Physiology, Kiev, Ukraine
| | - W S Vanessa Ho
- Vascular Biology Research Centre. St. George's, University of London, London, UK
| | - Samuel J Jackson
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Thomas E Williams
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sarah Al-Izki
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ioanna Sevastou
- Department of Medicinal Chemistry, UCL Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Masahiro Okuyama
- Department of Medicinal Chemistry, UCL Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Wolfgang F Graier
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Lesley A Stevenson
- Vascular Biology Research Centre. St. George's, University of London, London, UK
| | - Carolyn Tanner
- Department of Biomedical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Ruth Ross
- Department of Biomedical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Roger G Pertwee
- Department of Biomedical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Christopher M Henstridge
- Neurosciences Institute, Division of Pathology and Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Andrew J Irving
- Neurosciences Institute, Division of Pathology and Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Jesse Schulman
- Canbex Therapeutics Ltd, London BioScience Innovation Centre, London, UK
| | - Keith Powell
- Canbex Therapeutics Ltd, London BioScience Innovation Centre, London, UK
| | - Mark D Baker
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gavin Giovannoni
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Department of Neuroinflammation, UCL Institute of Neurology, University College London, London, UK
| | - David L Selwood
- Department of Medicinal Chemistry, UCL Wolfson Institute for Biomedical Research, University College London, London, UK
| |
Collapse
|