101
|
Milani G, Budriesi R, Tavazzani E, Cavalluzzi MM, Mattioli LB, Miniero DV, Delre P, Belviso BD, Denegri M, Cuocci C, Rotondo NP, De Palma A, Gualdani R, Caliandro R, Mangiatordi GF, Kumawat A, Camilloni C, Priori S, Lentini G. hERG stereoselective modulation by mexiletine-derived ureas: Molecular docking study, synthesis, and biological evaluation. Arch Pharm (Weinheim) 2023; 356:e2300116. [PMID: 37460390 DOI: 10.1002/ardp.202300116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 10/06/2023]
Abstract
Long QT syndrome (LQTS) is a disorder of cardiac electrophysiology resulting in life-threatening arrhythmias; nowadays, only a few drugs are available for the management of LQTS. Focusing our attention on LQT2, one of the most common subtypes of LQTS caused by mutations in the human ether-à-go-go-related gene (hERG), in the present work, the stereoselectivity of the recently discovered mexiletine-derived urea 8 was investigated on the hERG potassium channel. According to preliminary in silico predictions, in vitro studies revealed a stereoselective behavior, with the meso form showing the greatest hERG opening activity. In addition, functional studies on guinea pig isolated left atria, aorta, and ileum demonstrated that 8 does not present any cardiac or intestinal liability in our ex vivo studies. Due to its overall profile, (R,S)-8 paves the way for the design and development of a new series of compounds potentially useful in the treatment of both congenital and drug-induced forms of LQTS.
Collapse
Affiliation(s)
- Gualtiero Milani
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Roberta Budriesi
- Department of Pharmacy and Biotechnology, Food Chemistry and Nutraceutical Lab, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | | | | | - Laura Beatrice Mattioli
- Department of Pharmacy and Biotechnology, Food Chemistry and Nutraceutical Lab, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Daniela Valeria Miniero
- Department of Biosciences, Biotechnologies, and Environment, University Aldo Moro of Bari, Bari, Italy
| | - Pietro Delre
- Chemistry Department, University of Bari Aldo Moro, Bari, Italy
- CNR-Institute of Crystallography, Bari, Italy
| | | | | | | | - Natalie Paola Rotondo
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Annalisa De Palma
- Department of Biosciences, Biotechnologies, and Environment, University Aldo Moro of Bari, Bari, Italy
| | - Roberta Gualdani
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | | | | | - Amit Kumawat
- Department of Biosciences, University of Milan, Milano, Italy
| | - Carlo Camilloni
- Department of Biosciences, University of Milan, Milano, Italy
| | - Silvia Priori
- ICS-Maugeri IRCCS, Pavia, Italy
- Molecular Cardiology, Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Giovanni Lentini
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
102
|
Alexander SPH, Mathie AA, Peters JA, Veale EL, Striessnig J, Kelly E, Armstrong JF, Faccenda E, Harding SD, Davies JA, Aldrich RW, Attali B, Baggetta AM, Becirovic E, Biel M, Bill RM, Caceres AI, Catterall WA, Conner AC, Davies P, De Clerq K, Delling M, Di Virgilio F, Falzoni S, Fenske S, Fortuny-Gomez A, Fountain S, George C, Goldstein SAN, Grimm C, Grissmer S, Ha K, Hammelmann V, Hanukoglu I, Hu M, Ijzerman AP, Jabba SV, Jarvis M, Jensen AA, Jordt SE, Kaczmarek LK, Kellenberger S, Kennedy C, King B, Kitchen P, Liu Q, Lynch JW, Meades J, Mehlfeld V, Nicke A, Offermanns S, Perez-Reyes E, Plant LD, Rash L, Ren D, Salman MM, Sieghart W, Sivilotti LG, Smart TG, Snutch TP, Tian J, Trimmer JS, Van den Eynde C, Vriens J, Wei AD, Winn BT, Wulff H, Xu H, Yang F, Fang W, Yue L, Zhang X, Zhu M. The Concise Guide to PHARMACOLOGY 2023/24: Ion channels. Br J Pharmacol 2023; 180 Suppl 2:S145-S222. [PMID: 38123150 PMCID: PMC11339754 DOI: 10.1111/bph.16178] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
The Concise Guide to PHARMACOLOGY 2023/24 is the sixth in this series of biennial publications. The Concise Guide provides concise overviews, mostly in tabular format, of the key properties of approximately 1800 drug targets, and over 6000 interactions with about 3900 ligands. There is an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (https://www.guidetopharmacology.org/), which provides more detailed views of target and ligand properties. Although the Concise Guide constitutes almost 500 pages, the material presented is substantially reduced compared to information and links presented on the website. It provides a permanent, citable, point-in-time record that will survive database updates. The full contents of this section can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.16178. Ion channels are one of the six major pharmacological targets into which the Guide is divided, with the others being: G protein-coupled receptors, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to mid-2023, and supersedes data presented in the 2021/22, 2019/20, 2017/18, 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the Nomenclature and Standards Committee of the International Union of Basic and Clinical Pharmacology (NC-IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human drug targets, where appropriate.
Collapse
Affiliation(s)
- Stephen P H Alexander
- School of Life Sciences, University of Nottingham Medical School, Nottingham, NG7 2UH, UK
| | - Alistair A Mathie
- School of Engineering, Arts, Science and Technology, University of Suffolk, Ipswich, IP4 1QJ, UK
| | - John A Peters
- Neurosci-ence Division, Medical Education Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Emma L Veale
- Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Anson Building, Central Avenue, Chatham Maritime, Chatham, Kent, ME4 4TB, UK
| | - Jörg Striessnig
- Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, A-6020, Innsbruck, Austria
| | - Eamonn Kelly
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Jane F Armstrong
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Elena Faccenda
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Simon D Harding
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Jamie A Davies
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | | | | | | | | | - Martin Biel
- Ludwig Maximilian University of Munich, Munich, Germany
| | | | | | | | | | - Paul Davies
- Tufts University School of Medicine, Boston, USA
| | | | - Markus Delling
- University of California San Francisco, San Francisco, USA
| | | | | | | | | | | | - Chandy George
- Nanyang Technological University, Singapore, Singapore
| | | | | | | | - Kotdaji Ha
- University of California San Francisco, San Francisco, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Annette Nicke
- Ludwig Maximilian University of Munich, Munich, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research/JW Goethe University, Bad Nauheim/Frankfurt, Germany
| | | | | | | | - Dejian Ren
- University of Pennsylvania, Philadelphia, USA
| | | | | | | | | | | | - Jinbin Tian
- University of Texas at Houston, Houston, USA
| | | | | | | | | | | | | | | | | | | | - Lixia Yue
- University of Connecticut, Farmington, USA
| | | | - Michael Zhu
- University of Texas at Houston, Houston, USA
| |
Collapse
|
103
|
Maria SA, Kumar A, Wilfred PM, Shanthi M, Peedicayil J. Inhibition of Contractility of Isolated Caprine Detrusor by the Calcium Channel Blocker Cilnidipine and Reversal by Calcium Channel Openers. CURRENT THERAPEUTIC RESEARCH 2023; 99:100717. [PMID: 37869401 PMCID: PMC10589763 DOI: 10.1016/j.curtheres.2023.100717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/26/2023] [Indexed: 10/24/2023]
Abstract
Background Cilnidipine is a fourth-generation calcium channel blocker that is clinically used to treat hypertension. It is a dihydropyridine that blocks L- and N-type calcium channels. The inhibitory effect of cilnidipine on isolated detrusor muscle contractility has not been studied. This study investigated the inhibitory effect of cilnidipine on isolated caprine (goat) detrusor muscle contractility and the reversal of the inhibition by calcium channel openers. Methods Fourteen caprine detrusor strips were made to contract using 80 mM potassium chloride before and after addition of three concentrations (20, 40, and 60 µM) of cilnidipine. Two reversal agents, the L-type calcium channel opener FPL64716, and the N-type calcium channel opener GV-58, were investigated for their ability to reverse the inhibitory effect of 40 µΜ cilnidipine on potassium chloride-induced detrusor contractility. Results Cilnidipine caused a dose-dependent and statistically significant inhibition of detrusor contractility at all concentrations of cilnidipine used (20, 40, and 60 µΜ). The inhibitory effect of 40 µM cilnidipine on detrusor contractility was significantly reversed by the addition of FPL64716 and GV-58. Conclusions Cilnidipine inhibits the contractility of the isolated detrusor by blocking L- and N-type calcium channels. Cilnidipine could be evaluated for treating clinical conditions requiring relaxation of the detrusor such as overactive bladder.
Collapse
Affiliation(s)
- Steffi A. Maria
- Department of Pharmacology & Clinical Pharmacology, Christian Medical College, Vellore, India
| | - Aniket Kumar
- Department of Pharmacology & Clinical Pharmacology, Christian Medical College, Vellore, India
| | - Premila M. Wilfred
- Department of Pharmacology & Clinical Pharmacology, Christian Medical College, Vellore, India
| | - Margaret Shanthi
- Department of Pharmacology & Clinical Pharmacology, Christian Medical College, Vellore, India
| | - Jacob Peedicayil
- Department of Pharmacology & Clinical Pharmacology, Christian Medical College, Vellore, India
| |
Collapse
|
104
|
Del Rivero Morfin PJ, Chavez DS, Jayaraman S, Yang L, Kochiss AL, Colecraft HM, Liu XS, Marx SO, Rajadhyaksha AM, Ben-Johny M. A Genetically Encoded Actuator Selectively Boosts L-type Calcium Channels in Diverse Physiological Settings. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.558856. [PMID: 37790372 PMCID: PMC10542531 DOI: 10.1101/2023.09.22.558856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
L-type Ca 2+ channels (Ca V 1.2/1.3) convey influx of calcium ions (Ca 2+ ) that orchestrate a bevy of biological responses including muscle contraction and gene transcription. Deficits in Ca V 1 function play a vital role in cardiac and neurodevelopmental disorders. Yet conventional pharmacological approaches to upregulate Ca V 1 are limited, as excessive Ca 2+ influx leads to cytotoxicity. Here, we develop a genetically encoded enhancer of Ca V 1.2/1.3 channels (GeeC) to manipulate Ca 2+ entry in distinct physiological settings. Specifically, we functionalized a nanobody that targets the Ca V macromolecular complex by attaching a minimal effector domain from a Ca V enhancer-leucine rich repeat containing protein 10 (Lrrc10). In cardiomyocytes, GeeC evoked a 3-fold increase in L-type current amplitude. In neurons, GeeC augmented excitation-transcription (E-T) coupling. In all, GeeC represents a powerful strategy to boost Ca V 1.2/1.3 function in distinct physiological settings and, in so doing, lays the groundwork to illuminate new insights on neuronal and cardiac physiology and disease.
Collapse
|
105
|
Smith PA. Neuropathic pain; what we know and what we should do about it. FRONTIERS IN PAIN RESEARCH 2023; 4:1220034. [PMID: 37810432 PMCID: PMC10559888 DOI: 10.3389/fpain.2023.1220034] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Neuropathic pain can result from injury to, or disease of the nervous system. It is notoriously difficult to treat. Peripheral nerve injury promotes Schwann cell activation and invasion of immunocompetent cells into the site of injury, spinal cord and higher sensory structures such as thalamus and cingulate and sensory cortices. Various cytokines, chemokines, growth factors, monoamines and neuropeptides effect two-way signalling between neurons, glia and immune cells. This promotes sustained hyperexcitability and spontaneous activity in primary afferents that is crucial for onset and persistence of pain as well as misprocessing of sensory information in the spinal cord and supraspinal structures. Much of the current understanding of pain aetiology and identification of drug targets derives from studies of the consequences of peripheral nerve injury in rodent models. Although a vast amount of information has been forthcoming, the translation of this information into the clinical arena has been minimal. Few, if any, major therapeutic approaches have appeared since the mid 1990's. This may reflect failure to recognise differences in pain processing in males vs. females, differences in cellular responses to different types of injury and differences in pain processing in humans vs. animals. Basic science and clinical approaches which seek to bridge this knowledge gap include better assessment of pain in animal models, use of pain models which better emulate human disease, and stratification of human pain phenotypes according to quantitative assessment of signs and symptoms of disease. This can lead to more personalized and effective treatments for individual patients. Significance statement: There is an urgent need to find new treatments for neuropathic pain. Although classical animal models have revealed essential features of pain aetiology such as peripheral and central sensitization and some of the molecular and cellular mechanisms involved, they do not adequately model the multiplicity of disease states or injuries that may bring forth neuropathic pain in the clinic. This review seeks to integrate information from the multiplicity of disciplines that seek to understand neuropathic pain; including immunology, cell biology, electrophysiology and biophysics, anatomy, cell biology, neurology, molecular biology, pharmacology and behavioral science. Beyond this, it underlines ongoing refinements in basic science and clinical practice that will engender improved approaches to pain management.
Collapse
Affiliation(s)
- Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
106
|
Borges FS, Protachevicz PR, Souza DLM, Bittencourt CF, Gabrick EC, Bentivoglio LE, Szezech JD, Batista AM, Caldas IL, Dura-Bernal S, Pena RFO. The Roles of Potassium and Calcium Currents in the Bistable Firing Transition. Brain Sci 2023; 13:1347. [PMID: 37759949 PMCID: PMC10527161 DOI: 10.3390/brainsci13091347] [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: 08/14/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Healthy brains display a wide range of firing patterns, from synchronized oscillations during slow-wave sleep to desynchronized firing during movement. These physiological activities coexist with periods of pathological hyperactivity in the epileptic brain, where neurons can fire in synchronized bursts. Most cortical neurons are pyramidal regular spiking (RS) cells with frequency adaptation and do not exhibit bursts in current-clamp experiments (in vitro). In this work, we investigate the transition mechanism of spike-to-burst patterns due to slow potassium and calcium currents, considering a conductance-based model of a cortical RS cell. The joint influence of potassium and calcium ion channels on high synchronous patterns is investigated for different synaptic couplings (gsyn) and external current inputs (I). Our results suggest that slow potassium currents play an important role in the emergence of high-synchronous activities, as well as in the spike-to-burst firing pattern transitions. This transition is related to the bistable dynamics of the neuronal network, where physiological asynchronous states coexist with pathological burst synchronization. The hysteresis curve of the coefficient of variation of the inter-spike interval demonstrates that a burst can be initiated by firing states with neuronal synchronization. Furthermore, we notice that high-threshold (IL) and low-threshold (IT) ion channels play a role in increasing and decreasing the parameter conditions (gsyn and I) in which bistable dynamics occur, respectively. For high values of IL conductance, a synchronous burst appears when neurons are weakly coupled and receive more external input. On the other hand, when the conductance IT increases, higher coupling and lower I are necessary to produce burst synchronization. In light of our results, we suggest that channel subtype-specific pharmacological interactions can be useful to induce transitions from pathological high bursting states to healthy states.
Collapse
Affiliation(s)
- Fernando S. Borges
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Center for Mathematics, Computation and Cognition, Federal University of ABC, São Bernardo do Campo 09606-045, Brazil
| | | | - Diogo L. M. Souza
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
| | - Conrado F. Bittencourt
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
| | - Enrique C. Gabrick
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
| | - Lucas E. Bentivoglio
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
| | - José D. Szezech
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
- Department of Mathematics and Statistics, State University of Ponta Grossa, Ponta Grossa 84030-900, Brazil
| | - Antonio M. Batista
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
- Department of Mathematics and Statistics, State University of Ponta Grossa, Ponta Grossa 84030-900, Brazil
| | - Iberê L. Caldas
- Institute of Physics, University of São Paulo, São Paulo 05508-090, Brazil
| | - Salvador Dura-Bernal
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Center for Biomedical Imaging and Neuromodulation, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Rodrigo F. O. Pena
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL 33458, USA
| |
Collapse
|
107
|
Arias HR, Tae HS, Micheli L, Yousuf A, Manetti D, Romanelli MN, Ghelardini C, Adams DJ, Di Cesare Mannelli L. The Antinociceptive Activity of (E)-3-(thiophen-2-yl)- N -(p-tolyl)acrylamide in Mice Is Reduced by (E)-3-(furan-2-yl)- N -methyl- N -(p-tolyl)acrylamide Through Opposing Modulatory Mechanisms at the α7 Nicotinic Acetylcholine Receptor. Anesth Analg 2023; 137:691-701. [PMID: 37058425 PMCID: PMC10408732 DOI: 10.1213/ane.0000000000006461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND The primary objective of this study was to characterize the pharmacological and behavioral activity of 2 novel compounds, DM497 [(E)-3-(thiophen-2-yl)- N -(p-tolyl)acrylamide] and DM490 [(E)-3-(furan-2-yl)- N -methyl- N -(p-tolyl)acrylamide], structural derivatives of PAM-2, a positive allosteric modulator of the α7 nicotinic acetylcholine receptor (nAChR). METHODS A mouse model of oxaliplatin-induced neuropathic pain (2.4 mg/kg, 10 injections) was used to test the pain-relieving properties of DM497 and DM490. To assess possible mechanisms of action, the activity of these compounds was determined at heterologously expressed α7 and α9α10 nAChRs, and voltage-gated N-type calcium channel (Ca V 2.2) using electrophysiological techniques. RESULTS Cold plate tests indicated that 10 mg/kg DM497 was able to decrease neuropathic pain in mice induced by the chemotherapeutic agent oxaliplatin. In contrast, DM490 induced neither pro- nor antinociceptive activity but inhibited DM497's effect at equivalent dose (30 mg/kg). These effects are not a product of changes in motor coordination or locomotor activity. At α7 nAChRs, DM497 potentiated whereas DM490 inhibited its activity. In addition, DM490 antagonized the α9α10 nAChR with >8-fold higher potency than that for DM497. In contrast, DM497 and DM490 had minimal inhibitory activity at the Ca V 2.2 channel. Considering that DM497 did not increase the mouse exploratory activity, an indirect anxiolytic mechanism was not responsible for the observed antineuropathic effect. CONCLUSIONS The antinociceptive activity of DM497 and the concomitant inhibitory effect of DM490 are mediated by opposing modulatory mechanisms on the α7 nAChR, whereas the involvement of other possible nociception targets such as the α9α10 nAChR and Ca V 2.2 channel can be ruled out.
Collapse
Affiliation(s)
- Hugo R. Arias
- From the Department of Pharmacology and Physiology, College of Osteopathic Medicine, Oklahoma State University Center for Health Sciences, Tahlequah, Oklahoma
| | - Han-Shen Tae
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales, Australiaand Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA)
| | | | - Arsalan Yousuf
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales, Australiaand Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA)
| | - Dina Manetti
- Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Florence, Italy
| | - Maria Novella Romanelli
- Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Florence, Italy
| | | | - David J. Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales, Australiaand Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA)
| | | |
Collapse
|
108
|
Zhang Y, Wei Y, Zheng T, Tao Y, Sun Y, Jiang D, Tao J. Adiponectin receptor 1-mediated stimulation of Cav3.2 channels in trigeminal ganglion neurons induces nociceptive behaviors in mice. J Headache Pain 2023; 24:117. [PMID: 37620777 PMCID: PMC10463856 DOI: 10.1186/s10194-023-01658-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Adipokines, including adiponectin, are implicated in nociceptive pain; however, the underlying cellular and molecular mechanisms remain unknown. METHODS Using electrophysiological recording, immunostaining, molecular biological approaches and animal behaviour tests, we elucidated a pivotal role of adiponectin in regulating membrane excitability and pain sensitivity by manipulating Cav3.2 channels in trigeminal ganglion (TG) neurons. RESULTS Adiponectin enhanced T-type Ca2+ channel currents (IT) in TG neurons through the activation of adiponectin receptor 1 (adipoR1) but independently of heterotrimeric G protein-mediated signaling. Coimmunoprecipitation revealed a physical association between AdipoR1 and casein kinase II alpha-subunits (CK2α) in the TG, and inhibiting CK2 activity by chemical inhibitor or siRNA targeting CK2α prevented the adiponectin-induced IT response. Adiponectin significantly activated protein kinase C (PKC), and this effect was abrogated by CK2α knockdown. Adiponectin increased the membrane abundance of PKC beta1 (PKCβ1). Blocking PKCβ1 pharmacologically or genetically abrogated the adiponectin-induced IT increase. In heterologous expression systems, activation of adipoR1 induced a selective enhancement of Cav3.2 channel currents, dependent on PKCβ1 signaling. Functionally, adiponectin increased TG neuronal excitability and induced mechanical pain hypersensitivity, both attenuated by T-type channel blockade. In a trigeminal neuralgia model induced by chronic constriction injury of infraorbital nerve, blockade of adipoR1 signaling suppressed mechanical allodynia, which was prevented by silencing Cav3.2. CONCLUSION Our study elucidates a novel signaling cascade wherein adiponectin stimulates TG Cav3.2 channels via adipoR1 coupled to a novel CK2α-dependent PKCβ1. This process induces neuronal hyperexcitability and pain hypersensitivity. Insight into adipoR-Cav3.2 signaling in sensory neurons provides attractive targets for pain treatment.
Collapse
Affiliation(s)
- Yuan Zhang
- Clinical Research Center of Neurological Disease & Department of Geriatrics, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004 People’s Republic of China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123 People’s Republic of China
| | - Yuan Wei
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123 People’s Republic of China
| | - Tingting Zheng
- Clinical Research Center of Neurological Disease & Department of Geriatrics, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004 People’s Republic of China
| | - Yu Tao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123 People’s Republic of China
| | - Yufang Sun
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123 People’s Republic of China
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123 People’s Republic of China
| | - Dongsheng Jiang
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, 81377 Munich, Germany
| | - Jin Tao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123 People’s Republic of China
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123 People’s Republic of China
| |
Collapse
|
109
|
Borges FS, Protachevicz PR, Souza DLM, Bittencourt CF, Gabrick EC, Bentivoglio LE, Szezech JD, Batista AM, Caldas IL, Dura-Bernal S, Pena RFO. The Role of Potassium and Calcium Currents in the Bistable Firing Transition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553625. [PMID: 37645875 PMCID: PMC10462112 DOI: 10.1101/2023.08.16.553625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Healthy brains display a wide range of firing patterns, from synchronized oscillations during slowwave sleep to desynchronized firing during movement. These physiological activities coexist with periods of pathological hyperactivity in the epileptic brain, where neurons can fire in synchronized bursts. Most cortical neurons are pyramidal regular spiking cells (RS) with frequency adaptation and do not exhibit bursts in current-clamp experiments ( in vitro ). In this work, we investigate the transition mechanism of spike-to-burst patterns due to slow potassium and calcium currents, considering a conductance-based model of a cortical RS cell. The joint influence of potassium and calcium ion channels on high synchronous patterns is investigated for different synaptic couplings ( g syn ) and external current inputs ( I ). Our results suggest that slow potassium currents play an important role in the emergence of high-synchronous activities, as well as in the spike-to-burst firing pattern transitions. This transition is related to bistable dynamics of the neuronal network, where physiological asynchronous states coexist with pathological burst synchronization. The hysteresis curve of the coefficient of variation of the inter-spike interval demonstrates that a burst can be initiated by firing states with neuronal synchronization. Furthermore, we notice that high-threshold ( I L ) and low-threshold ( I T ) ion channels play a role in increasing and decreasing the parameter conditions ( g syn and I ) in which bistable dynamics occur, respectively. For high values of I L conductance, a synchronous burst appears when neurons are weakly coupled and receive more external input. On the other hand, when the conductance I T increases, higher coupling and lower I are necessary to produce burst synchronization. In light of our results, we suggest that channel subtype-specific pharmacological interactions can be useful to induce transitions from pathological high bursting states to healthy states.
Collapse
Affiliation(s)
- Fernando S Borges
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
- Center for Mathematics, Computation, and Cognition, Federal University of ABC, 09606-045 São Bernardo do Campo, SP, Brazil
| | | | - Diogo L M Souza
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - Conrado F Bittencourt
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - Enrique C Gabrick
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - Lucas E Bentivoglio
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - José D Szezech
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
- Department of Mathematics and Statistics, State University of Ponta Grossa, Ponta Grossa, Brazil
| | - Antonio M Batista
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
- Department of Mathematics and Statistics, State University of Ponta Grossa, Ponta Grossa, Brazil
| | - Iberê L Caldas
- Institute of Physics, University of São Paulo, 05508-090 São Paulo, SP, Brazil
| | - Salvador Dura-Bernal
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
- Center for Biomedical Imaging and Neuromodulation, The Nathan S. Kline Institute for Psychiatric Research, New York, USA
| | - Rodrigo F O Pena
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida, USA
| |
Collapse
|
110
|
Zhong L, Cao X, Li L, He Y, Liu Y, Chen W, Yang F, Xiao N, Zhang J, He H. Renzhu Ointment Regulates L-Type Voltage-Dependent Calcium Channel in Mice Model of Senna-Induced Diarrhea by Transdermal Administration. Drug Des Devel Ther 2023; 17:2355-2368. [PMID: 37588013 PMCID: PMC10426434 DOI: 10.2147/dddt.s419626] [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] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/21/2023] [Indexed: 08/18/2023] Open
Abstract
Purpose In China, herbal preparation is commonly administered transdermally for treating pediatric diarrhea. However, few studies have probed into their antidiarrheal mechanisms. This study was designed to investigate the antidiarrheal effect of Renzhu ointment (Renzhuqigao, RZQG) and its underlying mechanisms via transdermal administration. Methods The main components of RZQG were confirmed by gas chromatography-mass spectrometry (GC-MS). The effect of RZQG on L-type voltage-dependent calcium channel (L-VDCC) was evaluated by CaCl2- and ACh-induced contraction in isolated colon. The antidiarrheal efficacy of RZQG was further investigated by the senna-induced diarrhea mice based on the frequency of loose stools, diarrhea rate and index, fecal moisture content, and the basal tension of the colon. Additionally, the protein expression of CACNA1C, CACNA1D, cAMP, and PKA were detected with Western blot and immunohistochemistry (IHC). Results GC-MS analysis determined 14 components in RZQG. In vitro, RZQG relaxed the CaCl2- and ACh-induced tension, while nifedipine (a L-VDCC inhibitor) and H-89 (a PKA inhibitor) decreased the relaxation. In vivo, animal model showed that transdermal administration of RZQG exhibited a significant reduction in the frequency of loose stools, diarrhea rate and index, fecal moisture content and the basal tension. Compared to the model group, the colon of mice treated with RZQG showed lower expression of CACNA1C, CACNA1D, cAMP, and PKA. IHC results showed that cAMP was downregulated in colonic smooth muscle after RZQG treatment. Conclusion RZQG improved diarrhea symptoms and down-regulated the expression of CACNA1C and CACNA1D via transdermal administration, which is closely associated with the cAMP/PKA signaling pathway in colonic smooth muscle.
Collapse
Affiliation(s)
- Lian Zhong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Xiaoyu Cao
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China
| | - Li Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Yuanyuan He
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Yanxia Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Weiwei Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Fuzhen Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Ni Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Jun Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Huifen He
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People’s Republic of China
| |
Collapse
|
111
|
Ni K, Liu H, Lai K, Shen L, Li X, Wang J, Shi H. Upregulation of A-type potassium channels suppresses neuronal excitability in hypoxic neonatal mice. FEBS J 2023; 290:4092-4106. [PMID: 37059697 DOI: 10.1111/febs.16799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/22/2023] [Accepted: 04/13/2023] [Indexed: 04/16/2023]
Abstract
Neuronal excitability is a critical feature of central nervous system development, playing a fundamental role in the functional maturation of brain regions, including the hippocampus, cerebellum, auditory and visual systems. The present study aimed to determine the mechanism by which hypoxia causes brain dysfunction through perturbation of neuronal excitability in a hypoxic neonatal mouse model. Functional brain development was assessed in humans using the Gesell Development Diagnosis Scale. In mice, gene transcription was evaluated via mRNA sequencing and quantitative PCR; furthermore, patch clamp recordings assessed potassium currents. Clinical observations revealed disrupted functional brain development in 6- and 18-month-old hypoxic neonates, and those born with normal hearing screening unexpectedly exhibited impaired central auditory function at 3 months. In model mice, CA1 pyramidal neurons exhibited reduced spontaneous activity, largely induced by excitatory synaptic input suppression, despite the elevated membrane excitability of hypoxic neurons compared to that of control neurons. In hypoxic neurons, Kcnd3 gene transcription was upregulated, confirming upregulated hippocampal Kv 4.3 expression. A-type potassium currents were enhanced, and Kv 4.3 participated in blocking excitatory presynaptic inputs. Elevated Kv 4.3 activity in pyramidal neurons under hypoxic conditions inhibited excitatory presynaptic inputs and further decreased neuronal excitability, disrupting functional brain development in hypoxic neonates.
Collapse
Affiliation(s)
- Kun Ni
- Department of Otorhinolaryngology-Head & Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Otorhinolaryngology-Head & Neck Surgery, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hanwei Liu
- Department of Otorhinolaryngology-Head & Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ke Lai
- Department of Otorhinolaryngology-Head & Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Shen
- Department of Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Li
- Department of Otorhinolaryngology-Head & Neck Surgery, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiping Wang
- Department of Otorhinolaryngology-Head & Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibo Shi
- Department of Otorhinolaryngology-Head & Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
112
|
Topczewska A, Giacalone E, Pratt WS, Migliore M, Dolphin AC, Shah MM. T-type Ca 2+ and persistent Na + currents synergistically elevate ventral, not dorsal, entorhinal cortical stellate cell excitability. Cell Rep 2023; 42:112699. [PMID: 37368752 PMCID: PMC10687207 DOI: 10.1016/j.celrep.2023.112699] [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: 03/24/2022] [Revised: 03/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Dorsal and ventral medial entorhinal cortex (mEC) regions have distinct neural network firing patterns to differentially support functions such as spatial memory. Accordingly, mEC layer II dorsal stellate neurons are less excitable than ventral neurons. This is partly because the densities of inhibitory conductances are higher in dorsal than ventral neurons. Here, we report that T-type Ca2+ currents increase 3-fold along the dorsal-ventral axis in mEC layer II stellate neurons, with twice as much CaV3.2 mRNA in ventral mEC compared with dorsal mEC. Long depolarizing stimuli trigger T-type Ca2+ currents, which interact with persistent Na+ currents to elevate the membrane voltage and spike firing in ventral, not dorsal, neurons. T-type Ca2+ currents themselves prolong excitatory postsynaptic potentials (EPSPs) to enhance their summation and spike coupling in ventral neurons only. These findings indicate that T-type Ca2+ currents critically influence the dorsal-ventral mEC stellate neuron excitability gradient and, thereby, mEC dorsal-ventral circuit activity.
Collapse
Affiliation(s)
| | | | - Wendy S Pratt
- Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Michele Migliore
- Institute of Biophysics, National Research Council, 90146 Palermo, Italy
| | - Annette C Dolphin
- Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Mala M Shah
- Pharmacology, School of Pharmacy, University College London, London WC1N 4AX, UK.
| |
Collapse
|
113
|
Rangel-Galván M, Rangel-Galván V, Rangel-Huerta A. T-type calcium channel modulation by hydrogen sulfide in neuropathic pain conditions. Front Pharmacol 2023; 14:1212800. [PMID: 37529702 PMCID: PMC10387653 DOI: 10.3389/fphar.2023.1212800] [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: 04/26/2023] [Accepted: 07/05/2023] [Indexed: 08/03/2023] Open
Abstract
Neuropathic pain can appear as a direct or indirect nerve damage lesion or disease that affects the somatosensory nervous system. If the neurons are damaged or indirectly stimulated, immune cells contribute significantly to inflammatory and neuropathic pain. After nerve injury, peripheral macrophages/spinal microglia accumulate around damaged neurons, producing endogenous hydrogen sulfide (H2S) through the cystathionine-γ-lyase (CSE) enzyme. H2S has a pronociceptive modulation on the Cav3.2 subtype, the predominant Cav3 isoform involved in pain processes. The present review provides relevant information about H2S modulation on the Cav3.2 T-type channels in neuropathic pain conditions. We have discussed that the dual effect of H2S on T-type channels is concentration-dependent, that is, an inhibitory effect is seen at low concentrations of 10 µM and an augmentation effect on T-current at 100 µM. The modulation mechanism of the Cav3.2 channel by H2S involves the direct participation of the redox/Zn2+ affinity site located in the His191 in the extracellular loop of domain I of the channel, involving a group of extracellular cysteines, comprising C114, C123, C128, and C1333, that can modify the local redox environment. The indirect interaction pathways involve the regulation of the Cav3.2 channel through cytokines, kinases, and post-translational regulators of channel expression. The findings conclude that the CSE/H2S/Cav3.2 pathway could be a promising therapeutic target for neuropathic pain disorders.
Collapse
Affiliation(s)
- Maricruz Rangel-Galván
- Biothecnology Department, Metropolitan Polytechnic University of Puebla, Puebla, Puebla, Mexico
| | - Violeta Rangel-Galván
- Nursing and Physiotherapy Department, University of Professional Development, Tijuana, Baja California, Mexico
| | - Alejandro Rangel-Huerta
- Faculty of Computer Science, Meritorious Autonomous University of Puebla, Puebla, Puebla, Mexico
| |
Collapse
|
114
|
Li P, Qin D, Chen T, Hou W, Song X, Yin S, Song M, Fernando WCHA, Chen X, Sun Y, Wang J. Dysregulated Rbfox2 produces aberrant splicing of Ca V1.2 calcium channel in diabetes-induced cardiac hypertrophy. Cardiovasc Diabetol 2023; 22:168. [PMID: 37415128 PMCID: PMC10324275 DOI: 10.1186/s12933-023-01894-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND L-type Ca2+ channel CaV1.2 is essential for cardiomyocyte excitation, contraction and gene transcription in the heart, and abnormal functions of cardiac CaV1.2 channels are presented in diabetic cardiomyopathy. However, the underlying mechanisms are largely unclear. The functions of CaV1.2 channels are subtly modulated by splicing factor-mediated alternative splicing (AS), but whether and how CaV1.2 channels are alternatively spliced in diabetic heart remains unknown. METHODS Diabetic rat models were established by using high-fat diet in combination with low dose streptozotocin. Cardiac function and morphology were assessed by echocardiography and HE staining, respectively. Isolated neonatal rat ventricular myocytes (NRVMs) were used as a cell-based model. Cardiac CaV1.2 channel functions were measured by whole-cell patch clamp, and intracellular Ca2+ concentration was monitored by using Fluo-4 AM. RESULTS We find that diabetic rats develop diastolic dysfunction and cardiac hypertrophy accompanied by an increased CaV1.2 channel with alternative exon 9* (CaV1.2E9*), but unchanged that with alternative exon 8/8a or exon 33. The splicing factor Rbfox2 expression is also increased in diabetic heart, presumably because of dominate-negative (DN) isoform. Unexpectedly, high glucose cannot induce the aberrant expressions of CaV1.2 exon 9* and Rbfox2. But glycated serum (GS), the mimic of advanced glycation end-products (AGEs), upregulates CaV1.2E9* channels proportion and downregulates Rbfox2 expression in NRVMs. By whole-cell patch clamp, we find GS application hyperpolarizes the current-voltage curve and window currents of cardiac CaV1.2 channels. Moreover, GS treatment raises K+-triggered intracellular Ca2+ concentration ([Ca2+]i), enlarges cell surface area of NRVMs and induces hypertrophic genes transcription. Consistently, siRNA-mediated knockdown of Rbfox2 in NRVMs upregulates CaV1.2E9* channel, shifts CaV1.2 window currents to hyperpolarization, increases [Ca2+]i and induces cardiomyocyte hypertrophy. CONCLUSIONS AGEs, not glucose, dysregulates Rbfox2 which thereby increases CaV1.2E9* channels and hyperpolarizes channel window currents. These make the channels open at greater negative potentials and lead to increased [Ca2+]i in cardiomyocytes, and finally induce cardiomyocyte hypertrophy in diabetes. Our work elucidates the underlying mechanisms for CaV1.2 channel regulation in diabetic heart, and targeting Rbfox2 to reset the aberrantly spliced CaV1.2 channel might be a promising therapeutic approach in diabetes-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Pengpeng Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Dongxia Qin
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Tiange Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Wei Hou
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xinyu Song
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Shumin Yin
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Miaomiao Song
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - W C Hewith A Fernando
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiaojie Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yu Sun
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Juejin Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
115
|
Chen Z, Mondal A, Abderemane-Ali F, Jang S, Niranjan S, Montaño JL, Zaro BW, Minor DL. EMC chaperone-Ca V structure reveals an ion channel assembly intermediate. Nature 2023; 619:410-419. [PMID: 37196677 PMCID: PMC10896479 DOI: 10.1038/s41586-023-06175-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 05/05/2023] [Indexed: 05/19/2023]
Abstract
Voltage-gated ion channels (VGICs) comprise multiple structural units, the assembly of which is required for function1,2. Structural understanding of how VGIC subunits assemble and whether chaperone proteins are required is lacking. High-voltage-activated calcium channels (CaVs)3,4 are paradigmatic multisubunit VGICs whose function and trafficking are powerfully shaped by interactions between pore-forming CaV1 or CaV2 CaVα1 (ref. 3), and the auxiliary CaVβ5 and CaVα2δ subunits6,7. Here we present cryo-electron microscopy structures of human brain and cardiac CaV1.2 bound with CaVβ3 to a chaperone-the endoplasmic reticulum membrane protein complex (EMC)8,9-and of the assembled CaV1.2-CaVβ3-CaVα2δ-1 channel. These structures provide a view of an EMC-client complex and define EMC sites-the transmembrane (TM) and cytoplasmic (Cyto) docks; interaction between these sites and the client channel causes partial extraction of a pore subunit and splays open the CaVα2δ-interaction site. The structures identify the CaVα2δ-binding site for gabapentinoid anti-pain and anti-anxiety drugs6, show that EMC and CaVα2δ interactions with the channel are mutually exclusive, and indicate that EMC-to-CaVα2δ hand-off involves a divalent ion-dependent step and CaV1.2 element ordering. Disruption of the EMC-CaV complex compromises CaV function, suggesting that the EMC functions as a channel holdase that facilitates channel assembly. Together, the structures reveal a CaV assembly intermediate and EMC client-binding sites that could have wide-ranging implications for the biogenesis of VGICs and other membrane proteins.
Collapse
Affiliation(s)
- Zhou Chen
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Abhisek Mondal
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Fayal Abderemane-Ali
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Seil Jang
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Sangeeta Niranjan
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - José L Montaño
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Balyn W Zaro
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, USA.
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
| |
Collapse
|
116
|
Sahagian M, Mastrocco A, Prittie J. Phenibut toxicosis in a dog. J Vet Emerg Crit Care (San Antonio) 2023; 33:472-476. [PMID: 37436877 DOI: 10.1111/vec.13313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 07/14/2023]
Abstract
OBJECTIVE To describe the successful treatment of severe neurological and cardiovascular abnormalities in a dog following ingestion of the neuropsychotropic drug, phenibut. CASE SUMMARY A 2-year-old neutered male Weimaraner was found unresponsive and laterally recumbent in his urine after ingesting approximately 1600 mg/kg of phenibut. On presentation to an emergency clinic, the dog was neurologically inappropriate, tachycardic, hypertensive, and exhibiting a profoundly decreased respiratory rate. Because of progressive clinical signs, electrolyte abnormalities, increased hepatic enzyme activity and bilirubin concentrations, and the development of pigmenturia, referral to specialist care was sought. On presentation, the dog was intermittently somnolent and then maniacal. Sinus tachycardia persisted, and hyperthermia was documented. Hospitalization for supportive care was undertaken, and the dog was administered IV fluids, flumazenil, antiepileptics, and IV lipid emulsion therapy. The dog developed hypoglycemia and treated with dextrose supplementation. Progressive increases in liver enzyme activities as well as pronounced increase in creatine kinase activity, consistent with rhabdomyolysis, were noted. Over the course of 48 hours, the hypoglycemia resolved, and clinical signs significantly improved. Ultimately, the dog was discharged with improved clinical signs, with the owner reporting that 1 week after discharge, a full recovery had been made, and no residual clinical signs persisted. NEW INFORMATION PROVIDED To the authors' knowledge, there are no previous reports of phenibut intoxication in small animals. The growing availability and use of this drug by people in the past several years highlight the need for a greater understanding of its effects in companion animals.
Collapse
Affiliation(s)
- Michael Sahagian
- Department of Emergency and Critical Care, The Animal Medical Center, New York, New York, USA
| | - Alicia Mastrocco
- Department of Emergency and Critical Care, The Animal Medical Center, New York, New York, USA
| | - Jennifer Prittie
- Department of Emergency and Critical Care, The Animal Medical Center, New York, New York, USA
| |
Collapse
|
117
|
Kim Y, Clemens EG, Farner JM, Londono-Barbaran A, Grab DJ, Dumler JS. Spotted fever rickettsia-induced microvascular endothelial barrier dysfunction is delayed by the calcium channel blocker benidipine. Biochem Biophys Res Commun 2023; 663:96-103. [PMID: 37121130 PMCID: PMC10362780 DOI: 10.1016/j.bbrc.2023.04.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/17/2023] [Indexed: 05/02/2023]
Abstract
The tick-borne bacterium Rickettsia parkeri is an obligate intracellular pathogen that belongs to spotted fever group rickettsia (SFGR). The SFG pathogens are characterized by their ability to infect and rapidly proliferate inside host vascular endothelial cells that eventually result in impairment of vascular endothelium barrier functions. Benidipine, a wide range dihydropyridine calcium channel blocker, is used to prevent and treat cardiovascular diseases. In this study, we tested whether benidipine has protective effects against rickettsia-induced microvascular endothelial cell barrier dysfunction in vitro. We utilized an in vitro vascular model consisting of transformed human brain microvascular endothelial cells (tHBMECs) and continuously monitored transendothelial electric resistance (TEER) across the cell monolayer. We found that during the late stages of infection when we observed TEER decrease and when there was a gradual increase of the cytoplasmic [Ca2+], benidipine prevented these rickettsia-induced effects. In contrast, nifedipine, another cardiovascular dihydropyridine channel blocker specific for L-type Ca2+ channels, did not prevent R. parkeri-induced drop of TEER. Additionally, neither drug was bactericidal. These data suggest that growth of R. parkeri inside endothelial cells is associated with impairment of endothelial cell monolayer integrity due to Ca2+ flooding through specific, benidipine-sensitive T- or N/Q-type Ca2+ channels but not through nifedipine-sensitive L-type Ca2+ channels. Further study will be required to discern the exact nature of the Ca2+ channels and Ca2+ transporting system(s) involved, any contributions of the pathogen toward this process, as well as the suitability of benidipine and new dihydropyridine derivatives as complimentary therapeutic drugs against Rickettsia-induced vascular failure.
Collapse
Affiliation(s)
- Yuri Kim
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA; Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Emily G Clemens
- Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Jennifer M Farner
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA; Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Andres Londono-Barbaran
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA; Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Dennis J Grab
- Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - J Stephen Dumler
- Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
118
|
Kessi M, Chen B, Pan L, Yang L, Yang L, Peng J, He F, Yin F. Disruption of mitochondrial and lysosomal functions by human CACNA1C variants expressed in HEK 293 and CHO cells. Front Mol Neurosci 2023; 16:1209760. [PMID: 37448958 PMCID: PMC10336228 DOI: 10.3389/fnmol.2023.1209760] [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: 04/21/2023] [Accepted: 05/30/2023] [Indexed: 07/18/2023] Open
Abstract
Objective To investigate the pathogenesis of three novel de novo CACNA1C variants (p.E411D, p.V622G, and p.A272V) in causing neurodevelopmental disorders and arrhythmia. Methods Several molecular experiments were carried out on transfected human embryonic kidney 293 (HEK 293) and Chinese hamster ovary (CHO) cells to explore the effects of p.E411D, p.V622G, and p.A272V variants on electrophysiology, mitochondrial and lysosomal functions. Electrophysiological studies, RT-qPCR, western blot, apoptosis assay, mito-tracker fluorescence intensity, lyso-tracker fluorescence intensity, mitochondrial calcium concentration test, and cell viability assay were performed. Besides, reactive oxygen species (ROS) levels, ATP levels, mitochondrial copy numbers, mitochondrial complex I, II, and cytochrome c functions were measured. Results The p.E411D variant was found in a patient with attention deficit-hyperactive disorder (ADHD), and moderate intellectual disability (ID). This mutant demonstrated reduced calcium current density, mRNA, and protein expression, and it was localized in the nucleus, cytoplasm, lysosome, and mitochondria. It exhibited an accelerated apoptosis rate, impaired autophagy, and mitophagy. It also demonstrated compromised mitochondrial cytochrome c oxidase, complex I, and II enzymes, abnormal mitochondrial copy numbers, low ATP levels, abnormal mitochondria fluorescence intensity, impaired mitochondrial fusion and fission, and elevated mitochondrial calcium ions. The p.V622G variant was identified in a patient who presented with West syndrome and moderate global developmental delay. The p.A272V variant was found in a patient who presented with epilepsy and mild ID. Both mutants (p.V622G and p.A272V) exhibited reduced calcium current densities, decreased mRNA and protein expressions, and they were localized in the nucleus, cytoplasm, lysosome, and mitochondria. They exhibited accelerated apoptosis and proliferation rates, impaired autophagy, and mitophagy. They also exhibited abnormal mitochondrial cytochrome c oxidase, complex I and II enzymes, abnormal mitochondrial copy numbers, low ATP, high ROS levels, abnormal mitochondria fluorescence intensity, impaired mitochondrial fusion and fission, as well as elevated mitochondrial calcium ions. Conclusion The p.E411D, p.V622G and p.A272V mutations of human CACNA1C reduce the expression level of CACNA1C proteins, and impair mitochondrial and lysosomal functions. These effects induced by CACNA1C variants may contribute to the pathogenesis of CACNA1C-related disorders.
Collapse
Affiliation(s)
- Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Baiyu Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Langui Pan
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Li Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
119
|
Al-Hassany L, Boucherie DM, Creeney H, van Drie RWA, Farham F, Favaretto S, Gollion C, Grangeon L, Lyons H, Marschollek K, Onan D, Pensato U, Stanyer E, Waliszewska-Prosół M, Wiels W, Chen HZ, Amin FM. Future targets for migraine treatment beyond CGRP. J Headache Pain 2023; 24:76. [PMID: 37370051 DOI: 10.1186/s10194-023-01567-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/14/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Migraine is a disabling and chronic neurovascular headache disorder. Trigeminal vascular activation and release of calcitonin gene-related peptide (CGRP) play a pivotal role in the pathogenesis of migraine. This knowledge has led to the development of CGRP(-receptor) therapies. Yet, a substantial proportion of patients do not respond to these treatments. Therefore, alternative targets for future therapies are warranted. The current narrative review provides a comprehensive overview of the pathophysiological role of these possible non-CGRP targets in migraine. FINDINGS We covered targets of the metabotropic receptors (pituitary adenylate cyclase-activating polypeptide (PACAP), vasoactive intestinal peptide (VIP), amylin, and adrenomedullin), intracellular targets (nitric oxide (NO), phosphodiesterase-3 (PDE3) and -5 (PDE5)), and ion channels (potassium, calcium, transient receptor potential (TRP), and acid-sensing ion channels (ASIC)). The majority of non-CGRP targets were able to induce migraine-like attacks, except for (i) calcium channels, as it is not yet possible to directly target channels to elucidate their precise involvement in migraine; (ii) TRP channels, activation of which can induce non-migraine headache; and (iii) ASICs, as their potential in inducing migraine attacks has not been investigated thus far. Drugs that target its receptors exist for PACAP, NO, and the potassium, TRP, and ASIC channels. No selective drugs exist for the other targets, however, some existing (migraine) treatments appear to indirectly antagonize responses to amylin, adrenomedullin, and calcium channels. Drugs against PACAP, NO, potassium channels, TRP channels, and only a PAC1 antibody have been tested for migraine treatment, albeit with ambiguous results. CONCLUSION While current research on these non-CGRP drug targets has not yet led to the development of efficacious therapies, human provocation studies using these targets have provided valuable insight into underlying mechanisms of migraine headaches and auras. Further studies are needed on these alternative therapies in non-responders of CGRP(-receptor) targeted therapies with the ultimate aim to pave the way towards a headache-free future for all migraine patients.
Collapse
Affiliation(s)
- Linda Al-Hassany
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Deirdre M Boucherie
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Hannah Creeney
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Ruben W A van Drie
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Cardiology, Division of Experimental Cardiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Fatemeh Farham
- Department of Headache, Iranian Centre of Neurological Researchers, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Silvia Favaretto
- Headache Center, Neurology Clinic, University Hospital of Padua, Padua, Italy
| | - Cédric Gollion
- Department of Neurology, University Hospital of Toulouse, Toulouse, France
| | - Lou Grangeon
- Neurology Department, Rouen University Hospital, Rouen, France
| | - Hannah Lyons
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Karol Marschollek
- Department of Neurology, Wroclaw Medical University, Wrocław, Poland
| | - Dilara Onan
- Spine Health Unit, Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Umberto Pensato
- Neurology and Stroke Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Humanitas University, Pieve Emanuele, Milan, Italy
| | - Emily Stanyer
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | | | - Wietse Wiels
- Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hui Zhou Chen
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Faisal Mohammad Amin
- Danish Headache Center, Department of Neurology, Faculty of Health and Medical Sciences, Rigshospitalet Glostrup, University of Copenhagen, Copenhagen, Denmark.
- Department of Neurorehabilitation/Traumatic Brain Injury, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
120
|
Cunha-Garcia D, Monteiro-Fernandes D, Correia JS, Neves-Carvalho A, Vilaça-Ferreira AC, Guerra-Gomes S, Viana JF, Oliveira JF, Teixeira-Castro A, Maciel P, Duarte-Silva S. Genetic Ablation of Inositol 1,4,5-Trisphosphate Receptor Type 2 (IP 3R2) Fails to Modify Disease Progression in a Mouse Model of Spinocerebellar Ataxia Type 3. Int J Mol Sci 2023; 24:10606. [PMID: 37445783 PMCID: PMC10341520 DOI: 10.3390/ijms241310606] [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: 05/15/2023] [Revised: 06/07/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3) is a rare neurodegenerative disease caused by an abnormal polyglutamine expansion within the ataxin-3 protein (ATXN3). This leads to neurodegeneration of specific brain and spinal cord regions, resulting in a progressive loss of motor function. Despite neuronal death, non-neuronal cells, including astrocytes, are also involved in SCA3 pathogenesis. Astrogliosis is a common pathological feature in SCA3 patients and animal models of the disease. However, the contribution of astrocytes to SCA3 is not clearly defined. Inositol 1,4,5-trisphosphate receptor type 2 (IP3R2) is the predominant IP3R in mediating astrocyte somatic calcium signals, and genetically ablation of IP3R2 has been widely used to study astrocyte function. Here, we aimed to investigate the relevance of IP3R2 in the onset and progression of SCA3. For this, we tested whether IP3R2 depletion and the consecutive suppression of global astrocytic calcium signalling would lead to marked changes in the behavioral phenotype of a SCA3 mouse model, the CMVMJD135 transgenic line. This was achieved by crossing IP3R2 null mice with the CMVMJD135 mouse model and performing a longitudinal behavioral characterization of these mice using well-established motor-related function tests. Our results demonstrate that IP3R2 deletion in astrocytes does not modify SCA3 progression.
Collapse
Affiliation(s)
- Daniela Cunha-Garcia
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Daniela Monteiro-Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Joana Sofia Correia
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Andreia Neves-Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Ana Catarina Vilaça-Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Sónia Guerra-Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - João Filipe Viana
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - João Filipe Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
- IPCA-EST-2Ai, Polytechnic Institute of Cávado and Ave, Applied Artificial Intelligence Laboratory, Campus of IPCA, 4750-810 Barcelos, Portugal
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| |
Collapse
|
121
|
Ding W, Zhao X, Wang H, Wang Y, Liu Y, Gong L, Lin S, Liu C, Li Y. Effect of Terahertz Electromagnetic Field on the Permeability of Potassium Channel Kv1.2. Int J Mol Sci 2023; 24:10271. [PMID: 37373419 DOI: 10.3390/ijms241210271] [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: 04/18/2023] [Revised: 06/10/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
In this paper, the influence of external terahertz electromagnetic fields with different frequencies of 4 THz, 10 THz, 15 THz, and 20 THz on the permeability of the Kv1.2 voltage-gated potassium ion channel on the nerve cell membrane was studied using the combined model of the "Constant Electric Field-Ion Imbalance" method by molecular dynamics. We found that although the applied terahertz electric field does not produce strong resonance with the -C=O groups of the conservative sequence T-V-G-Y-G amino acid residue of the selective filter (SF) of the channel, it would affect the stability of the electrostatic bond between potassium ions and the carbonyl group of T-V-G-Y-G of SF, and it would affect the stability of the hydrogen bond between water molecules and oxygen atoms of the hydroxyl group of the 374THR side chain at the SF entrance, changing the potential and occupied states of ions in the SF and the occurrence probability of the permeation mode of ions and resulting in the change in the permeability of the channel. Compared with no external electric field, when the external electric field with 15 THz frequency is applied, the lifetime of the hydrogen bond is reduced by 29%, the probability of the "soft knock on" mode is decreased by 46.9%, and the ion flux of the channel is activated by 67.7%. Our research results support the view that compared to "direct knock-on", "soft knock-on" is a slower permeation mode.
Collapse
Affiliation(s)
- Wen Ding
- Key Laboratory for Physical Electronics and Devices of the Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, China
- School of Electronic Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xiaofei Zhao
- Key Laboratory for Physical Electronics and Devices of the Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, China
- School of Electronic Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hongguang Wang
- Key Laboratory for Physical Electronics and Devices of the Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, China
- School of Electronic Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yize Wang
- Key Laboratory for Physical Electronics and Devices of the Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, China
- School of Electronic Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yanjiang Liu
- Key Laboratory for Physical Electronics and Devices of the Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, China
- School of Electronic Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Lirong Gong
- Key Laboratory for Physical Electronics and Devices of the Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, China
- School of Electronic Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Shu Lin
- Key Laboratory for Physical Electronics and Devices of the Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, China
- School of Electronic Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Chunliang Liu
- Key Laboratory for Physical Electronics and Devices of the Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, China
- School of Electronic Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yongdong Li
- Key Laboratory for Physical Electronics and Devices of the Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, China
- School of Electronic Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
122
|
Chen Z, Mondal A, Minor DL. Structural basis for Ca Vα 2δ:gabapentin binding. Nat Struct Mol Biol 2023; 30:735-739. [PMID: 36973510 PMCID: PMC10896480 DOI: 10.1038/s41594-023-00951-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/28/2023] [Indexed: 03/29/2023]
Abstract
Gabapentinoid drugs for pain and anxiety act on the CaVα2δ-1 and CaVα2δ-2 subunits of high-voltage-activated calcium channels (CaV1s and CaV2s). Here we present the cryo-EM structure of the gabapentin-bound brain and cardiac CaV1.2/CaVβ3/CaVα2δ-1 channel. The data reveal a binding pocket in the CaVα2δ-1 dCache1 domain that completely encapsulates gabapentin and define CaVα2δ isoform sequence variations that explain the gabapentin binding selectivity of CaVα2δ-1 and CaVα2δ-2.
Collapse
Affiliation(s)
- Zhou Chen
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Abhisek Mondal
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
- Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, USA.
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
123
|
Filippini L, Ortner NJ, Kaserer T, Striessnig J. Ca v 1.3-selective inhibitors of voltage-gated L-type Ca 2+ channels: Fact or (still) fiction? Br J Pharmacol 2023; 180:1289-1303. [PMID: 36788128 PMCID: PMC10953394 DOI: 10.1111/bph.16060] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/17/2022] [Accepted: 01/29/2023] [Indexed: 02/16/2023] Open
Abstract
Voltage-gated L-type Ca2+ -channels (LTCCs) are the target of Ca2+ -channel blockers (CCBs), which are in clinical use for the evidence-based treatment of hypertension and angina. Their cardiovascular effects are largely mediated by the Cav 1.2-subtype. However, based on our current understanding of their physiological and pathophysiological roles, Cav 1.3 LTCCs also appear as attractive drug targets for the therapy of various diseases, including treatment-resistant hypertension, spasticity after spinal cord injury and neuroprotection in Parkinson's disease. Since CCBs inhibit both Cav 1.2 and Cav 1.3, Cav 1.3-selective inhibitors would be valuable tools to validate the therapeutic potential of Cav 1.3 channel inhibition in preclinical models. Despite a number of publications reporting the discovery of Cav 1.3-selective blockers, their selectivity remains controversial. We conclude that at present no pharmacological tools exist that are suitable to confirm or refute a role of Cav 1.3 channels in cellular responses. We also suggest essential criteria for a small molecule to be considered Cav 1.3-selective.
Collapse
Affiliation(s)
- Ludovica Filippini
- Department of Pharmacology and Toxicology and Center of Molecular BiosciencesUniversity of InnsbruckInnsbruckAustria
- Department of Pharmaceutical Chemistry, Institute of PharmacyUniversity of InnsbruckInnsbruckAustria
| | - Nadine J. Ortner
- Department of Pharmacology and Toxicology and Center of Molecular BiosciencesUniversity of InnsbruckInnsbruckAustria
| | - Teresa Kaserer
- Department of Pharmaceutical Chemistry, Institute of PharmacyUniversity of InnsbruckInnsbruckAustria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology and Center of Molecular BiosciencesUniversity of InnsbruckInnsbruckAustria
| |
Collapse
|
124
|
Dong XY. Calcium Ion Channels in Saccharomyces cerevisiae. J Fungi (Basel) 2023; 9:jof9050524. [PMID: 37233235 DOI: 10.3390/jof9050524] [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: 03/29/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Regulating calcium ion (Ca2+) channels to improve the cell cycle and metabolism is a promising technology, ensuring increased cell growth, differentiation, and/or productivity. In this regard, the composition and structure of Ca2+ channels play a vital role in controlling the gating states. In this review, Saccharomyces cerevisiae, as a model eukaryotic organism and an essential industrial microorganism, was used to discuss the effect of its type, composition, structure, and gating mechanism on the activity of Ca2+ channels. Furthermore, the advances in the application of Ca2+ channels in pharmacology, tissue engineering, and biochemical engineering are summarized, with a special focus on exploring the receptor site of Ca2+ channels for new drug design strategies and different therapeutic uses, targeting Ca2+ channels to produce functional replacement tissues, creating favorable conditions for tissue regeneration, and regulating Ca2+ channels to enhance biotransformation efficiency.
Collapse
Affiliation(s)
- Xiao-Yu Dong
- College of Life and Health, Dalian University, Dalian 116622, China
| |
Collapse
|
125
|
Ali MY, Gadotti VM, Huang S, Garcia-Caballero A, Antunes FTT, Jung HA, Choi JS, Zamponi GW. Icariside II, a Prenyl-Flavonol, Alleviates Inflammatory and Neuropathic Pain by Inhibiting T-Type Calcium Channels and USP5-Cav3.2 Interactions. ACS Chem Neurosci 2023; 14:1859-1869. [PMID: 37116219 DOI: 10.1021/acschemneuro.3c00083] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
Cav3.2 channels play an important role in the afferent nociceptive pathway, which is responsible for both physiological and pathological pain transmission. Cav3.2 channels are upregulated during neuropathic pain or peripheral inflammation in part due to an increased association with the deubiquitinase USP5. In this study, we investigated nine naturally occurring flavonoid derivatives which we tested for their abilities to inhibit transiently expressed Cav3.2 channels and their interactions with USP5. Icariside II (ICA-II), one of the flavonols studied, inhibited the biochemical interactions between USP5 and Cav3.2 and concomitantly and effectively blocked Cav3.2 channels. Molecular docking analysis predicts that ICA-II binds to the cUBP domain and the Cav3.2 interaction region. In addition, ICA-II was predicted to interact with residues in close proximity to the Cav3.2 channel's fenestrations, thus accounting for the observed blocking activity. In mice with inflammatory and neuropathic pain, ICA-II inhibited both phases of the formalin-induced nocifensive responses and abolished thermal hyperalgesia induced by injection of complete Freund's adjuvant (CFA) into the hind paw. Furthermore, ICA-II produced significant and long-lasting thermal anti-hyperalgesia in female mice, whereas Cav3.2 null mice were resistant to the action of ICA-II. Altogether, our data show that ICA-II has analgesic activity via an action on Cav3.2 channels.
Collapse
Affiliation(s)
- Md Yousof Ali
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
- Zymedyne Therapeutics, Calgary, AB T2N4G4, Canada
| | - Vinicius M Gadotti
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
- Zymedyne Therapeutics, Calgary, AB T2N4G4, Canada
| | - Sun Huang
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
| | - Agustin Garcia-Caballero
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Zymedyne Therapeutics, Calgary, AB T2N4G4, Canada
| | - Flavia T T Antunes
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
| | - Hyun Ah Jung
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Jae Sue Choi
- Department of Food and Life Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
| |
Collapse
|
126
|
Bouron A. Neuronal Store-Operated Calcium Channels. Mol Neurobiol 2023:10.1007/s12035-023-03352-5. [PMID: 37118324 DOI: 10.1007/s12035-023-03352-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/13/2023] [Indexed: 04/30/2023]
Abstract
The endoplasmic reticulum (ER) is the major intracellular calcium (Ca2+) storage compartment in eukaryotic cells. In most instances, the mobilization of Ca2+ from this store is followed by a delayed and sustained uptake of Ca2+ through Ca2+-permeable channels of the cell surface named store-operated Ca2+ channels (SOCCs). This gives rise to a store-operated Ca2+ entry (SOCE) that has been thoroughly investigated in electrically non-excitable cells where it is the principal regulated Ca2+ entry pathway. The existence of this Ca2+ route in neurons has long been a matter of debate. However, a growing body of experimental evidence indicates that the recruitment of Ca2+ from neuronal ER Ca2+ stores generates a SOCE. The present review summarizes the main studies supporting the presence of a depletion-dependent Ca2+ entry in neurons. It also addresses the question of the molecular composition of neuronal SOCCs, their expression, pharmacological properties, as well as their physiological relevance.
Collapse
Affiliation(s)
- Alexandre Bouron
- Université Grenoble Alpes, CNRS, CEA, Inserm UA13 BGE, 38000, Grenoble, France.
| |
Collapse
|
127
|
Ovsepian SV, Waxman SG. Gene therapy for chronic pain: emerging opportunities in target-rich peripheral nociceptors. Nat Rev Neurosci 2023; 24:252-265. [PMID: 36658346 DOI: 10.1038/s41583-022-00673-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2022] [Indexed: 01/20/2023]
Abstract
With sweeping advances in precision delivery systems and manipulation of the genomes and transcriptomes of various cell types, medical biotechnology offers unprecedented selectivity for and control of a wide variety of biological processes, forging new opportunities for therapeutic interventions. This perspective summarizes state-of-the-art gene therapies enabled by recent innovations, with an emphasis on the expanding universe of molecular targets that govern the activity and function of primary sensory neurons and which might be exploited to effectively treat chronic pain.
Collapse
Affiliation(s)
- Saak V Ovsepian
- School of Science, Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, UK.
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
128
|
Kang Y, Xu L, Dong J, Huang Y, Yuan X, Li R, Chen L, Wang Z, Ji X. Calcium-based nanotechnology for cancer therapy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
129
|
Ritzer A, Roeschl T, Nay S, Rudakova E, Volk T. Rapid Pacing Decreases L-type Ca 2+ Current and Alters Cacna1c Isogene Expression in Primary Cultured Rat Left Ventricular Myocytes. J Membr Biol 2023; 256:257-269. [PMID: 36995425 DOI: 10.1007/s00232-023-00284-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 03/14/2023] [Indexed: 03/31/2023]
Abstract
The L-type calcium current (ICaL) is the first step in cardiac excitation-contraction-coupling and plays an important role in regulating contractility, but also in electrical and mechanical remodeling. Primary culture of cardiomyocytes, a widely used tool in cardiac ion channel research, is associated with substantial morphological, functional and electrical changes some of which may be prevented by electrical pacing. We therefore investigated ICaL directly after cell isolation and after 24 h of primary culture with and without regular pacing at 1 and 3 Hz in rat left ventricular myocytes. Moreover, we analyzed total mRNA expression of the pore forming subunit of the L-type Ca2+ channel (cacna1c) as well as the expression of splice variants of its exon 1 that contribute to specificity of ICaL in different tissue such as cardiac myocytes or smooth muscle. 24 h incubation without pacing decreased ICaL density by ~ 10% only. Consistent with this decrease we observed a decrease in the expression of total cacna1c and of exon 1a, the dominant variant of cardiomyocytes, while expression of exon 1b and 1c increased. Pacing for 24 h at 1 and 3 Hz led to a substantial decrease in ICaL density by 30%, mildly slowed ICaL inactivation and shifted steady-state inactivation to more negative potentials. Total cacna1c mRNA expression was substantially decreased by pacing, as was the expression of exon 1b and 1c. Taken together, electrical silence introduces fewer alterations in ICaL density and cacna1c mRNA expression than pacing for 24 h and should therefore be the preferred approach for primary culture of cardiomyocytes.
Collapse
Affiliation(s)
- Anne Ritzer
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Waldstraße 6, 91054, Erlangen, Germany
| | - Tobias Roeschl
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Waldstraße 6, 91054, Erlangen, Germany
| | - Sandra Nay
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Waldstraße 6, 91054, Erlangen, Germany
| | - Elena Rudakova
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Waldstraße 6, 91054, Erlangen, Germany
| | - Tilmann Volk
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Waldstraße 6, 91054, Erlangen, Germany.
- Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
| |
Collapse
|
130
|
Leyva-Leyva M, Sandoval A, Morales-Lázaro SL, Corzo-López A, Felix R, González-Ramírez R. Identification of Dp140 and α1-syntrophin as novel molecular interactors of the neuronal Ca V2.1 channel. Pflugers Arch 2023; 475:595-606. [PMID: 36964781 DOI: 10.1007/s00424-023-02803-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/21/2023] [Accepted: 03/06/2023] [Indexed: 03/26/2023]
Abstract
The primary function of dystrophin is to form a link between the cytoskeleton and the extracellular matrix. In addition to this crucial structural function, dystrophin also plays an essential role in clustering and organizing several signaling proteins, including ion channels. Proteomic analysis of the whole rodent brain has stressed the role of some components of the dystrophin-associated glycoprotein complex (DGC) as potential interacting proteins of the voltage-gated Ca2+ channels of the CaV2 subfamily. The interaction of CaV2 with signaling and scaffolding proteins, such as the DGC components, may influence their function, stability, and location in neurons. This work aims to study the interaction between dystrophin and CaV2.1. Our immunoprecipitation data showed the presence of a complex formed by CaV2.1, CaVα2δ-1, CaVβ4e, Dp140, and α1-syntrophin in the brain. Furthermore, proximity ligation assays (PLA) showed that CaV2.1 and CaVα2δ-1 interact with dystrophin in the hippocampus and cerebellum. Notably, Dp140 and α1-syntrophin increase CaV2.1 protein stability, half-life, permanence in the plasma membrane, and current density through recombinant CaV2.1 channels. Therefore, we have identified the Dp140 and α1-syntrophin as novel interaction partners of CaV2.1 channels in the mammalian brain. Consistent with previous findings, our work provides evidence of the role of DGC in anchoring and clustering CaV channels in a macromolecular complex.
Collapse
Affiliation(s)
- Margarita Leyva-Leyva
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Universidad Nacional Autónoma de México (UNAM), Ciudad de Mexico, México
| | - Alejandro Sandoval
- School of Medicine FES Iztacala, National Autonomous University of México (UNAM), Tlalnepantla, Mexico
| | - Sara Luz Morales-Lázaro
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - Alejandra Corzo-López
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Ricardo Felix
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico.
| | - Ricardo González-Ramírez
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico.
| |
Collapse
|
131
|
Pulgar VM, Harp J, Reeves TE. Molecular Modeling and Potential Ca2+ Channel Blocker Activity of Diphenylmethoxypiperidine Derivatives. CHEMISTRY 2023. [DOI: 10.3390/chemistry5020050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Molecular interactions of 4-diphenylmethoxy-1-methylpiperidine derivatives with the calcium channel CaV1.1 (pdb:6JP5) are described. All the compounds tested, previously shown to inhibit adrenergic vascular contractions, display similar binding energetics and interactions with the trans-membrane domain of 6JP5 on the opposite side relative to the channel pore, where nifedipine, a known dihydropyridine Ca2+ channel blocker binds. Additionally, the compounds tested inhibit Ca2+-dependent contractions in isolated mouse mesenteric arteries. Thus, diphenylpyraline analogs may exert their anticontractile effects, at least partially, by blocking vascular Ca2+ channels.
Collapse
|
132
|
Oz M, Lorke DE, Howarth FC. Transient receptor potential vanilloid 1 (TRPV1)-independent actions of capsaicin on cellular excitability and ion transport. Med Res Rev 2023. [PMID: 36916676 DOI: 10.1002/med.21945] [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: 06/14/2022] [Revised: 01/17/2023] [Accepted: 02/26/2023] [Indexed: 03/15/2023]
Abstract
Capsaicin is a naturally occurring alkaloid derived from chili pepper that is responsible for its hot pungent taste. Capsaicin is known to exert multiple pharmacological actions, including analgesia, anticancer, anti-inflammatory, antiobesity, and antioxidant effects. The transient receptor potential vanilloid subfamily member 1 (TRPV1) is the main receptor mediating the majority of the capsaicin effects. However, numerous studies suggest that the TRPV1 receptor is not the only target for capsaicin. An increasing number of studies indicates that capsaicin, at low to mid µM ranges, not only indirectly through TRPV1-mediated Ca2+ increases, but also directly modulates the functions of voltage-gated Na+ , K+ , and Ca2+ channels, as well as ligand-gated ion channels and other ion transporters and enzymes involved in cellular excitability. These TRPV1-independent effects are mediated by alterations of the biophysical properties of the lipid membrane and subsequent modulation of the functional properties of ion channels and by direct binding of capsaicin to the channels. The present study, for the first time, systematically categorizes this diverse range of non-TRPV1 targets and discusses cellular and molecular mechanisms mediating TRPV1-independent effects of capsaicin in excitable, as well as nonexcitable cells.
Collapse
Affiliation(s)
- Murat Oz
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Safat, Kuwait
| | - Dietrich E Lorke
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates.,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Frank C Howarth
- Department of Physiology, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| |
Collapse
|
133
|
Wang W, Yao J, Li W, Wu Y, Duan H, Xu C, Tian X, Li S, Tan Q, Zhang D. Epigenome-wide association study in Chinese monozygotic twins identifies DNA methylation loci associated with blood pressure. Clin Epigenetics 2023; 15:38. [PMID: 36869404 PMCID: PMC9985232 DOI: 10.1186/s13148-023-01457-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Hypertension is a crucial risk factor for developing cardiovascular disease and reducing life expectancy. We aimed to detect DNA methylation (DNAm) variants potentially related to systolic blood pressure (SBP) and diastolic blood pressure (DBP) by conducting epigenome-wide association studies in 60 and 59 Chinese monozygotic twin pairs, respectively. METHODS Genome-wide DNA methylation profiling in whole blood of twins was performed using Reduced Representation Bisulfite Sequencing, yielding 551,447 raw CpGs. Association between DNAm of single CpG and blood pressure was tested by applying generalized estimation equation. Differentially methylated regions (DMRs) were identified by comb-P approach. Inference about Causation through Examination of Familial Confounding was utilized to perform the causal inference. Ontology enrichment analysis was performed using Genomic Regions Enrichment of Annotations Tool. Candidate CpGs were quantified using Sequenom MassARRAY platform in a community population. Weighted gene co-expression network analysis (WGCNA) was conducted using gene expression data. RESULTS The median age of twins was 52 years (95% range 40, 66). For SBP, 31 top CpGs (p < 1 × 10-4) and 8 DMRs were identified, with several DMRs within NFATC1, CADM2, IRX1, COL5A1, and LRAT. For DBP, 43 top CpGs (p < 1 × 10-4) and 12 DMRs were identified, with several DMRs within WNT3A, CNOT10, and DAB2IP. Important pathways, such as Notch signaling pathway, p53 pathway by glucose deprivation, and Wnt signaling pathway, were significantly enriched for SBP and DBP. Causal inference analysis suggested that DNAm at top CpGs within NDE1, MYH11, SRRM1P2, and SMPD4 influenced SBP, while SBP influenced DNAm at CpGs within TNK2. DNAm at top CpGs within WNT3A influenced DBP, while DBP influenced DNAm at CpGs within GNA14. Three CpGs mapped to WNT3A and one CpG mapped to COL5A1 were validated in a community population, with a hypermethylated and hypomethylated direction in hypertension cases, respectively. Gene expression analysis by WGCNA further identified some common genes and enrichment terms. CONCLUSION We detect many DNAm variants that may be associated with blood pressure in whole blood, particularly the loci within WNT3A and COL5A1. Our findings provide new clues to the epigenetic modification underlying hypertension pathogenesis.
Collapse
Affiliation(s)
- Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, Qingdao, 266021, Shandong, China
| | - Jie Yao
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, Qingdao, 266021, Shandong, China
- Jiangsu Health Development Research Center, Nanjing, Jiangsu, China
| | - Weilong Li
- Population Research Unit, Faculty of Social Sciences, University of Helsinki, Helsinki, Finland
| | - Yili Wu
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, Qingdao, 266021, Shandong, China
| | - Haiping Duan
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Chunsheng Xu
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Xiaocao Tian
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Shuxia Li
- Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Qihua Tan
- Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, Qingdao, 266021, Shandong, China.
| |
Collapse
|
134
|
Török F, Tezcan K, Filippini L, Fernández-Quintero ML, Zanetti L, Liedl KR, Drexel RS, Striessnig J, Ortner NJ. Germline de novo variant F747S extends the phenotypic spectrum of CACNA1D Ca2+ channelopathies. Hum Mol Genet 2023; 32:847-859. [PMID: 36208199 PMCID: PMC9941835 DOI: 10.1093/hmg/ddac248] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/22/2022] [Accepted: 09/30/2022] [Indexed: 11/14/2022] Open
Abstract
Germline gain-of-function missense variants in the pore-forming Cav1.3 α1-subunit (CACNA1D gene) confer high risk for a severe neurodevelopmental disorder with or without endocrine symptoms. Here, we report a 4-week-old new-born with the novel de novo missense variant F747S with a so far not described prominent jittering phenotype in addition to symptoms previously reported for CACNA1D mutations including developmental delay, elevated aldosterone level and transient hypoglycemia. We confirmed the pathogenicity of this variant in whole-cell patch-clamp experiments with wild-type and F747S mutant channels heterologously expressed together with α2δ1 and cytosolic β3 or membrane-bound β2a subunits. Mutation F747S caused the quantitatively largest shift in the voltage dependence of activation (-28 mV) reported so far for CACNA1D germline mutations. It also shifted inactivation to more negative voltages, slowed the time course of current inactivation and slowed current deactivation upon repolarization with both co-expressed β-subunits. In silico modelling and molecular docking, simulations revealed that this gain-of-function phenotype can be explained by formation of a novel inter-domain hydrogen bond between mutant residues S747 (IIS6) with N1145 (IIIS6) stabilizing selectively the activated open channel state. F747S displayed 2-6-fold increased sensitivity for the L-type Ca2+ channel blocker isradipine compared to wild type. Our data confirm the pathogenicity of the F747S variant with very strong gain-of-function gating changes, which may contribute to the novel jittering phenotype. Increased sensitivity for isradipine suggests this drug for potential symptomatic off-label treatment for carriers of this mutation.
Collapse
Affiliation(s)
- Ferenc Török
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Kamer Tezcan
- Department of Genetics, Kaiser Permanente, Sacramento, CA 95825, USA
| | - Ludovica Filippini
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Monica L Fernández-Quintero
- Department of General, Inorganic and Theoretical Chemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Lucia Zanetti
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Klaus R Liedl
- Department of General, Inorganic and Theoretical Chemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Raphaela S Drexel
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Nadine J Ortner
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| |
Collapse
|
135
|
Jang W, Oh M, Cho EH, Baek M, Kim C. Drosophila pain sensitization and modulation unveiled by a novel pain model and analgesic drugs. PLoS One 2023; 18:e0281874. [PMID: 36795675 PMCID: PMC9934396 DOI: 10.1371/journal.pone.0281874] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 02/01/2023] [Indexed: 02/17/2023] Open
Abstract
In mammals, pain is regulated by the combination of an ascending stimulating and descending inhibitory pain pathway. It remains an intriguing question whether such pain pathways are of ancient origin and conserved in invertebrates. Here we report a new Drosophila pain model and use it to elucidate the pain pathways present in flies. The model employs transgenic flies expressing the human capsaicin receptor TRPV1 in sensory nociceptor neurons, which innervate the whole fly body, including the mouth. Upon capsaicin sipping, the flies abruptly displayed pain-related behaviors such as running away, scurrying around, rubbing vigorously, and pulling at their mouth parts, suggesting that capsaicin stimulated nociceptors in the mouth via activating TRPV1. When reared on capsaicin-containing food, the animals died of starvation, demonstrating the degree of pain experienced. This death rate was reduced by treatment both with NSAIDs and gabapentin, analgesics that inhibit the sensitized ascending pain pathway, and with antidepressants, GABAergic agonists, and morphine, analgesics that strengthen the descending inhibitory pathway. Our results suggest Drosophila to possess intricate pain sensitization and modulation mechanisms similar to mammals, and we propose that this simple, non-invasive feeding assay has utility for high-throughput evaluation and screening of analgesic compounds.
Collapse
Affiliation(s)
- Wijeong Jang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Myungsok Oh
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Eun-Hee Cho
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Minwoo Baek
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Changsoo Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
- * E-mail:
| |
Collapse
|
136
|
Nunes EJ, Addy NA. L-type calcium channel regulation of dopamine activity in the ventral tegmental area to nucleus accumbens pathway: Implications for substance use, mood disorders and co-morbidities. Neuropharmacology 2023; 224:109336. [PMID: 36414149 PMCID: PMC11215796 DOI: 10.1016/j.neuropharm.2022.109336] [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: 06/30/2022] [Revised: 11/07/2022] [Accepted: 11/13/2022] [Indexed: 11/21/2022]
Abstract
L-type calcium channels (LTCCs), including the Cav1.2 and Cav1.3 LTCC subtypes, are important regulators of calcium entry into neurons, which mediates neurotransmitter release and synaptic plasticity. Cav1.2 and Cav1.3 are encoded by the CACNA1C and CACNA1D genes, respectively. These genes are implicated in substance use disorders and depression in humans, as demonstrated by genetic-wide association studies (GWAS). Pre-clinical models have also revealed a critical role of LTCCs on drug and mood related behavior, including the co-morbidity of substance use and mood disorders. Moreover, LTCCs have been shown to regulate the neuronal firing of dopamine (DA) neurons as well as drug and stress-induced plasticity within the ventral tegmental area (VTA) to nucleus accumbens (NAc) pathway. Thus, LTCCs are interesting targets for the treatment of neuropsychiatric diseases. In this review, we provide a brief introduction to voltage-gated calcium channels, specifically focusing on the LTCCs. We place particular emphasis on the ability of LTCCs to regulate DA neuronal activity and downstream signaling in the VTA to NAc pathway, and how such processes mediate substance use and mood disorder-related behavioral responses. We also discuss the bi-directional control of VTA LTCCs on drug and mood-related behaviors in pre-clinical models, with implications for co-morbid psychiatric diagnosis. We conclude with a section on the clinical implications of LTCC blockers, many which are already FDA approved as cardiac medications. Thus, pre-clinical and clinical work should examine the potential of LTCC blockers to be repurposed for neuropsychiatric illness. This article is part of the Special Issue on 'L-type calcium channel mechanisms in neuropsychiatric disorders'.
Collapse
Affiliation(s)
- Eric J. Nunes
- Department of Psychiatry, Yale School of Medicine
- Yale Tobacco Center of Regulatory Science, Yale School of Medicine
| | - Nii A. Addy
- Department of Psychiatry, Yale School of Medicine
- Yale Tobacco Center of Regulatory Science, Yale School of Medicine
- Department of Cellular and Molecular Physiology, Yale School of Medicine
- Interdepartmental Neuroscience Program, Yale University
- Wu Tsai Institute, Yale University
| |
Collapse
|
137
|
Whole Exome Sequencing of Hemiplegic Migraine Patients Shows an Increased Burden of Missense Variants in CACNA1H and CACNA1I Genes. Mol Neurobiol 2023; 60:3034-3043. [PMID: 36786913 PMCID: PMC10122627 DOI: 10.1007/s12035-023-03255-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/02/2023] [Indexed: 02/15/2023]
Abstract
Hemiplegic migraine (HM) is a rare subtype of migraine with aura. Given that causal missense mutations in the voltage-gated calcium channel α1A subunit gene CACNA1A have been identified in a subset of HM patients, we investigated whether HM patients without a mutation have an increased burden of such variants in the "CACNA1x gene family". Whole exome sequencing data of an Australian cohort of unrelated HM patients (n = 184), along with public data from gnomAD, as controls, was used to assess the burden of missense variants in CACNA1x genes. We performed both a variant and a subject burden test. We found a significant burden for the number of variants in CACNA1E (p = 1.3 × 10-4), CACNA1H (p < 2.2 × 10-16) and CACNA1I (p < 2.2 × 10-16). There was also a significant burden of subjects with missense variants in CACNA1E (p = 6.2 × 10-3), CACNA1H (p < 2.2 × 10-16) and CACNA1I (p < 2.2 × 10-16). Both the number of variants and number of subjects were replicated for CACNA1H (p = 3.5 × 10-8; p = 0.012) and CACNA1I (p = 0.019, p = 0.044), respectively, in a Dutch clinical HM cohort (n = 32), albeit that CACNA1I did not remain significant after multiple testing correction. Our data suggest that HM, in the absence of a single causal mutation, is a complex trait, in which an increased burden of missense variants in CACNA1H and CACNA1I may contribute to the risk of disease.
Collapse
|
138
|
Čonkaš J, Sabol M, Ozretić P. 'Toxic Masculinity': What Is Known about the Role of Androgen Receptors in Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2023; 24:ijms24043766. [PMID: 36835177 PMCID: PMC9965076 DOI: 10.3390/ijms24043766] [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] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), the most prevalent cancer in the head and neck region, develops from the mucosal epithelium of the upper aerodigestive tract. Its development directly correlates with alcohol and/or tobacco consumption and infection with human papillomavirus. Interestingly, the relative risk for HNSCC is up to five times higher in males, so it is considered that the endocrine microenvironment is another risk factor. A gender-specific risk for HNSCC suggests either the existence of specific risk factors that affect only males or that females have defensive hormonal and metabolic features. In this review, we summarized the current knowledge about the role of both nuclear and membrane androgen receptors (nAR and mARs, respectively) in HNSCC. As expected, the significance of nAR is much better known; it was shown that increased nAR expression was observed in HNSCC, while treatment with dihydrotestosterone increased proliferation, migration, and invasion of HNSCC cells. For only three out of five currently known mARs-TRPM8, CaV1.2, and OXER1-it was shown either their increased expression in various types of HNSCC or that their increased activity enhanced the migration and invasion of HNSCC cells. The primary treatments for HNSCC are surgery and radiotherapy, but targeted immunotherapies are on the rise. On the other hand, given the evidence of elevated nAR expression in HNSCC, this receptor represents a potential target for antiandrogen therapy. Moreover, there is still plenty of room for further examination of mARs' role in HNSCC diagnosis, prognosis, and treatment.
Collapse
|
139
|
Structure-Function Studies of Sponge-Derived Compounds on the Cardiac Ca V3.1 Channel. Int J Mol Sci 2023; 24:ijms24043429. [PMID: 36834837 PMCID: PMC9962600 DOI: 10.3390/ijms24043429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
T-type calcium (CaV3) channels are involved in cardiac automaticity, development, and excitation-contraction coupling in normal cardiac myocytes. Their functional role becomes more pronounced in the process of pathological cardiac hypertrophy and heart failure. Currently, no CaV3 channel inhibitors are used in clinical settings. To identify novel T-type calcium channel ligands, purpurealidin analogs were electrophysiologically investigated. These compounds are alkaloids produced as secondary metabolites by marine sponges, and they exhibit a broad range of biological activities. In this study, we identified the inhibitory effect of purpurealidin I (1) on the rat CaV3.1 channel and conducted structure-activity relationship studies by characterizing the interaction of 119 purpurealidin analogs. Next, the mechanism of action of the four most potent analogs was investigated. Analogs 74, 76, 79, and 99 showed a potent inhibition on the CaV3.1 channel with IC50's at approximately 3 μM. No shift of the activation curve could be observed, suggesting that these compounds act like a pore blocker obstructing the ion flow by binding in the pore region of the CaV3.1 channel. A selectivity screening showed that these analogs are also active on hERG channels. Collectively, a new class of CaV3 channel inhibitors has been discovered and the structure-function studies provide new insights into the synthetic design of drugs and the mechanism of interaction with T-type CaV channels.
Collapse
|
140
|
Zhu J, Qiu H, Guo W. Probing ion binding in the selectivity filter of the Ca v1.1 channel with molecular dynamics. Biophys J 2023; 122:496-505. [PMID: 36587239 PMCID: PMC9941718 DOI: 10.1016/j.bpj.2022.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 11/11/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022] Open
Abstract
Cav1.1 is the voltage-gated calcium channel essential for the contraction of skeletal muscles upon membrane potential changes. Structural determination of the Cav1.1 channel opens the avenue toward understanding of the structure-function relationship of voltage-gated calcium channels. Here, we show that there exist two Ca2+-binding sites, termed S1 and S2, within the selectivity filter of Cav1.1 through extensive molecular dynamics simulations on various initial ion arrangement configurations. The formation of both binding sites is associated with the four Glu residues (Glu292/614/1014/1323) that constitute the so-called EEEE locus. At the S1 site near the extracellular side, the Ca2+ ion is coordinated with the negatively charged carboxylic groups of these Glu residues and of the Asp615 residue either in a direct way or via an intermediate water molecule. At the S2 site, Ca2+ binding shows two distinct states: an upper state involving two out of the four Glu residues in the EEEE locus and a lower state involving only one Glu residue. In addition, there exist two recruitment sites for Ca2+ above the entrance of the filter. These findings promote the understanding of mechanism for ion permeation and selectivity in calcium channels.
Collapse
Affiliation(s)
- Junliang Zhu
- Key Laboratory for Intelligent Nano Materials and Devices of the Ministry of Education, State Key Laboratory of Mechanics and Control of Mechanical Structures, Institute for Frontier Science, Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Hu Qiu
- Key Laboratory for Intelligent Nano Materials and Devices of the Ministry of Education, State Key Laboratory of Mechanics and Control of Mechanical Structures, Institute for Frontier Science, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
| | - Wanlin Guo
- Key Laboratory for Intelligent Nano Materials and Devices of the Ministry of Education, State Key Laboratory of Mechanics and Control of Mechanical Structures, Institute for Frontier Science, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
| |
Collapse
|
141
|
Matthews LG, Puryear CB, Correia SS, Srinivasan S, Belfort GM, Pan MK, Kuo SH. T-type calcium channels as therapeutic targets in essential tremor and Parkinson's disease. Ann Clin Transl Neurol 2023; 10:462-483. [PMID: 36738196 PMCID: PMC10109288 DOI: 10.1002/acn3.51735] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 02/05/2023] Open
Abstract
Neuronal action potential firing patterns are key components of healthy brain function. Importantly, restoring dysregulated neuronal firing patterns has the potential to be a promising strategy in the development of novel therapeutics for disorders of the central nervous system. Here, we review the pathophysiology of essential tremor and Parkinson's disease, the two most common movement disorders, with a focus on mechanisms underlying the genesis of abnormal firing patterns in the implicated neural circuits. Aberrant burst firing of neurons in the cerebello-thalamo-cortical and basal ganglia-thalamo-cortical circuits contribute to the clinical symptoms of essential tremor and Parkinson's disease, respectively, and T-type calcium channels play a key role in regulating this activity in both the disorders. Accordingly, modulating T-type calcium channel activity has received attention as a potentially promising therapeutic approach to normalize abnormal burst firing in these diseases. In this review, we explore the evidence supporting the theory that T-type calcium channel blockers can ameliorate the pathophysiologic mechanisms underlying essential tremor and Parkinson's disease, furthering the case for clinical investigation of these compounds. We conclude with key considerations for future investigational efforts, providing a critical framework for the development of much needed agents capable of targeting the dysfunctional circuitry underlying movement disorders such as essential tremor, Parkinson's disease, and beyond.
Collapse
Affiliation(s)
| | - Corey B Puryear
- Praxis Precision Medicines, Boston, Massachusetts, 02110, USA
| | | | - Sharan Srinivasan
- Praxis Precision Medicines, Boston, Massachusetts, 02110, USA.,Department of Neurology, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | | | - Ming-Kai Pan
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, 10051, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, 10617, Taiwan.,Department of Medical Research, National Taiwan University Hospital, Taipei, 10002, Taiwan.,Cerebellar Research Center, National Taiwan University Hospital, Yun-Lin Branch, Yun-Lin, 64041, Taiwan
| | - Sheng-Han Kuo
- Department of Neurology, Columbia University, New York, New York, 10032, USA.,Initiative for Columbia Ataxia and Tremor, Columbia University, New York, New York, 10032, USA
| |
Collapse
|
142
|
Mustafá ER, Weiss N. Extended spectrum of Ca v1.3 channelopathies. Pflugers Arch 2023; 475:147-149. [PMID: 36307590 DOI: 10.1007/s00424-022-02766-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 02/01/2023]
Affiliation(s)
- Emilio R Mustafá
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Norbert Weiss
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
143
|
Chege BM, Nyaga NM, Kaur PS, Misigo WO, Khan N, Wanyonyi WC, Mwangi PW. The significant antidyslipidemic, hypoglycemic, antihyperglycemic, and antiobesity activities of the aqueous extracts of Agave Sisalana juice are partly mediated via modulation of calcium signaling pathways. Heliyon 2023; 9:e12400. [PMID: 36816233 PMCID: PMC9932365 DOI: 10.1016/j.heliyon.2022.e12400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/15/2022] [Accepted: 12/08/2022] [Indexed: 12/26/2022] Open
Abstract
Plant species in the genus Agave, including Agave sisalana, have found extensive application in African and Asian traditional medicine. Inspired by the use of the edible sweet sap known as Aguamiel (obtained from specific mature agave species such as Agave salmiana) in Mexico by diabetic patients to improve their diabetic condition, this study investigated the effects of Agave sisalana extracts prepared by lyophilization, fermentation, and saponin extraction from sisal juice in a rodent model of metabolic syndrome. The metabolic syndrome was induced by administering a high fat and high fructose diet to freshly weaned Sprague-Dawley rats for eight weeks. The A. sisalana extracts possessed significant hypoglycemic effects [3.883 ± 0.371 mmol/L (normal group) vs. 8.183 ± 0.5845 mmol/L (negative control) vs. 3.767 ± 0.2716 mmol/L (positive control) vs. 4.167 ± 0.4602 mmol/L (FSP) vs. 4.533 ± 0.3169 mmol/L (FerSP) vs. 3.5 ± 0.2309 mmol/L (FS LD) vs. 3.867 ± 0.3353 mmol/L (FS HD) vs. 4.617 ± 0.2725 mmol/L (FerS LD) vs. 4.383 ± 0.3114 mmol/L (FerS HD): p < 0.0001]. The extracts also possessed significant antihyperlipidemic effects with significant differences in total serum cholesterol between the groups [1.398 ± 0.1232 mmol/L (normal group) vs. 4.225 ± 0.4135 mmol/L (negative control) vs. 1.582 ± 0.154 mmol/L (positive control) vs. 1.245 ± 0.0911 mmol/L (FSP) vs. 1.393 ± 0.1423 mmol/L (FerSP) vs. 1.387 ± 0.0924 mmol/L (FS LD) vs. 1.761 ± 0.1495 mmol/L (FS HD) vs. 1.698 ± 0.1294 mmol/L (FerS LD) vs. 1.6975 ± 0.0982 mmol/L (FerS HD): p < 0.0001]. Further, significant antiobesity effects of the A.sisalana extracts were observed with significant differences in weight among the groups [196.3 ± 6.49 g (normal group) vs. 298.9 ± 6.67 g (negative control) vs. 215.3 ± 6.06 g (positive control) vs. 195.4 ± 3.92 g (FSP) vs. 213.1 ± 5.21 g (FerSP) vs. 190.8 ± 6.49 g (FS LD) vs. 198.9 ± 4.31 g (FS HD) vs. 204.7 ± 4.78 g (FerS LD) vs. 208.7 ± 6.21 g (FerS HD): p < 0.0001]. Network pharmacology studies indicated that the chemical components found in sisal juice primarily exert their effects by modulating the voltage-gated calcium channels CACNA1S, CACNA1D, and CACNA1C, in the beta cells of the islets of Langerhans.
Collapse
Affiliation(s)
| | | | | | | | - Nelson Khan
- University of Nairobi, Department of Biochemistry, Kenya
| | | | | |
Collapse
|
144
|
Naudin L, Raison-Aubry L, Buhry L. A general pattern of non-spiking neuron dynamics under the effect of potassium and calcium channel modifications. J Comput Neurosci 2023; 51:173-186. [PMID: 36371576 DOI: 10.1007/s10827-022-00840-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/08/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022]
Abstract
Electrical activity of excitable cells results from ion exchanges through cell membranes, so that genetic or epigenetic changes in genes encoding ion channels are likely to affect neuronal electrical signaling throughout the brain. There is a large literature on the effect of variations in ion channels on the dynamics of spiking neurons that represent the main type of neurons found in the vertebrate nervous systems. Nevertheless, non-spiking neurons are also ubiquitous in many nervous tissues and play a critical role in the processing of some sensory systems. To our knowledge, however, how conductance variations affect the dynamics of non-spiking neurons has never been assessed. Based on experimental observations reported in the biological literature and on mathematical considerations, we first propose a phenotypic classification of non-spiking neurons. Then, we determine a general pattern of the phenotypic evolution of non-spiking neurons as a function of changes in calcium and potassium conductances. Furthermore, we study the homeostatic compensatory mechanisms of ion channels in a well-posed non-spiking retinal cone model. We show that there is a restricted range of ion conductance values for which the behavior and phenotype of the neuron are maintained. Finally, we discuss the implications of the phenotypic changes of individual cells at the level of neuronal network functioning of the C. elegans worm and the retina, which are two non-spiking nervous tissues composed of neurons with various phenotypes.
Collapse
Affiliation(s)
- Loïs Naudin
- Laboratoire Lorrain de Recherche en Informatique et ses Applications, CNRS, Université de Lorraine, Nancy, France.
| | - Laetitia Raison-Aubry
- Laboratoire Lorrain de Recherche en Informatique et ses Applications, CNRS, Université de Lorraine, Nancy, France
| | - Laure Buhry
- Laboratoire Lorrain de Recherche en Informatique et ses Applications, CNRS, Université de Lorraine, Nancy, France.
| |
Collapse
|
145
|
Gao Y, Xu S, Cui X, Xu H, Qiu Y, Wei Y, Dong Y, Zhu B, Peng C, Liu S, Zhang XC, Sun J, Huang Z, Zhao Y. Molecular insights into the gating mechanisms of voltage-gated calcium channel Ca V2.3. Nat Commun 2023; 14:516. [PMID: 36720859 PMCID: PMC9889812 DOI: 10.1038/s41467-023-36260-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/23/2023] [Indexed: 02/02/2023] Open
Abstract
High-voltage-activated R-type CaV2.3 channel plays pivotal roles in many physiological activities and is implicated in epilepsy, convulsions, and other neurodevelopmental impairments. Here, we determine the high-resolution cryo-electron microscopy (cryo-EM) structure of human CaV2.3 in complex with the α2δ1 and β1 subunits. The VSDII is stabilized in the resting state. Electrophysiological experiments elucidate that the VSDII is not required for channel activation, whereas the other VSDs are essential for channel opening. The intracellular gate is blocked by the W-helix. A pre-W-helix adjacent to the W-helix can significantly regulate closed-state inactivation (CSI) by modulating the association and dissociation of the W-helix with the gate. Electrostatic interactions formed between the negatively charged domain on S6II, which is exclusively conserved in the CaV2 family, and nearby regions at the alpha-interacting domain (AID) and S4-S5II helix are identified. Further functional analyses indicate that these interactions are critical for the open-state inactivation (OSI) of CaV2 channels.
Collapse
Affiliation(s)
- Yiwei Gao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shuai Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaoli Cui
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China
| | - Hao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yunlong Qiu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yiqing Wei
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yanli Dong
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Boling Zhu
- Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Chao Peng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Shiqi Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Xuejun Cai Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jianyuan Sun
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China.,The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China. .,IDG/McGovern Institute for Brain Research, Peking University, Beijing, China.
| | - Yan Zhao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China. .,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing, China. .,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
146
|
Vitamin C Modes of Action in Calcium-Involved Signaling in the Brain. Antioxidants (Basel) 2023; 12:antiox12020231. [PMID: 36829790 PMCID: PMC9952025 DOI: 10.3390/antiox12020231] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Vitamin C (ascorbic acid) is well known for its potent antioxidant properties, as it can neutralize ROS and free radicals, thereby protecting cellular elements from oxidative stress. It predominantly exists as an ascorbate anion and after oxidation to dehydroascorbic acid and further breakdown, is removed from the cells. In nervous tissue, a progressive decrease in vitamin C level or its prolonged deficiency have been associated with an increased risk of disturbances in neurotransmission, leading to dysregulation in brain function. Therefore, understanding the regulatory function of vitamin C in antioxidant defence and identification of its molecular targets deserves more attention. One of the key signalling ions is calcium and a transient rise in its concentration is crucial for all neuronal processes. Extracellular Ca2+ influx (through specific ion channels) or Ca2+ release from intracellular stores (endoplasmic reticulum, mitochondria) are precisely controlled. Ca2+ regulates the functioning of the CNS, including growth, development, myelin formation, synthesis of catecholamines, modulation of neurotransmission and antioxidant protection. A growing body of evidence indicates a unique role for vitamin C in these processes. In this short review, we focus on vitamin C in the regulation of calcium-involved pathways under physiological and stress conditions in the brain.
Collapse
|
147
|
Modelling altered signalling of G-protein coupled receptors in inflamed environment to advance drug design. Sci Rep 2023; 13:607. [PMID: 36635362 PMCID: PMC9837128 DOI: 10.1038/s41598-023-27699-w] [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: 05/03/2022] [Accepted: 01/05/2023] [Indexed: 01/14/2023] Open
Abstract
We previously reported the successful design, synthesis and testing of the prototype opioid painkiller NFEPP that does not elicit adverse side effects. The design process of NFEPP was based on mathematical modelling of extracellular interactions between G-protein coupled receptors (GPCRs) and ligands, recognizing that GPCRs function differently under pathological versus healthy conditions. We now present an additional and novel stochastic model of GPCR function that includes intracellular dissociation of G-protein subunits and modulation of plasma membrane calcium channels and their dependence on parameters of inflamed and healthy tissue (pH, radicals). The model is validated against in vitro experimental data for the ligands NFEPP and fentanyl at different pH values and radical concentrations. We observe markedly reduced binding affinity and calcium channel inhibition for NFEPP at normal pH compared to lower pH, in contrast to the effect of fentanyl. For increasing radical concentrations, we find enhanced constitutive G-protein activation but reduced ligand binding affinity. Assessing the different effects, the results suggest that, compared to radicals, low pH is a more important determinant of overall GPCR function in an inflamed environment. Future drug design efforts should take this into account.
Collapse
|
148
|
Folacci M, Estaran S, Ménard C, Bertaud A, Rousset M, Roussel J, Thibaud JB, Vignes M, Chavanieu A, Charnet P, Cens T. Functional Characterization of Four Known Cav2.1 Variants Associated with Neurodevelopmental Disorders. MEMBRANES 2023; 13:96. [PMID: 36676903 PMCID: PMC9864995 DOI: 10.3390/membranes13010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 06/17/2023]
Abstract
Cav2.1 channels are expressed throughout the brain and are the predominant Ca2+ channels in the Purkinje cells. These cerebellar neurons fire spontaneously, and Cav2.1 channels are involved in the regular pacemaking activity. The loss of precision of the firing pattern of Purkinje cells leads to ataxia, a disorder characterized by poor balance and difficulties in performing coordinated movements. In this study, we aimed at characterizing functional and structural consequences of four variations (p.A405T in I-II loop and p.R1359W, p.R1667W and p.S1799L in IIIS4, IVS4, and IVS6 helices, respectively) identified in patients exhibiting a wide spectrum of disorders including ataxia symptoms. Functional analysis using two major Cav2.1 splice variants (Cav2.1+e47 and Cav2.1-e47) in Xenopus laevis oocytes, revealed a lack of effect upon A405T substitution and a significant loss-of-function caused by R1359W, whereas R1667W and S1799L caused both channel gain-of-function and loss-of-function, in a splice variant-dependent manner. Structural analysis revealed the loss of interactions with S1, S2, and S3 helices upon R1359W and R1667W substitutions, but a lack of obvious structural changes with S1799L. Computational modeling suggests that biophysical changes induced by Cav2.1 pathogenic mutations might affect action potential frequency in Purkinje cells.
Collapse
|
149
|
Ortner NJ. CACNA1D-Related Channelopathies: From Hypertension to Autism. Handb Exp Pharmacol 2023. [PMID: 36592224 DOI: 10.1007/164_2022_626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Tightly controlled Ca2+ influx through voltage-gated Ca2+ channels (Cavs) is indispensable for proper physiological function. Thus, it is not surprising that Cav loss and/or gain of function have been implicated in human pathology. Deficiency of Cav1.3 L-type Ca2+ channels (LTCCs) causes deafness and bradycardia, whereas several genetic variants of CACNA1D, the gene encoding the pore-forming α1 subunit of Cav1.3, have been linked to various disease phenotypes, such as hypertension, congenital hypoglycemia, or autism. These variants include not only common polymorphisms associated with an increased disease risk, but also rare de novo missense variants conferring high risk. This review provides a concise summary of disease-associated CACNA1D variants, whereas the main focus lies on de novo germline variants found in individuals with a neurodevelopmental disorder of variable severity. Electrophysiological recordings revealed activity-enhancing gating changes induced by these de novo variants, and tools to predict their pathogenicity and to study the resulting pathophysiological consequences will be discussed. Despite the low number of affected patients, potential phenotype-genotype correlations and factors that could impact the severity of symptoms will be covered. Since increased channel activity is assumed as the disease-underlying mechanism, pharmacological inhibition could be a treatment option. In the absence of Cav1.3-selective blockers, dihydropyridine LTCC inhibitors clinically approved for the treatment of hypertension may be used for personalized off-label trials. Findings from in vitro studies and treatment attempts in some of the patients seem promising as outlined. Taken together, due to advances in diagnostic sequencing techniques the number of reported CACNA1D variants in human diseases is constantly rising. Evidence from in silico, in vitro, and in vivo disease models can help to predict the pathogenic potential of such variants and to guide diagnosis and treatment in the clinical practice when confronted with patients harboring CACNA1D variants.
Collapse
Affiliation(s)
- Nadine J Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
150
|
Waclawiková B, Cesar Telles de Souza P, Schwalbe M, Neochoritis CG, Hoornenborg W, Nelemans SA, Marrink SJ, El Aidy S. Potential binding modes of the gut bacterial metabolite, 5-hydroxyindole, to the intestinal L-type calcium channels and its impact on the microbiota in rats. Gut Microbes 2023; 15:2154544. [PMID: 36511640 PMCID: PMC9754111 DOI: 10.1080/19490976.2022.2154544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Intestinal microbiota and microbiota-derived metabolites play a key role in regulating the host physiology. Recently, we have identified a gut-bacterial metabolite, namely 5-hydroxyindole, as a potent stimulant of intestinal motility via its modulation of L-type voltage-gated calcium channels located on the intestinal smooth muscle cells. Dysregulation of L-type voltage-gated calcium channels is associated with various gastrointestinal motility disorders, including constipation, making L-type voltage-gated calcium channels an important target for drug development. Nonetheless, the majority of currently available drugs are associated with alteration of the gut microbiota. Using 16S rRNA sequencing this study shows that, when administered orally, 5-hydroxyindole has only marginal effects on the rat cecal microbiota. Molecular dynamics simulations propose potential-binding pockets of 5-hydroxyindole in the α1 subunit of the L-type voltage-gated calcium channels and when its stimulatory effect on the rat colonic contractility was compared to 16 different analogues, ex-vivo, 5-hydroxyindole stood as the most potent enhancer of the intestinal contractility. Overall, the present findings imply a potential role of microbiota-derived metabolites as candidate therapeutics for targeted treatment of slow intestinal motility-related disorders including constipation.
Collapse
Affiliation(s)
- Barbora Waclawiková
- Host-Microbe Metabolic Interactions, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, The Netherlands
| | - Paulo Cesar Telles de Souza
- Molecular Microbiology and Structural Biochemistry (MMSB - UMR 5086), CNRS & University of Lyon, Lyon, France
| | - Markus Schwalbe
- Host-Microbe Metabolic Interactions, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, The Netherlands
| | | | - Warner Hoornenborg
- Department of Behavioral Neurosciences, Cluster Neurobiology, Groningen Institute of for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Sieger A. Nelemans
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Siewert J. Marrink
- Molecular Dynamics, Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen, Groningen, The Netherlands
| | - Sahar El Aidy
- Host-Microbe Metabolic Interactions, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, The Netherlands,CONTACT Sahar El Aidy Host-Microbe Metabolic Interactions, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, The Netherlands
| |
Collapse
|