1
|
Yao X, Gao S, Yan N. Structural biology of voltage-gated calcium channels. Channels (Austin) 2024; 18:2290807. [PMID: 38062897 PMCID: PMC10761187 DOI: 10.1080/19336950.2023.2290807] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Voltage-gated calcium (Cav) channels mediate Ca2+ influx in response to membrane depolarization, playing critical roles in diverse physiological processes. Dysfunction or aberrant regulation of Cav channels can lead to life-threatening consequences. Cav-targeting drugs have been clinically used to treat cardiovascular and neuronal disorders for several decades. This review aims to provide an account of recent developments in the structural dissection of Cav channels. High-resolution structures have significantly advanced our understanding of the working and disease mechanisms of Cav channels, shed light on the molecular basis for their modulation, and elucidated the modes of actions (MOAs) of representative drugs and toxins. The progress in structural studies of Cav channels lays the foundation for future drug discovery efforts targeting Cav channelopathies.
Collapse
Affiliation(s)
- Xia Yao
- TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Shuai Gao
- TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Shenzhen Medical Academy of Research and Translation, Shenzhen, China
| |
Collapse
|
2
|
Ciesluk B, Inglis DG, Parke A, Troup LJ. Systematic review: The relationship between gabapentinoids, etizolam, and drug related deaths in Scotland. PLoS One 2024; 19:e0310655. [PMID: 39383139 PMCID: PMC11463789 DOI: 10.1371/journal.pone.0310655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/04/2024] [Indexed: 10/11/2024] Open
Abstract
In recent years Scotland has been experiencing a disproportionally high number of drug related deaths compared to other European countries, causing significant individual, societal and economic burden. A possible cause of this is the increase in average number of substances involved in Scottish drug related deaths, as well as the changing pattern of substances involved. Opioids, cocaine, and alcohol have been consistently involved in the culture of drug use in Scotland, however recently National Records Scotland have identified that designer benzodiazepines such as etizolam, and prescription drugs such as gabapentinoids are increasingly being detected in Scottish toxicology reports. A systematic literature review following PRISMA guidelines was conducted through searching PubMed and Google Scholar to identify peer-reviewed articles published in English between 2013 and 2023 that investigated Scottish population data on gabapentinoids and etizolam to establish their contribution to the rise in Scottish drug related deaths. 18 studies were included in the review. A high use prevalence of etizolam and gabapentinoids in Scotland has been identified, with both substance-related deaths showing recent increase, marked since 2015. This pattern is replicated in the Scottish prison system. There has also been a significant increase of gabapentinoids prescriptions in Scotland. Polydrug use was identified as the most common determinant of both etizolam and gabapentinoids related adverse effects and fatality in Scotland, especially concurrent opioid use. The results indicate the literature on individual characteristics of Scottish at-risk users of gabapentinoids and etizolam is limited, however the data shows both substances are being used by older cohort, with adverse effects seen more in older women.
Collapse
Affiliation(s)
- Beata Ciesluk
- University of the West of Scotland, School of Education and Social Sciences, Paisley, Scotland, United Kingdom
| | - Dr. Greig Inglis
- University of the West of Scotland, School of Education and Social Sciences, Paisley, Scotland, United Kingdom
| | - Adrian Parke
- University of the West of Scotland, School of Education and Social Sciences, Paisley, Scotland, United Kingdom
| | - Lucy J. Troup
- University of the West of Scotland, School of Education and Social Sciences, Paisley, Scotland, United Kingdom
| |
Collapse
|
3
|
Scott MB, Kammermeier PJ. Rat Sympathetic Neuron Calcium Channels Are Insensitive to Gabapentin. Pharmaceuticals (Basel) 2024; 17:1237. [PMID: 39338399 PMCID: PMC11435301 DOI: 10.3390/ph17091237] [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: 08/02/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
The gabapentenoids such as gabapentin (GP) and pregabalin are approved for the treatment of chronic pain, but their utility is limited by persistent side effects. These adverse effects result from GPs affecting many types of neurons and muscle cells, not just the pain-sensing neurons that are the intended targets. We have recently discovered a type of peripheral neuron, rat sympathetic neurons from the superior cervical ganglion (SCG), that is uniquely insensitive to GP effects. Currents were measured using whole-cell patch-clamp electrophysiology from cells in primary culture from either the SCG or the Nodose Ganglion (NDG) as a positive control for the effects of GP. We find that the calcium current density was dramatically reduced by GP pretreatment in NDG neurons, but that neurons from the SCG were resistant. Further, when GP was cytoplasmically injected into these neurons, the resistance of SCG neurons to GP treatment persisted. These data demonstrate that rat sympathetic neurons appear to be uniquely resistant to GP treatment. These results may help us to better understand the mechanism of action of, and resistance to, GP in altering calcium channel current density, which may help to develop future treatments with fewer side effects.
Collapse
Affiliation(s)
| | - Paul J. Kammermeier
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
4
|
Bats C, Coombs ID, Farrant M, Cull-Candy SG. α-Bungarotoxin labelling of AMPA receptor-associated TARPs in living neurons. Neuroscience 2024:S0306-4522(24)00440-8. [PMID: 39209103 DOI: 10.1016/j.neuroscience.2024.08.036] [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: 05/21/2024] [Revised: 07/30/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
In mammalian central neurons AMPARs are clustered at glutamatergic synapses where they mediate fast excitatory transmission. In addition to four pore-forming subunits (GluA1-4), AMPARs contain auxiliary transmembrane AMPAR regulatory proteins (γ2, γ3, γ4, γ5, γ7 or γ8) whose incorporation can vary between neuron types, brain regions, and stages of development. As well as modulating the functional properties of AMPARs, these auxiliary subunits play a central role in AMPAR trafficking. Directly visualizing TARPs could therefore provide a valuable insight into mechanisms underlying these processes. Although antibodies are routinely used for the detection of surface proteins, our experience suggests anti-TARP antibodies are too bulky to access their target, possibly due to close interactions between the extracellular domains of TARP and AMPAR subunits. We therefore assessed the utility of a small monovalent probe - fluorescent α-bungarotoxin (α-Btx) - for TARP labelling in living neurons. We inserted the bungarotoxin binding site (BBS) within the extracellular domain of TARPs to enable their detection in cells exposed to fluorescent α-Btx. Focusing on the prototypical TARP γ2, we demonstrate that the small size of fluorescent α-Btx allows it to bind to the BBS-tagged TARP when associated with AMPARs. Importantly, labelled γ2 enhances AMPAR function in the same way as unmodified γ2. In living neurons, fluorescent α-Btx-labelled γ2 associates with AMPAR clusters at synapses. As a proof-of-principle, we employed our method to compare the surface trafficking of γ2 and γ7 in cerebellar stellate neurons. Our approach provides a simple way to visualize TARPs within AMPARs in living cells.
Collapse
Affiliation(s)
- Cecile Bats
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Ian D Coombs
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Mark Farrant
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.
| | - Stuart G Cull-Candy
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
5
|
Garza-Carbajal A, Bavencoffe A, Herrera JJ, Johnson KN, Walters ET, Dessauer CW. Mechanism of gabapentinoid potentiation of opioid effects on cyclic AMP signaling in neuropathic pain. Proc Natl Acad Sci U S A 2024; 121:e2405465121. [PMID: 39145932 PMCID: PMC11348325 DOI: 10.1073/pnas.2405465121] [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: 03/15/2024] [Accepted: 07/10/2024] [Indexed: 08/16/2024] Open
Abstract
Over half of spinal cord injury (SCI) patients develop opioid-resistant chronic neuropathic pain. Safer alternatives to opioids for treatment of neuropathic pain are gabapentinoids (e.g., pregabalin and gabapentin). Clinically, gabapentinoids appear to amplify opioid effects, increasing analgesia and overdose-related adverse outcomes, but in vitro proof of this amplification and its mechanism are lacking. We previously showed that after SCI, sensitivity to opioids is reduced by fourfold to sixfold in rat sensory neurons. Here, we demonstrate that after injury, gabapentinoids restore normal sensitivity of opioid inhibition of cyclic AMP (cAMP) generation, while reducing nociceptor hyperexcitability by inhibiting voltage-gated calcium channels (VGCCs). Increasing intracellular Ca2+ or activation of L-type VGCCs (L-VGCCs) suffices to mimic SCI effects on opioid sensitivity, in a manner dependent on the activity of the Raf1 proto-oncogene, serine/threonine-protein kinase C-Raf, but independent of neuronal depolarization. Together, our results provide a mechanism for potentiation of opioid effects by gabapentinoids after injury, via reduction of calcium influx through L-VGCCs, and suggest that other inhibitors targeting these channels may similarly enhance opioid treatment of neuropathic pain.
Collapse
Affiliation(s)
- Anibal Garza-Carbajal
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| | - Alexis Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| | - Juan J. Herrera
- Department of Diagnostic and Interventional Imaging, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| | - Kayla N. Johnson
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| |
Collapse
|
6
|
Page KM, Gumerov VM, Dahimene S, Zhulin IB, Dolphin AC. The importance of cache domains in α 2δ proteins and the basis for their gabapentinoid selectivity. Channels (Austin) 2023; 17:2167563. [PMID: 36735378 PMCID: PMC9901441 DOI: 10.1080/19336950.2023.2167563] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
In this hybrid review, we have first collected and reviewed available information on the structure and function of the enigmatic cache domains in α2δ proteins. These are organized into two double cache (dCache_1) domains, and they are present in all α2δ proteins. We have also included new data on the key function of these domains with respect to amino acid and gabapentinoid binding to the universal amino acid-binding pocket, which is present in α2δ-1 and α2δ-2. We have now identified the reason why α2δ-3 and α2δ-4 do not bind gabapentinoid drugs or amino acids with bulky side chains. In relation to this, we have determined that the bulky amino acids Tryptophan and Phenylalanine prevent gabapentin from inhibiting cell surface trafficking of α2δ-1. Together, these novel data shed further light on the importance of the cache domains in α2δ proteins.
Collapse
Affiliation(s)
- Karen M Page
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Vadim M Gumerov
- Department of Microbiology and Translational Data Analytics Institute, The Ohio State University, Columbus, OH, USA
| | - Shehrazade Dahimene
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Igor B Zhulin
- Department of Microbiology and Translational Data Analytics Institute, The Ohio State University, Columbus, OH, USA
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
- CONTACT Annette C Dolphin Dolphin Department of Neuroscience, Physiology and Pharmacology, University College London, LondonWC1E 6BT, UK
| |
Collapse
|
7
|
Zhang Y, Wang T, Cai Y, Cui T, Kuah M, Vicini S, Wang T. Role of α2δ-3 in regulating calcium channel localization at presynaptic active zones during homeostatic plasticity. Front Mol Neurosci 2023; 16:1253669. [PMID: 38025261 PMCID: PMC10662314 DOI: 10.3389/fnmol.2023.1253669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
The homeostatic modulation of synaptic transmission is an evolutionarily conserved mechanism that is critical for stabilizing the nervous system. At the Drosophila neuromuscular junction (NMJ), presynaptic homeostatic potentiation (PHP) compensates for impairments in postsynaptic glutamate receptors due to pharmacological blockade or genetic deletion. During PHP, there is an increase in presynaptic neurotransmitter release, counteracting postsynaptic changes and restoring excitation to baseline levels. Previous studies have shown that α2δ-3, an auxiliary subunit of voltage-gated calcium channels (VGCCs), is essential for both the rapid induction and sustained expression of PHP at the Drosophila NMJ. However, the molecular mechanisms by which α2δ-3 regulates neurotransmitter release during PHP remain to be elucidated. In this study, we utilized electrophysiological, confocal imaging, and super-resolution imaging approaches to explore how α2δ-3 regulates synaptic transmission during PHP. Our findings suggest that α2δ-3 governs PHP by controlling the localization of the calcium channel pore-forming α1 subunit at presynaptic release sites, or active zones. Moreover, we examined the role of two structural domains within α2δ-3 in regulating neurotransmitter release and calcium channel localization. Our results highlight that these domains in α2δ-3 serve distinct functions in controlling synaptic transmission and presynaptic calcium channel abundance, at baseline in the absence of perturbations and during PHP. In summary, our research offers compelling evidence that α2δ-3 is an indispensable signaling component for controlling calcium channel trafficking and stabilization in homeostatic plasticity.
Collapse
Affiliation(s)
- Yanfeng Zhang
- Department of Pediatric Neurology, First Hospital of Jilin University, Changchun, Jilin, China
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Ting Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Yimei Cai
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Tao Cui
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Michelle Kuah
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Stefano Vicini
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Tingting Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
8
|
Cain SM, Alles SRA, Gopaul R, Bernier LP, Yung AC, Bauman A, Yang Y, Baker GB, Kozlowski P, MacVicar BA, Snutch TP. Chronic pregabalin treatment protects against spreading depolarization and alters hippocampal synaptic characteristics in a model of familial hemiplegic migraine-type 1. Mol Brain 2023; 16:76. [PMID: 37924146 PMCID: PMC10623724 DOI: 10.1186/s13041-023-01062-6] [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: 04/25/2023] [Accepted: 10/04/2023] [Indexed: 11/06/2023] Open
Abstract
Familial hemiplegic migraine type-1 (FHM-1) is a form of migraine with aura caused by mutations in the P/Q-type (Cav2.1) voltage-gated calcium channel. Pregabalin, used clinically in the treatment of chronic pain and epilepsy, inhibits P/Q-type calcium channel activity and recent studies suggest that it may have potential for the treatment of migraine. Spreading Depolarization (SD) is a neurophysiological phenomenon that can occur during migraine with aura by propagating a wave of silenced neuronal function through cortex and sometimes subcortical brain structures. Here, utilizing an optogenetic stimulation technique optimized to allow for non-invasive initiation of cortical SD, we demonstrate that chronic pregabalin administration [12 mg/kg/day (s.c.)] in vivo increased the threshold for cortical spreading depolarization in transgenic mice harboring the clinically-relevant Cav2.1S218L mutation (S218L). In addition, chronic pregabalin treatment limited subcortical propagation of recurrent spreading depolarization events to the striatum and hippocampus in both wild-type and S218L mice. To examine contributing underlying mechanisms of action of chronic pregabalin, we performed whole-cell patch-clamp electrophysiology in CA1 neurons in ex vivo brain slices from mice treated with chronic pregabalin vs vehicle. In WT mice, chronic pregabalin produced a decrease in spontaneous excitatory postsynaptic current (sEPSC) amplitude with no effect on frequency. In contrast, in S218L mice chronic pregabalin produced an increase in sEPSC amplitude and decreased frequency. These electrophysiological findings suggest that in FHM-1 mice chronic pregabalin acts through both pre- and post-synaptic mechanisms in CA1 hippocampal neurons to elicit FHM-1 genotype-specific inhibitory action. The results highlight the potential of chronic pregabalin to limit recurrent SD to subcortical brain structures during pathophysiological events in both the genetically-normal and FHM-1 brain. The work further provides insights into FHM-1 pathophysiology and the potential for chronic pregabalin treatment to prevent SD in migraineurs.
Collapse
Affiliation(s)
- Stuart M Cain
- Michael Smith Laboratories, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, Canada
| | - Sascha R A Alles
- Michael Smith Laboratories, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Ray Gopaul
- Michael Smith Laboratories, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, Canada
| | - Louis-Philippe Bernier
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, Canada
| | - Andrew C Yung
- UBC MRI Research Facility, University of British Columbia, Vancouver, Canada
| | - Andrew Bauman
- UBC MRI Research Facility, University of British Columbia, Vancouver, Canada
| | - Yi Yang
- Michael Smith Laboratories, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, Canada
| | - Glen B Baker
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Canada
| | - Piotr Kozlowski
- UBC MRI Research Facility, University of British Columbia, Vancouver, Canada
| | - Brian A MacVicar
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, Canada
| | - Terrance P Snutch
- Michael Smith Laboratories, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada.
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
9
|
Rissardo JP, Medeiros Araujo de Matos U, Fornari Caprara AL. Gabapentin-Associated Movement Disorders: A Literature Review. MEDICINES (BASEL, SWITZERLAND) 2023; 10:52. [PMID: 37755242 PMCID: PMC10536490 DOI: 10.3390/medicines10090052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Gabapentin (GBP)-induced movement disorders (MDs) are under-recognized adverse drug reactions. They are commonly not discussed with patients, and their sudden occurrence can lead to misdiagnosis. This literature review aims to evaluate the clinical-epidemiological profile, pathological mechanisms, and management of GBP-associated MD. METHODS Two reviewers identified and assessed relevant reports in six databases without language restriction between 1990 and 2023. RESULTS A total of 99 reports of 204 individuals who developed a MD associated with GBP were identified. The MDs encountered were 135 myoclonus, 22 dyskinesias, 7 dystonia, 3 akathisia, 3 stutterings, 1 myokymia, and 1 parkinsonism. The mean and median ages were 54.54 (SD: 17.79) and 57 years (age range: 10-89), respectively. Subjects were predominantly male (53.57%). The mean and median doses of GBP when the MD occurred were 1324.66 (SD: 1117.66) and 1033 mg/daily (GBP dose range: 100-9600), respectively. The mean time from GBP-onset to GBP-associated MD was 4.58 weeks (SD: 8.08). The mean recovery time after MD treatment was 4.17 days (SD: 4.87). The MD management involved GBP discontinuation. A total of 82.5% of the individuals had a full recovery in the follow-up period. CONCLUSIONS Myoclonus (GRADE A) and dyskinesia (GRADE C) were the most common movement disorders associated with GBP.
Collapse
|
10
|
Oh KH, Xiong A, Choe JY, Richmond JE, Kim H. Active Zone Trafficking of CaV2/UNC-2 Channels Is Independent of β/CCB-1 and α2δ/UNC-36 Subunits. J Neurosci 2023; 43:5142-5157. [PMID: 37160370 PMCID: PMC10343168 DOI: 10.1523/jneurosci.2264-22.2023] [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: 12/09/2022] [Revised: 03/28/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023] Open
Abstract
The CaV2 voltage-gated calcium channel is the major conduit of calcium ions necessary for neurotransmitter release at presynaptic active zones (AZs). The CaV2 channel is a multimeric complex that consists of a pore-forming α1 subunit and two auxiliary β and α2δ subunits. Although auxiliary subunits are critical for channel function, whether they are required for α1 trafficking is unresolved. Using endogenously fluorescent protein-tagged CaV2 channel subunits in Caenorhabditis elegans, we show that UNC-2/α1 localizes to AZs even in the absence of CCB-1/β or UNC-36/α2δ, albeit at low levels. When UNC-2 is manipulated to be trapped in the endoplasmic reticulum (ER), CCB-1 and UNC-36 fail to colocalize with UNC-2 in the ER, indicating that they do not coassemble with UNC-2 in the ER. Moreover, blocking ER-associated degradation does not further increase presynaptic UNC-2 channels in ccb-1 or unc-36 mutants, indicating that UNC-2 levels are not regulated in the ER. An unc-2 mutant lacking C-terminal AZ protein interaction sites with intact auxiliary subunit binding sites displays persistent presynaptic UNC-2 localization and a prominent increase of UNC-2 channels in nonsynaptic axonal regions, underscoring a protective role of auxiliary subunits against UNC-2 degradation. In the absence of UNC-2, presynaptic CCB-1 and UNC-36 are profoundly diminished to barely detectable levels, indicating that UNC-2 is required for the presynaptic localization of CCB-1 and UNC-36. Together, our findings demonstrate that although the pore-forming subunit does not require auxiliary subunits for its trafficking and transport to AZs, it recruits auxiliary subunits to stabilize and expand calcium channel signalosomes.SIGNIFICANCE STATEMENT Synaptic transmission in the neuron hinges on the coupling of synaptic vesicle exocytosis with calcium influx. This calcium influx is mediated by CaV2 voltage-gated calcium channels. These channels consist of one pore-forming α1 subunit and two auxiliary β and α2δ subunits. The auxiliary subunits enhance channel function and regulate the overall level of channels at presynaptic terminals. However, it is not settled how these auxiliary subunits regulate the overall channel level. Our study in C. elegans finds that although the auxiliary subunits do not coassemble with α1 and aid trafficking, they are recruited to α1 and stabilize the channel complex at presynaptic terminals. Our study suggests that drugs that target the auxiliary subunits can directly destabilize and have an impact on CaV2 channels.
Collapse
Affiliation(s)
- Kelly H Oh
- Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| | - Ame Xiong
- Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| | - Jun-Yong Choe
- Department of Chemistry, East Carolina University, Greenville, North Carolina 27858
| | - Janet E Richmond
- Department of Biological Sciences, University of Illinois, Chicago, Illinois 60607
| | - Hongkyun Kim
- Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| |
Collapse
|
11
|
Ferron L, Gandini MA, Zamponi GW. Fighting pain: the structure of gabapentin and its binding site in the Ca vα 2δ subunit. Nat Struct Mol Biol 2023:10.1038/s41594-023-01013-8. [PMID: 37286823 DOI: 10.1038/s41594-023-01013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Affiliation(s)
- Laurent Ferron
- Department of Clinical Neuroscience, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Maria A Gandini
- Department of Clinical Neuroscience, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Gerald W Zamponi
- Department of Clinical Neuroscience, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
12
|
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: 5.5] [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
|
13
|
Tyagi S, Sarveswaran N, Higerd-Rusli GP, Liu S, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. Conserved but not critical: Trafficking and function of NaV1.7 are independent of highly conserved polybasic motifs. Front Mol Neurosci 2023; 16:1161028. [PMID: 37008789 PMCID: PMC10060856 DOI: 10.3389/fnmol.2023.1161028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Non-addictive treatment of chronic pain represents a major unmet clinical need. Peripheral voltage-gated sodium (NaV) channels are an attractive target for pain therapy because they initiate and propagate action potentials in primary afferents that detect and transduce noxious stimuli. NaV1.7 sets the gain on peripheral pain-signaling neurons and is the best validated peripheral ion channel involved in human pain, and previous work has shown that it is transported in vesicles in sensory axons which also carry Rab6a, a small GTPase known to be involved in vesicular packaging and axonal transport. Understanding the mechanism of the association between Rab6a and NaV1.7 could inform therapeutic modalities to decrease trafficking of NaV1.7 to the distal axonal membrane. Polybasic motifs (PBM) have been shown to regulate Rab-protein interactions in a variety of contexts. In this study, we explored whether two PBMs in the cytoplasmic loop that joins domains I and II of human NaV1.7 were responsible for association with Rab6a and regulate axonal trafficking of the channel. Using site-directed mutagenesis we generated NaV1.7 constructs with alanine substitutions in the two PBMs. Voltage-clamp recordings showed that the constructs retain wild-type like gating properties. Optical Pulse-chase Axonal Long-distance (OPAL) imaging in live sensory axons shows that mutations of these PBMs do not affect co-trafficking of Rab6a and NaV1.7, or the accumulation of the channel at the distal axonal surface. Thus, these polybasic motifs are not required for interaction of NaV1.7 with the Rab6a GTPase, or for trafficking of the channel to the plasma membrane.
Collapse
Affiliation(s)
- Sidharth Tyagi
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Nivedita Sarveswaran
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Grant P. Higerd-Rusli
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Shujun Liu
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Fadia B. Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Stephen G. Waxman
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
- *Correspondence: Stephen G. Waxman,
| | - Sulayman D. Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, United States
- Sulayman D. Dib-Hajj,
| |
Collapse
|
14
|
Aneja J, Singh J. Pregabalin Use/Misuse: A Source of Consternation in Western Punjab. Indian J Psychol Med 2023; 45:206-207. [PMID: 36925504 PMCID: PMC10011841 DOI: 10.1177/02537176221122389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Jitender Aneja
- Dept. of Psychiatry, All India Institute of Medical Sciences, Bathinda, Punjab, India
| | - Jawahar Singh
- Dept. of Psychiatry, All India Institute of Medical Sciences, Bathinda, Punjab, India
| |
Collapse
|
15
|
Warlick H, Leon L, Patel R, Filoramo S, Knipe R, Joubran E, Levy A, Nguyen H, Rey J. Application of gabapentinoids and novel compounds for the treatment of benzodiazepine dependence: the glutamatergic model. Mol Biol Rep 2023; 50:1765-1784. [PMID: 36456769 DOI: 10.1007/s11033-022-08110-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 11/09/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND Current approaches for managing benzodiazepine (BZD) withdrawal symptoms are daunting for clinicians and patients, warranting novel treatment and management strategies. This review discusses the pharmacodynamic properties of BZDs, gabapentinoids (GBPs), endozepines, and novel GABAergic compounds associated with potential clinical benefits for BZD-dependent patients. The objective of this study was to review the complex neuromolecular changes occurring within the GABAergic and glutamatergic systems during the BZD tolerance and withdrawal periods while also examining the mechanism by which GBPs and alternative pharmacological therapies may attenuate withdrawal symptoms. METHODS AND RESULTS An elaborative literature review was conducted using multiple platforms, including the National Center for Biotechnology (NCBI), AccessMedicine, ScienceDirect, pharmacology textbooks, clinical trial data, case reports, and PubChem. Our literature analysis revealed that many distinctive neuroadaptive mechanisms are involved in the GABAergic and glutamatergic systems during BZD tolerance and withdrawal. Based on this data, we hypothesize that GBPs may attenuate the overactive glutamatergic system during the withdrawal phase by an indirect presynaptic glutamatergic mechanism dependent on the α2δ1 subunit expression. CONCLUSIONS GBPs may benefit individuals undergoing BZD withdrawal, given that the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor current significantly increases during abrupt BZD withdrawal in animal studies. This may be a conceivable explanation for the effectiveness of GBPs in treating both alcohol withdrawal symptoms and BZD withdrawal symptoms in some recent studies. Finally, natural and synthetic GABAergic compounds with unique pharmacodynamic properties were found to exert potential clinical benefits as BZD substitutes in animal studies, though human studies are lacking.
Collapse
Affiliation(s)
- Halford Warlick
- Dr. Kiran C. Patel College Of Osteopathic Medicine, Nova Southeastern University, Davie, FL, USA.
| | - Lexie Leon
- Dr. Kiran C. Patel College Of Osteopathic Medicine, Nova Southeastern University, Davie, FL, USA
| | - Rudresh Patel
- Dr. Kiran C. Patel College Of Osteopathic Medicine, Nova Southeastern University, Davie, FL, USA
| | - Stefanie Filoramo
- Dr. Kiran C. Patel College Of Osteopathic Medicine, Nova Southeastern University, Davie, FL, USA
| | - Ryan Knipe
- Dr. Kiran C. Patel College Of Osteopathic Medicine, Nova Southeastern University, Davie, FL, USA
| | - Ernesto Joubran
- Dr. Kiran C. Patel College Of Osteopathic Medicine, Nova Southeastern University, Davie, FL, USA
| | - Arkene Levy
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, FL, USA
| | - Hoang Nguyen
- Dr. Kiran C. Patel College Of Osteopathic Medicine, Nova Southeastern University, Davie, FL, USA
| | - Jose Rey
- College of Pharmacy, Nova Southeastern University, Davie, FL, USA
| |
Collapse
|
16
|
Higerd-Rusli GP, Tyagi S, Liu S, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. The fates of internalized Na V1.7 channels in sensory neurons: Retrograde cotransport with other ion channels, axon-specific recycling, and degradation. J Biol Chem 2023; 299:102816. [PMID: 36539035 PMCID: PMC9843449 DOI: 10.1016/j.jbc.2022.102816] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 12/05/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
Neuronal function relies on the maintenance of appropriate levels of various ion channels at the cell membrane, which is accomplished by balancing secretory, degradative, and recycling pathways. Neuronal function further depends on membrane specialization through polarized distribution of specific proteins to distinct neuronal compartments such as axons. Voltage-gated sodium channel NaV1.7, a threshold channel for firing action potentials in nociceptors, plays a major role in human pain, and its abundance in the plasma membrane is tightly regulated. We have recently characterized the anterograde axonal trafficking of NaV1.7 channels in Rab6A-positive vesicles, but the fate of internalized channels is not known. Membrane proteins that have undergone endocytosis can be directed into multiple pathways including those for degradation, recycling to the membrane, and transcytosis. Here, we demonstrate NaV1.7 endocytosis and dynein-dependent retrograde trafficking in Rab7-containing late endosomes together with other axonal membrane proteins using real-time imaging of live neurons. We show that some internalized NaV1.7 channels are delivered to lysosomes within the cell body, and that there is no evidence for NaV1.7 transcytosis. In addition, we show that NaV1.7 is recycled specifically to the axonal membrane as opposed to the soma membrane, suggesting a novel mechanism for the development of neuronal polarity. Together, these results shed light on the mechanisms by which neurons maintain excitable membranes and may inform efforts to target ion channel trafficking for the treatment of disorders of excitability.
Collapse
Affiliation(s)
- Grant P Higerd-Rusli
- MD/PhD Program, Yale University School of Medicine, New Haven, Connecticut, USA; Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Sidharth Tyagi
- MD/PhD Program, Yale University School of Medicine, New Haven, Connecticut, USA; Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Shujun Liu
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Fadia B Dib-Hajj
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA.
| | - Sulayman D Dib-Hajj
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA.
| |
Collapse
|
17
|
Hessenberger M, Haddad S, Obermair GJ. Pathophysiological Roles of Auxiliary Calcium Channel α 2δ Subunits. Handb Exp Pharmacol 2023; 279:289-316. [PMID: 36598609 DOI: 10.1007/164_2022_630] [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] [Indexed: 01/05/2023]
Abstract
α2δ proteins serve as auxiliary subunits of voltage-gated calcium channels, which are essential components of excitable cells such as skeletal and heart muscles, nerve cells of the brain and the peripheral nervous system, as well as endocrine cells. Over the recent years, α2δ proteins have been identified as critical regulators of synaptic functions, including the formation and differentiation of synapses. These functions require signalling mechanisms which are partly independent of calcium channels. Hence, in light of these features it is not surprising that the genes encoding for the four α2δ isoforms have recently been linked to neurological and neurodevelopmental disorders including epilepsy, autism spectrum disorders, schizophrenia, and depressive and bipolar disorders. Despite the increasing number of identified disease-associated mutations, the underlying pathophysiological mechanisms are only beginning to emerge. However, a thorough understanding of the pathophysiological role of α2δ proteins ideally serves two purposes: first, it will contribute to our understanding of general pathological mechanisms in synaptic disorders. Second, it may support the future development of novel and specific treatments for brain disorders. In this context, it is noteworthy that the antiepileptic and anti-allodynic drugs gabapentin and pregabalin both act via binding to α2δ proteins and are among the top sold drugs for treating neuropathic pain. In this book chapter, we will discuss recent developments in our understanding of the functions of α2δ proteins, both as calcium channel subunits and as independent regulatory entities. Furthermore, we present and summarize recently identified and likely pathogenic mutations in the genes encoding α2δ proteins and discuss potential underlying pathophysiological consequences at the molecular and structural level.
Collapse
Affiliation(s)
- Manuel Hessenberger
- Division Physiology, Department Pharmacology, Physiology and Microbiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Sabrin Haddad
- Division Physiology, Department Pharmacology, Physiology and Microbiology, Karl Landsteiner University of Health Sciences, Krems, Austria
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gerald J Obermair
- Division Physiology, Department Pharmacology, Physiology and Microbiology, Karl Landsteiner University of Health Sciences, Krems, Austria.
| |
Collapse
|
18
|
Abstract
Vesicles mediate the trafficking of membranes/proteins in the endocytic and secretory pathways. These pathways are regulated by small GTPases of the Rab family. Rab proteins belong to the Ras superfamily of GTPases, which are significantly involved in various intracellular trafficking and signaling processes in the nervous system. Rab11 is known to play a key role especially in recycling many proteins, including receptors important for signal transduction and preservation of functional activities of nerve cells. Rab11 activity is controlled by GEFs (guanine exchange factors) and GAPs (GTPase activating proteins), which regulate its function through modulating GTP/GDP exchange and the intrinsic GTPase activity, respectively. Rab11 is involved in the transport of several growth factor molecules important for the development and repair of neurons. Overexpression of Rab11 has been shown to significantly enhance vesicle trafficking. On the other hand, a reduced expression of Rab11 was observed in several neurodegenerative diseases. Current evidence appears to support the notion that Rab11 and its cognate proteins may be potential targets for therapeutic intervention. In this review, we briefly discuss the function of Rab11 and its related interaction partners in intracellular pathways that may be involved in neurodegenerative processes.
Collapse
Affiliation(s)
| | - Jiri Novotny
- Jiri Novotny, Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic.
| |
Collapse
|
19
|
Ramgoolam KH, Dolphin AC. Capsaicin-Induced Endocytosis of Endogenous Presynaptic Ca V2.2 in DRG-Spinal Cord Co-Cultures Inhibits Presynaptic Function. FUNCTION 2022; 4:zqac058. [PMID: 36540890 PMCID: PMC9761886 DOI: 10.1093/function/zqac058] [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: 10/03/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 11/26/2022] Open
Abstract
The N-type calcium channel, CaV2.2 is key to neurotransmission from the primary afferent terminals of dorsal root ganglion (DRG) neurons to their postsynaptic targets in the spinal cord. In this study, we have utilized CaV2.2_HA knock-in mice, because the exofacial epitope tag in CaV2.2_HA enables accurate detection and localization of endogenous CaV2.2. CaV2.2_HA knock-in mice were used as a source of DRGs to exclusively study the presynaptic expression of N-type calcium channels in co-cultures between DRG neurons and wild-type spinal cord neurons. CaV2.2_HA is strongly expressed on the cell surface, particularly in TRPV1-positive small and medium DRG neurons. Super-resolution images of the presynaptic terminals revealed an increase in CaV2.2_HA expression and increased association with the postsynaptic marker Homer over time in vitro. Brief application of the TRPV1 agonist, capsaicin, resulted in a significant down-regulation of cell surface CaV2.2_HA expression in DRG neuron somata. At their presynaptic terminals, capsaicin caused a reduction in CaV2.2_HA proximity to and co-localization with the active zone marker RIM 1/2, as well as a lower contribution of N-type channels to single action potential-mediated Ca2+ influx. The mechanism of this down-regulation of CaV2.2_HA involves a Rab11a-dependent trafficking process, since dominant-negative Rab11a (S25N) occludes the effect of capsaicin on presynaptic CaV2.2_HA expression, and also prevents the effect of capsaicin on action potential-induced Ca2+ influx. Taken together, these data suggest that capsaicin causes a decrease in cell surface CaV2.2_HA expression in DRG terminals via a Rab11a-dependent endosomal trafficking pathway.
Collapse
Affiliation(s)
- Krishma H Ramgoolam
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| |
Collapse
|
20
|
Pellegrino M, Ricci E, Ceraldi R, Nigro A, Bonofiglio D, Lanzino M, Morelli C. From HDAC to Voltage-Gated Ion Channels: What's Next? The Long Road of Antiepileptic Drugs Repositioning in Cancer. Cancers (Basel) 2022; 14:cancers14184401. [PMID: 36139561 PMCID: PMC9497059 DOI: 10.3390/cancers14184401] [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: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Although in the last decades the clinical outcome of cancer patients considerably improved, the major drawbacks still associated with chemotherapy are the unwanted side effects and the development of drug resistance. Therefore, a continuous effort in trying to discover new tumor markers, possibly of diagnostic, prognostic and therapeutic value, is being made. This review is aimed at highlighting the anti-tumor activity that several antiepileptic drugs (AEDs) exert in breast, prostate and other types of cancers, mainly focusing on their ability to block the voltage-gated Na+ and Ca++ channels, as well as to inhibit the activity of histone deacetylases (HDACs), all well-documented tumor markers and/or molecular targets. The existence of additional AEDs molecular targets is highly suspected. Therefore, the repurposing of already available drugs as adjuvants in cancer treatment would have several advantages, such as reductions in dose-related toxicity CVs will be sent in a separate mail to the indicated address of combined treatments, lower production costs, and faster approval for clinical use. Abstract Cancer is a major health burden worldwide. Although the plethora of molecular targets identified in the last decades and the deriving developed treatments, which significantly improved patients’ outcome, the occurrence of resistance to therapies remains the major cause of relapse and mortality. Thus, efforts in identifying new markers to be exploited as molecular targets in cancer therapy are needed. This review will first give a glance on the diagnostic and therapeutic significance of histone deacetylase (HDAC) and voltage gated ion channels (VGICs) in cancer. Nevertheless, HDAC and VGICs have also been reported as molecular targets through which antiepileptic drugs (AEDs) seem to exert their anticancer activity. This should be claimed as a great advantage. Indeed, due to the slowness of drug approval procedures, the attempt to turn to off-label use of already approved medicines would be highly preferable. Therefore, an updated and accurate overview of both preclinical and clinical data of commonly prescribed AEDs (mainly valproic acid, lamotrigine, carbamazepine, phenytoin and gabapentin) in breast, prostate, brain and other cancers will follow. Finally, a glance at the emerging attempt to administer AEDs by means of opportunely designed drug delivery systems (DDSs), so to limit toxicity and improve bioavailability, is also given.
Collapse
Affiliation(s)
| | | | | | | | | | - Marilena Lanzino
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| | - Catia Morelli
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| |
Collapse
|
21
|
Hong JSW, Atkinson LZ, Al-Juffali N, Awad A, Geddes JR, Tunbridge EM, Harrison PJ, Cipriani A. Gabapentin and pregabalin in bipolar disorder, anxiety states, and insomnia: Systematic review, meta-analysis, and rationale. Mol Psychiatry 2022; 27:1339-1349. [PMID: 34819636 PMCID: PMC9095464 DOI: 10.1038/s41380-021-01386-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/29/2021] [Accepted: 10/27/2021] [Indexed: 12/29/2022]
Abstract
The gabapentinoids, gabapentin, and pregabalin, target the α2δ subunits of voltage-gated calcium channels. Initially licensed for pain and seizures, they have become widely prescribed drugs. Many of these uses are off-label for psychiatric indications, and there is increasing concern about their safety, so it is particularly important to have good evidence to justify this usage. We conducted a systematic review and meta-analysis of the evidence for three of their common psychiatric uses: bipolar disorder, anxiety, and insomnia. Fifty-five double-blind randomised controlled trials (RCTs) and 15 open-label studies were identified. For bipolar disorder, four double-blind RCTs investigating gabapentin, and no double-blind RCTs investigating pregabalin, were identified. A quantitative synthesis could not be performed due to heterogeneity in the study population, design and outcome measures. Across the anxiety spectrum, a consistent but not universal effect in favour of gabapentinoids compared to placebo was seen (standardised mean difference [SMD] ranging between -2.25 and -0.25). Notably, pregabalin (SMD -0.55, 95% CI -0.92 to -0.18) and gabapentin (SMD -0.92, 95% CI -1.32 to -0.52) were more effective than placebo in reducing preoperative anxiety. In insomnia, results were inconclusive. We conclude that there is moderate evidence of the efficacy of gabapentinoids in anxiety states, but minimal evidence in bipolar disorder and insomnia and they should be used for these disorders only with strong justification. This recommendation applies despite the attractive pharmacological and genetic rationale for targeting voltage-gated calcium channels.
Collapse
Affiliation(s)
- James S. W. Hong
- grid.416938.10000 0004 0641 5119Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX UK
| | - Lauren Z. Atkinson
- grid.416938.10000 0004 0641 5119Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX UK ,grid.497865.10000 0004 0427 1035Oxford Centre for Human Brain Activity, University of Oxford, Oxford, UK ,grid.416938.10000 0004 0641 5119Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Noura Al-Juffali
- grid.416938.10000 0004 0641 5119Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX UK ,grid.416938.10000 0004 0641 5119Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Amine Awad
- grid.416938.10000 0004 0641 5119Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX UK ,grid.32224.350000 0004 0386 9924Department of Neurology, Massachusetts General Hospital, Boston, MA 02114 USA
| | - John R. Geddes
- grid.416938.10000 0004 0641 5119Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX UK ,grid.416938.10000 0004 0641 5119Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Elizabeth M. Tunbridge
- grid.416938.10000 0004 0641 5119Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX UK ,grid.416938.10000 0004 0641 5119Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Paul J. Harrison
- grid.416938.10000 0004 0641 5119Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX UK ,grid.416938.10000 0004 0641 5119Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Andrea Cipriani
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK. .,Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK.
| |
Collapse
|
22
|
Beeson KA, Westbrook GL, Schnell E. α2δ-2 is required for depolarization-induced suppression of excitation in Purkinje cells. J Physiol 2022; 600:111-122. [PMID: 34783012 PMCID: PMC8724408 DOI: 10.1113/jp282438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/11/2021] [Indexed: 01/03/2023] Open
Abstract
α2δ proteins (CACNA2D1-4) are required for normal neurological function and contribute to membrane trafficking of voltage-gated calcium channels, through which calcium entry initiates numerous physiological processes. However, it remains unclear how α2δ proteins influence calcium-mediated signalling to control neuronal output. Using whole-cell recordings of mouse Purkinje cells, we show that α2δ-2 is required for functional coupling of postsynaptic voltage-dependent calcium entry with calcium-dependent effector mechanisms controlling two different outputs, depolarization-induced suppression of excitation and spike afterhyperpolarization. Our findings indicate an important role for α2δ-2 proteins in regulating functional postsynaptic calcium channel coupling in neurons, providing new context for understanding the effects of α2δ mutations on neuronal circuit function and presenting additional potential avenues to manipulate α2δ-mediated signalling for therapeutic gain. KEY POINTS: Calcium influx, via voltage-dependent calcium channels, drives numerous neuronal signalling processes with precision achieved in part by tight coupling between calcium entry and calcium-dependent effectors. α2δ proteins are important for neurological function and contribute to calcium channel membrane trafficking, although how α2δ proteins influence postsynaptic calcium-dependent signalling is largely unexplored. Here it is shown that loss of α2δ-2 proteins disrupts functional calcium coupling to two different postsynaptic calcium-dependent signals in mouse Purkinje cell neurons, retrograde endocannabinoid signalling and the action potential afterhyperpolarization. The findings provide new insights into the control of calcium coupling as well as new roles for α2δ-2 proteins in neurons.
Collapse
Affiliation(s)
- Kathleen A. Beeson
- Neuroscience Graduate Program, OHSU, Portland, OR, 97239,Department of Anesthesiology and Perioperative Medicine, OHSU, Portland, OR, 97239
| | | | - Eric Schnell
- Department of Anesthesiology and Perioperative Medicine, OHSU, Portland, OR, 97239,Operative Care Division, Portland VA Health Care System, Portland, OR, 97239,Eric Schnell, MD, PhD,
| |
Collapse
|
23
|
Abstract
Managing chronic pain remains a major unmet clinical challenge. Patients can be treated with a range of interventions, but pharmacotherapy is the most common. These include opioids, antidepressants, calcium channel modulators, sodium channel blockers, and nonsteroidal anti-inflammatory drugs. Many of these drugs target a particular mechanism; however, chronic pain in many diseases is multifactorial and induces plasticity throughout the sensory neuroaxis. Furthermore, comorbidities such as depression, anxiety, and sleep disturbances worsen quality of life. Given the complexity of mechanisms and symptoms in patients, it is unsurprising that many fail to achieve adequate pain relief from a single agent. The efforts to develop novel drug classes with better efficacy have not always proved successful; a multimodal or combination approach to analgesia is an important strategy in pain control. Many patients frequently take more than one medication, but high-quality evidence to support various combinations is often sparse. Ideally, combining drugs would produce synergistic action to maximize analgesia and reduce side effects, although sub-additive and additive analgesia is still advantageous if additive side-effects can be avoided. In this review, we discuss pain mechanisms, drug actions, and the rationale for mechanism-led treatment selection.Abbreviations: COX - cyclooxygenase, CGRP - calcitonin gene-related peptide, CPM - conditioned pain modulation, NGF - nerve growth factor, NNT - number needed to treat, NMDA - N-methyl-d-aspartate, NSAID - nonsteroidal anti-inflammatory drugs, TCA - tricyclic antidepressant, SNRI - serotonin-noradrenaline reuptake inhibitor, QST - quantitative sensory testing.
Collapse
Affiliation(s)
- Ryan Patel
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - Anthony H Dickenson
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| |
Collapse
|
24
|
Tuluc P, Theiner T, Jacobo-Piqueras N, Geisler SM. Role of High Voltage-Gated Ca 2+ Channel Subunits in Pancreatic β-Cell Insulin Release. From Structure to Function. Cells 2021; 10:2004. [PMID: 34440773 PMCID: PMC8393260 DOI: 10.3390/cells10082004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
The pancreatic islets of Langerhans secrete several hormones critical for glucose homeostasis. The β-cells, the major cellular component of the pancreatic islets, secrete insulin, the only hormone capable of lowering the plasma glucose concentration. The counter-regulatory hormone glucagon is secreted by the α-cells while δ-cells secrete somatostatin that via paracrine mechanisms regulates the α- and β-cell activity. These three peptide hormones are packed into secretory granules that are released through exocytosis following a local increase in intracellular Ca2+ concentration. The high voltage-gated Ca2+ channels (HVCCs) occupy a central role in pancreatic hormone release both as a source of Ca2+ required for excitation-secretion coupling as well as a scaffold for the release machinery. HVCCs are multi-protein complexes composed of the main pore-forming transmembrane α1 and the auxiliary intracellular β, extracellular α2δ, and transmembrane γ subunits. Here, we review the current understanding regarding the role of all HVCC subunits expressed in pancreatic β-cell on electrical activity, excitation-secretion coupling, and β-cell mass. The evidence we review was obtained from many seminal studies employing pharmacological approaches as well as genetically modified mouse models. The significance for diabetes in humans is discussed in the context of genetic variations in the genes encoding for the HVCC subunits.
Collapse
Affiliation(s)
- Petronel Tuluc
- Centre for Molecular Biosciences, Department of Pharmacology and Toxicology, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (T.T.); (N.J.-P.); (S.M.G.)
| | | | | | | |
Collapse
|
25
|
Meyer JO, Dolphin AC. Rab11-dependent recycling of calcium channels is mediated by auxiliary subunit α 2δ-1 but not α 2δ-3. Sci Rep 2021; 11:10256. [PMID: 33986433 PMCID: PMC8119971 DOI: 10.1038/s41598-021-89820-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/16/2021] [Indexed: 11/18/2022] Open
Abstract
N-type voltage-gated calcium channels (CaV2.2) are predominantly expressed at presynaptic terminals, and their function is regulated by auxiliary α2δ and β subunits. All four mammalian α2δ subunits enhance calcium currents through CaV1 and CaV2 channels, and this increase is attributed, in part, to increased CaV expression at the plasma membrane. In the present study we provide evidence that α2δ-1, like α2δ-2, is recycled to the plasma membrane through a Rab11a-dependent endosomal recycling pathway. Using a dominant-negative Rab11a mutant, Rab11a(S25N), we show that α2δ-1 increases plasma membrane CaV2.2 expression by increasing the rate and extent of net forward CaV2.2 trafficking in a Rab11a-dependent manner. Dominant-negative Rab11a also reduces the ability of α2δ-1 to increase CaV2.2 expression on the cell-surface of hippocampal neurites. In contrast, α2δ-3 does not enhance rapid forward CaV2.2 trafficking, regardless of whether Rab11a(S25N) is present. In addition, whole-cell CaV2.2 currents are reduced by co-expression of Rab11a(S25N) in the presence of α2δ-1, but not α2δ-3. Taken together these data suggest that α2δ subtypes participate in distinct trafficking pathways which in turn influence the localisation and function of CaV2.2.
Collapse
Affiliation(s)
- James O Meyer
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
26
|
Wright CS, Robling AG, Farach-Carson MC, Thompson WR. Skeletal Functions of Voltage Sensitive Calcium Channels. Curr Osteoporos Rep 2021; 19:206-221. [PMID: 33721180 PMCID: PMC8216424 DOI: 10.1007/s11914-020-00647-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 12/15/2022]
Abstract
Voltage-sensitive calcium channels (VSCCs) are ubiquitous multimeric protein complexes that are necessary for the regulation of numerous physiological processes. VSCCs regulate calcium influx and various intracellular processes including muscle contraction, neurotransmission, hormone secretion, and gene transcription, with function specificity defined by the channel's subunits and tissue location. The functions of VSCCs in bone are often overlooked since bone is not considered an electrically excitable tissue. However, skeletal homeostasis and adaptation relies heavily on VSCCs. Inhibition or deletion of VSCCs decreases osteogenesis, impairs skeletal structure, and impedes anabolic responses to mechanical loading. RECENT FINDINGS: While the functions of VSCCs in osteoclasts are less clear, VSCCs have distinct but complementary functions in osteoblasts and osteocytes. PURPOSE OF REVIEW: This review details the structure, function, and nomenclature of VSCCs, followed by a comprehensive description of the known functions of VSCCs in bone cells and their regulation of bone development, bone formation, and mechanotransduction.
Collapse
Affiliation(s)
- Christian S Wright
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Alexander G Robling
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
- Department of Anatomy & Cell Biology, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic & Biomedical Sciences, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, 77054, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy & Cell Biology, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
27
|
Ferron L, Koshti S, Zamponi GW. The life cycle of voltage-gated Ca 2+ channels in neurons: an update on the trafficking of neuronal calcium channels. Neuronal Signal 2021; 5:NS20200095. [PMID: 33664982 PMCID: PMC7905535 DOI: 10.1042/ns20200095] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/15/2021] [Indexed: 01/26/2023] Open
Abstract
Neuronal voltage-gated Ca2+ (CaV) channels play a critical role in cellular excitability, synaptic transmission, excitation-transcription coupling and activation of intracellular signaling pathways. CaV channels are multiprotein complexes and their functional expression in the plasma membrane involves finely tuned mechanisms, including forward trafficking from the endoplasmic reticulum (ER) to the plasma membrane, endocytosis and recycling. Whether genetic or acquired, alterations and defects in the trafficking of neuronal CaV channels can have severe physiological consequences. In this review, we address the current evidence concerning the regulatory mechanisms which underlie precise control of neuronal CaV channel trafficking and we discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Laurent Ferron
- Department of Physiology and Pharmacology, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Saloni Koshti
- Department of Physiology and Pharmacology, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Gerald W. Zamponi
- Department of Physiology and Pharmacology, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
28
|
Studies on CRMP2 SUMOylation-deficient transgenic mice identify sex-specific Nav1.7 regulation in the pathogenesis of chronic neuropathic pain. Pain 2021; 161:2629-2651. [PMID: 32569093 DOI: 10.1097/j.pain.0000000000001951] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The sodium channel Nav1.7 is a master regulator of nociceptive input into the central nervous system. Mutations in this channel can result in painful conditions and produce insensitivity to pain. Despite being recognized as a "poster child" for nociceptive signaling and human pain, targeting Nav1.7 has not yet produced a clinical drug. Recent work has illuminated the Nav1.7 interactome, offering insights into the regulation of these channels and identifying potentially new druggable targets. Among the regulators of Nav1.7 is the cytosolic collapsin response mediator protein 2 (CRMP2). CRMP2, modified at lysine 374 (K374) by addition of a small ubiquitin-like modifier (SUMO), bound Nav1.7 to regulate its membrane localization and function. Corollary to this, preventing CRMP2 SUMOylation was sufficient to reverse mechanical allodynia in rats with neuropathic pain. Notably, loss of CRMP2 SUMOylation did not compromise other innate functions of CRMP2. To further elucidate the in vivo role of CRMP2 SUMOylation in pain, we generated CRMP2 K374A knock-in (CRMP2) mice in which Lys374 was replaced with Ala. CRMP2 mice had reduced Nav1.7 membrane localization and function in female, but not male, sensory neurons. Behavioral appraisal of CRMP2 mice demonstrated no changes in depressive or repetitive, compulsive-like behaviors and a decrease in noxious thermal sensitivity. No changes were observed in CRMP2 mice to inflammatory, acute, or visceral pain. By contrast, in a neuropathic model, CRMP2 mice failed to develop persistent mechanical allodynia. Our study suggests that CRMP2 SUMOylation-dependent control of peripheral Nav1.7 is a hallmark of chronic, but not physiological, neuropathic pain.
Collapse
|
29
|
How Postdoctoral Research in Paul Greengard's Laboratory Shaped My Scientific Career, Although I Never Did Another Phosphorylation Assay. J Neurosci 2021; 41:2070-2075. [PMID: 33558431 PMCID: PMC8018760 DOI: 10.1523/jneurosci.3002-20.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 11/21/2022] Open
Abstract
In this short review, I describe from personal experience how every step in the career of any scientist, no matter how disjointed and pragmatic each might seem at the time, will almost inevitably meld together, to help us all tackle novel projects. My postdoctoral research in Paul Greengard's laboratory, where I investigated neurotransmitter-mediated phosphorylation of Synapsin I, was instrumental in my career progression, and Paul's support was instrumental in my ability to make a leap into independent research.
Collapse
|
30
|
Bargel S, Becam J, Chanu L, Lanot T, Martin M, Vaucel J, Willeman T, Fabresse N. Les gabapentinoïdes : une revue de la littérature. TOXICOLOGIE ANALYTIQUE ET CLINIQUE 2021. [DOI: 10.1016/j.toxac.2020.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
31
|
Zajączkowska R, Mika J, Leppert W, Kocot-Kępska M, Malec-Milewska M, Wordliczek J. Mirogabalin-A Novel Selective Ligand for the α2δ Calcium Channel Subunit. Pharmaceuticals (Basel) 2021; 14:112. [PMID: 33572689 PMCID: PMC7911728 DOI: 10.3390/ph14020112] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
The efficacy of neuropathic pain control remains unsatisfactory. Despite the availability of a variety of therapies, a significant proportion of patients suffer from poorly controlled pain of this kind. Consequently, new drugs and treatments are still being sought to remedy the situation. One such new drug is mirogabalin, a selective ligand for the α2δ subunits of voltage-gated calcium channels (VGCC) developed by Sankyo group for the management of neuropathic pain. In 2019 in Japan, mirogabalin was approved for peripheral neuropathic pain following the encouraging results of clinical trials conducted with diabetic peripheral neuropathic pain (DPNP) and postherpetic neuralgia (PHN) patients. The ligand selectivity of mirogabalin for α2δ-1 and α2δ-2 and its slower dissociation rate for α2δ-1 than for α2δ-2 subunits of VGCC may contribute to its strong analgesic effects, wide safety margin, and relatively lower incidence of adverse effects compared to pregabalin and gabapentin. This article discusses the mechanism of action of mirogabalin, presents data on its pharmacodynamics and pharmacokinetics, and reviews the available experimental and clinical studies that have assessed the efficacy and safety of the drug in the treatment of selected neuropathic pain syndromes.
Collapse
Affiliation(s)
- Renata Zajączkowska
- Department of Interdisciplinary Intensive Care, Jagiellonian University Medical College, 31-008 Krakow, Poland; (R.Z.); (J.W.)
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Wojciech Leppert
- Laboratory of Quality of Life Research, Chair and Department of Palliative Medicine, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Magdalena Kocot-Kępska
- Department of Pain Research and Treatment, Jagiellonian University Medical College, 31-008 Krakow, Poland;
| | - Małgorzata Malec-Milewska
- Department of Anesthesiology and Intensive Care, Medical Center for Postgraduate Education, 01-813 Warsaw, Poland;
| | - Jerzy Wordliczek
- Department of Interdisciplinary Intensive Care, Jagiellonian University Medical College, 31-008 Krakow, Poland; (R.Z.); (J.W.)
| |
Collapse
|
32
|
Fonseca F, Lenahan W, Dart RC, Papaseit E, Dargan PI, Wood DM, Guareschi M, Maremmani I, Auriacombe M, Farré M, Scherbaum N, Torrens M. Non-medical Use of Prescription Gabapentinoids (Gabapentin and Pregabalin) in Five European Countries. Front Psychiatry 2021; 12:676224. [PMID: 33995154 PMCID: PMC8113698 DOI: 10.3389/fpsyt.2021.676224] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Non-medical use (NMU) of prescription GABA analogs (pregabalin and gabapentin) has been reported especially in opiate dependent persons. However, by now the prevalence of NMU of gabapentinoids in the general population has not been sufficiently evaluated. The aim of this research paper is to determine the prevalence of prescription GABA analog NMU and associated demographics in five European countries with special detail of Spain. Methods: The RADARS Survey of Non-Medical Use of Prescription Drugs Program (NMURx) is a harmonized series of contemporaneous cross-sectional surveys of adults conducted in multiple countries. NMURx collects data from the general population in each participating country about NMU of prescription drugs, illicit drugs, and associated demographics. NMU was defined as "using a medication without a doctor's prescription or for any reason other than what was recommended by their doctor." Responses from Spain (4Q2017, n=10,062) were analyzed in detail. Comparative data were available from France, Germany, Italy, and UK. Responses were collected using non-probability quota sampling and post-stratification population weighting was applied to reflect the national distributions of adults, based on age, gender, and census region. Rates of NMU and associated demographics were reported as rate of past 90-day NMU per 100,000 adult population with 95% confidence intervals. Results: Germany (1,197 per 100,000 adult population [95% CI: 1,004.3-1,379.1]) and United Kingdom (1,067 per 100,000 adult population [95% CI: 851.3-1,283.2]) presented the highest prevalence of gabapentinoids NMU. In Spain the prevalence of past 90 days GABA analog NMU was: 344.4, 95% (CI 204.8-484.0), with male predominance. Those who non-medically use GABA analogs had a higher prevalence of lifetime chronic pain, lifetime illicit drug use, and previous substance abuse treatment. In Spain, 20% of respondents who ever have used gabapentinoids, reported a lifetime NMU; the prevalence was higher for pregabalin 624 (6.2%) than for gabapentin 444 (4.4%). The main reasons for use were to self-treat pain and other medical conditions. Conclusions: The risk of NMU of gabapentinoids should not be neglected. Subjects with a history of chronic pain and lifetime substance use disorders had an increased risk of NMU of gabapentinoids.
Collapse
Affiliation(s)
- Francina Fonseca
- Hospital del Mar, Institut de Neuropsiquiatria i Addiccions (INAD), Barcelona, Spain.,Grup de Recerca en Addiccions, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain.,Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| | - William Lenahan
- Rocky Mountain Poison and Drug Safety, Denver Health and Hospital Authority, Denver, CO, United States
| | - Richard C Dart
- Rocky Mountain Poison and Drug Safety, Denver Health and Hospital Authority, Denver, CO, United States
| | - Esther Papaseit
- Clinical Pharmacology Unit, Hospital Universitari Germans Trias i Pujol (HUGTP-Germans Trias i Pujol Research Institute), Badalona, Spain.,Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| | - Paul I Dargan
- Clinical Toxicology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom.,Clinical Toxicology, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - David M Wood
- Clinical Toxicology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom.,Clinical Toxicology, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Marilena Guareschi
- Association for the Application of Neuroscientific Knowledge to Social Aims (AU-CNS), Lucca, Italy
| | - Icro Maremmani
- Vincent P. Dole Research Group, Santa Chiara University Hospital, University of Pisa, Pisa, Italy
| | - Marc Auriacombe
- Addiction Psychiatry Department, University of Bordeaux, Bordeaux, France.,Addiction Team, Sanpsy Centre National de la Recherche Scientifique (CNRS) USR 3413, Bordeaux, France.,Pôle Addictologie et Filière Régionale, Centre Hospitalier (CH) Charles Perrens and Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Magí Farré
- Clinical Pharmacology Unit, Hospital Universitari Germans Trias i Pujol (HUGTP-Germans Trias i Pujol Research Institute), Badalona, Spain.,Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| | - Norbert Scherbaum
- LVR-Hospital Essen, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Marta Torrens
- Hospital del Mar, Institut de Neuropsiquiatria i Addiccions (INAD), Barcelona, Spain.,Grup de Recerca en Addiccions, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain.,Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| |
Collapse
|
33
|
Hayes WJ, Ferdinand A, Neabore S, Kappes JA, Hayes KM, Berendse J. Patient Case Report: Gabapentin-Induced Hypoglycemia. J Pharm Pract 2020; 35:298-301. [PMID: 32990167 DOI: 10.1177/0897190020961229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Gabapentin is an analog of gamma-aminobutyric acid (GABA), but its complete mechanism is not well understood. Common adverse effects from gabapentin include somnolence, sedation, and dizziness. Hyperglycemia is listed as a possible adverse drug reaction in the labeling. Case reports describe hypoglycemia in patients with diabetes, peritoneal dialysis, and/or incomplete medication records. The following case report details a hypoglycemia episode as a potential result of a gabapentin use in a patient without diabetes. SUMMARY A 47-year old, 68 kg, white female presented to the emergency department with altered mental status. Her blood glucose level was 33 mg/dL. Gabapentin was started 1 week prior to the hypoglycemia episode. Her past medical history, concomitant medications, and other laboratory findings were not likely causes of her severe hypoglycemia. CONCLUSION Gabapentin appears to have effects on several voltage-gated calcium channels. Hypoglycemia may be due to gabapentin binding to the alpha2delta subunit of the calcium channels in the pancreas. Future research should investigate gabapentin and the potential for hypoglycemia.
Collapse
Affiliation(s)
- William J Hayes
- 15488South Dakota State University, College of Pharmacy, Monument Health-Flormann Clinic, Rapid City, SD, USA
| | | | - Stephan Neabore
- Internal Medicine Physician, Monument Health-Flormann Clinic, Rapid City, SD, USA
| | - John A Kappes
- 15488South Dakota State University, College of Pharmacy, Monument Health-Rapid City Hospital, Rapid City, SD, USA
| | - Katie M Hayes
- Monument Health-Rapid City Hospital, Rapid City, SD, USA
| | - Joseph Berendse
- 15488South Dakota State University, College of Pharmacy, VA Black Hills Health Care System, Fort Meade, SD, USA
| |
Collapse
|
34
|
CACNA1S haploinsufficiency confers resistance to New World arenavirus infection. Proc Natl Acad Sci U S A 2020; 117:19497-19506. [PMID: 32719120 DOI: 10.1073/pnas.1920551117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Understanding the genetics of susceptibility to infectious agents is of great importance to our ability to combat disease. Here, we show that voltage-gated calcium channels (VGCCs) are critical for cellular binding and entry of the New World arenaviruses Junín and Tacaribe virus, suggesting that zoonosis via these receptors could occur. Moreover, we demonstrate that α1s haploinsufficiency renders cells and mice more resistant to infection by these viruses. In addition to being more resistant to infection, haploinsufficient cells and mice required a lower dosage of VGCC antagonists to block infection. These studies underscore the importance of genetic variation in susceptibility to both viruses and pharmaceutics.
Collapse
|
35
|
Dolphin AC, Lee A. Presynaptic calcium channels: specialized control of synaptic neurotransmitter release. Nat Rev Neurosci 2020; 21:213-229. [PMID: 32161339 PMCID: PMC7873717 DOI: 10.1038/s41583-020-0278-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 11/09/2022]
Abstract
Chemical synapses are heterogeneous junctions formed between neurons that are specialized for the conversion of electrical impulses into the exocytotic release of neurotransmitters. Voltage-gated Ca2+ channels play a pivotal role in this process as they are the major conduits for the Ca2+ ions that trigger the fusion of neurotransmitter-containing vesicles with the presynaptic membrane. Alterations in the intrinsic function of these channels and their positioning within the active zone can profoundly alter the timing and strength of synaptic output. Advances in optical and electron microscopic imaging, structural biology and molecular techniques have facilitated recent breakthroughs in our understanding of the properties of voltage-gated Ca2+ channels that support their presynaptic functions. Here we examine the nature of these channels, how they are trafficked to and anchored within presynaptic boutons, and the mechanisms that allow them to function optimally in shaping the flow of information through neural circuits.
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
36
|
Buchta WC, Moutal A, Hines B, Garcia-Keller C, Smith ACW, Kalivas P, Khanna R, Riegel AC. Dynamic CRMP2 Regulation of CaV2.2 in the Prefrontal Cortex Contributes to the Reinstatement of Cocaine Seeking. Mol Neurobiol 2020; 57:346-357. [PMID: 31359322 PMCID: PMC6980501 DOI: 10.1007/s12035-019-01711-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/15/2019] [Indexed: 02/06/2023]
Abstract
Cocaine addiction remains a major health concern with limited effective treatment options. A better understanding of mechanisms underlying relapse may help inform the development of new pharmacotherapies. Emerging evidence suggests that collapsin response mediator protein 2 (CRMP2) regulates presynaptic excitatory neurotransmission and contributes to pathological changes during diseases, such as neuropathic pain and substance use disorders. We examined the role of CRMP2 and its interactions with a known binding partner, CaV2.2, in cocaine-seeking behavior. We employed the rodent self-administration model of relapse to drug seeking and focused on the prefrontal cortex (PFC) for its well-established role in reinstatement behaviors. Our results indicated that repeated cocaine self-administration resulted in a dynamic and persistent alteration in the PFC expression of CRMP2 and its binding partner, the CaV2.2 (N-type) voltage-gated calcium channel. Following cocaine self-administration and extinction training, the expression of both CRMP2 and CaV2.2 was reduced relative to yoked saline controls. By contrast, cued reinstatement potentiated CRMP2 expression and increased CaV2.2 expression above extinction levels. Lastly, we utilized the recently developed peptide myr-TAT-CBD3 to disrupt the interaction between CRMP2 and CaV2.2 in vivo. We assessed the reinstatement behavior after infusing this peptide directly into the medial PFC and found that it decreased cue-induced reinstatement of cocaine seeking. Taken together, these data suggest that neuroadaptations in the CRMP2/CaV2.2 signaling cascade in the PFC can facilitate drug-seeking behavior. Targeting such interactions has implications for the treatment of cocaine relapse behavior.
Collapse
Affiliation(s)
- William C Buchta
- Department of Neuroscience, Medical University of South Carolina (MUSC), 410C Basic Sciences Building, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona, Tucson, AZ, 85724, USA
| | - Bethany Hines
- Department of Neuroscience, Medical University of South Carolina (MUSC), 410C Basic Sciences Building, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Constanza Garcia-Keller
- Department of Neuroscience, Medical University of South Carolina (MUSC), 410C Basic Sciences Building, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Alexander C W Smith
- Department of Neuroscience, Medical University of South Carolina (MUSC), 410C Basic Sciences Building, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Peter Kalivas
- Department of Neuroscience, Medical University of South Carolina (MUSC), 410C Basic Sciences Building, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, AZ, 85724, USA
- Department of Anesthesiology, University of Arizona, Tucson, AZ, 85724, USA
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, USA
| | - Arthur C Riegel
- Department of Neuroscience, Medical University of South Carolina (MUSC), 410C Basic Sciences Building, 173 Ashley Avenue, Charleston, SC, 29425, USA.
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Department of Pharmacology, University of Arizona, Tucson, AZ, 85724, USA.
| |
Collapse
|
37
|
Eldahan KC, Williams HC, Cox DH, Gollihue JL, Patel SP, Rabchevsky AG. Paradoxical effects of continuous high dose gabapentin treatment on autonomic dysreflexia after complete spinal cord injury. Exp Neurol 2020; 323:113083. [DOI: 10.1016/j.expneurol.2019.113083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/23/2019] [Accepted: 10/13/2019] [Indexed: 12/15/2022]
|
38
|
Haworth AS, Brackenbury WJ. Emerging roles for multifunctional ion channel auxiliary subunits in cancer. Cell Calcium 2019; 80:125-140. [PMID: 31071485 PMCID: PMC6553682 DOI: 10.1016/j.ceca.2019.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/16/2019] [Accepted: 04/16/2019] [Indexed: 02/07/2023]
Abstract
Several superfamilies of plasma membrane channels which regulate transmembrane ion flux have also been shown to regulate a multitude of cellular processes, including proliferation and migration. Ion channels are typically multimeric complexes consisting of conducting subunits and auxiliary, non-conducting subunits. Auxiliary subunits modulate the function of conducting subunits and have putative non-conducting roles, further expanding the repertoire of cellular processes governed by ion channel complexes to processes such as transcellular adhesion and gene transcription. Given this expansive influence of ion channels on cellular behaviour it is perhaps no surprise that aberrant ion channel expression is a common occurrence in cancer. This review will focus on the conducting and non-conducting roles of the auxiliary subunits of various Ca2+, K+, Na+ and Cl- channels and the burgeoning evidence linking such auxiliary subunits to cancer. Several subunits are upregulated (e.g. Cavβ, Cavγ) and downregulated (e.g. Kvβ) in cancer, while other subunits have been functionally implicated as oncogenes (e.g. Navβ1, Cavα2δ1) and tumour suppressor genes (e.g. CLCA2, KCNE2, BKγ1) based on in vivo studies. The strengthening link between ion channel auxiliary subunits and cancer has exposed these subunits as potential biomarkers and therapeutic targets. However further mechanistic understanding is required into how these subunits contribute to tumour progression before their therapeutic potential can be fully realised.
Collapse
Affiliation(s)
- Alexander S Haworth
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK
| | - William J Brackenbury
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK.
| |
Collapse
|
39
|
Schifano F, Chiappini S. Pregabalin: A range of misuse-related unanswered questions. CNS Neurosci Ther 2019; 25:659-660. [PMID: 30834646 PMCID: PMC6488882 DOI: 10.1111/cns.13115] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 01/11/2023] Open
Affiliation(s)
- Fabrizio Schifano
- Psychopharmacology, Drug Misuse and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, UK
| | - Stefania Chiappini
- Psychopharmacology, Drug Misuse and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, UK
| |
Collapse
|
40
|
Abstract
Voltage-gated calcium (CaV) channels are associated with β and α2δ auxiliary subunits. This review will concentrate on the function of the α2δ protein family, which has four members. The canonical role for α2δ subunits is to convey a variety of properties on the CaV1 and CaV2 channels, increasing the density of these channels in the plasma membrane and also enhancing their function. More recently, a diverse spectrum of non-canonical interactions for α2δ proteins has been proposed, some of which involve competition with calcium channels for α2δ or increase α2δ trafficking and others which mediate roles completely unrelated to their calcium channel function. The novel roles for α2δ proteins which will be discussed here include association with low-density lipoprotein receptor-related protein 1 (LRP1), thrombospondins, α-neurexins, prion proteins, large conductance (big) potassium (BK) channels, and N-methyl-d-aspartate (NMDA) receptors.
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
41
|
Reuveny A, Shnayder M, Lorber D, Wang S, Volk T. Ma2/d promotes myonuclear positioning and association with the sarcoplasmic reticulum. Development 2018; 145:dev.159558. [PMID: 30093550 DOI: 10.1242/dev.159558] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 07/24/2018] [Indexed: 12/24/2022]
Abstract
The cytoplasm of striated myofibers contains a large number of membrane organelles, including sarcoplasmic reticulum (SR), T-tubules and the nuclear membrane. These organelles maintain a characteristic juxtaposition that appears to be essential for efficient inter-membranous exchange of RNA, proteins and ions. We found that the membrane-associated Muscle-specific α2/δ (Ma2/d) subunit of the Ca2+ channel complex localizes to the SR and T-tubules, and accumulates at the myonuclear surfaces. Furthermore, Ma2/d mutant larval muscles exhibit nuclear positioning defects, disruption of the nuclear-SR juxtapositioning, as well as impaired larval locomotion. Ma2/d localization at the nuclear membrane depends on the proper function of the nesprin ortholog Msp300 and the BAR domain protein Amphiphysin (Amph). Importantly, live imaging of muscle contraction in intact Drosophila larvae indicated altered distribution of Sarco/Endoplamic Reticulum Ca2+-ATPase (SERCA) around the myonuclei of Ma2/d mutant larvae. Co-immunoprecipitation analysis supports association between Ma2/d and Amph, and indirectly with Msp300. We therefore suggest that Ma2/d, in association with Msp300 and Amph, mediates interactions between the SR and the nuclear membrane.
Collapse
Affiliation(s)
- Adriana Reuveny
- Department of Molecular Genetics, Weizmann Institute, Rehovot 76100, Israel
| | - Marina Shnayder
- Department of Molecular Genetics, Weizmann Institute, Rehovot 76100, Israel
| | - Dana Lorber
- Department of Molecular Genetics, Weizmann Institute, Rehovot 76100, Israel
| | - Shuoshuo Wang
- Department of Molecular Genetics, Weizmann Institute, Rehovot 76100, Israel
| | - Talila Volk
- Department of Molecular Genetics, Weizmann Institute, Rehovot 76100, Israel
| |
Collapse
|
42
|
α-Neurexins Together with α2δ-1 Auxiliary Subunits Regulate Ca 2+ Influx through Ca v2.1 Channels. J Neurosci 2018; 38:8277-8294. [PMID: 30104341 DOI: 10.1523/jneurosci.0511-18.2018] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 07/03/2018] [Accepted: 07/10/2018] [Indexed: 11/21/2022] Open
Abstract
Action potential-evoked neurotransmitter release is impaired in knock-out neurons lacking synaptic cell-adhesion molecules α-neurexins (αNrxns), the extracellularly longer variants of the three vertebrate Nrxn genes. Ca2+ influx through presynaptic high-voltage gated calcium channels like the ubiquitous P/Q-type (CaV2.1) triggers release of fusion-ready vesicles at many boutons. α2δ Auxiliary subunits regulate trafficking and kinetic properties of CaV2.1 pore-forming subunits but it has remained unclear if this involves αNrxns. Using live cell imaging with Ca2+ indicators, we report here that the total presynaptic Ca2+ influx in primary hippocampal neurons of αNrxn triple knock-out mice of both sexes is reduced and involved lower CaV2.1-mediated transients. This defect is accompanied by lower vesicle release, reduced synaptic abundance of CaV2.1 pore-forming subunits, and elevated surface mobility of α2δ-1 on axons. Overexpression of Nrxn1α in αNrxn triple knock-out neurons is sufficient to restore normal presynaptic Ca2+ influx and synaptic vesicle release. Moreover, coexpression of Nrxn1α together with α2δ-1 subunits facilitates Ca2+ influx further but causes little augmentation together with a different subunit, α2δ-3, suggesting remarkable specificity. Expression of defined recombinant CaV2.1 channels in heterologous cells validates and extends the findings from neurons. Whole-cell patch-clamp recordings show that Nrxn1α in combination with α2δ-1, but not with α2δ-3, facilitates Ca2+ currents of recombinant CaV2.1 without altering channel kinetics. These results suggest that presynaptic Nrxn1α acts as a positive regulator of Ca2+ influx through CaV2.1 channels containing α2δ-1 subunits. We propose that this regulation represents an important way for neurons to adjust synaptic strength.SIGNIFICANCE STATEMENT Synaptic transmission between neurons depends on the fusion of neurotransmitter-filled vesicles with the presynaptic membrane, which subsequently activates postsynaptic receptors. Influx of calcium ions into the presynaptic terminal is the key step to trigger vesicle release and involves different subtypes of voltage-gated calcium channels. We study the regulation of calcium channels by neurexins, a family of synaptic cell-adhesion molecules that are essential for many synapse properties. Using optical measurements of calcium influx in cultured neurons and electrophysiological recordings of calcium currents from recombinant channels, we show that a major neurexin variant facilitates calcium influx through P/Q-type channels by interacting with their α2δ-1 auxiliary subunits. These results propose a novel way how neurons can modulate the strength of distinct synapses.
Collapse
|
43
|
Takahashi DK, Jin S, Prince DA. Gabapentin Prevents Progressive Increases in Excitatory Connectivity and Epileptogenesis Following Neocortical Trauma. Cereb Cortex 2018; 28:2725-2740. [PMID: 28981586 PMCID: PMC6041890 DOI: 10.1093/cercor/bhx152] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/30/2017] [Accepted: 06/01/2017] [Indexed: 11/12/2022] Open
Abstract
Neocortical injury initiates a cascade of events, some of which result in maladaptive epileptogenic reorganization of surviving neural circuits. Research focused on molecular and organizational changes that occur following trauma may reveal processes that underlie human post-traumatic epilepsy (PTE), a common and unfortunate consequence of traumatic brain injury. The latency between injury and development of PTE provides an opportunity for prophylactic intervention, once the key underlying mechanisms are understood. In rodent neocortex, injury to pyramidal neurons promotes axonal sprouting, resulting in increased excitatory circuitry that is one important factor promoting epileptogenesis. We used laser-scanning photostimulation of caged glutamate and whole-cell recordings in in vitro slices from injured neocortex to assess formation of new excitatory synapses, a process known to rely on astrocyte-secreted thrombospondins (TSPs), and to map the distribution of maladaptive circuit reorganization. We show that this reorganization is centered principally in layer V and associated with development of epileptiform activity. Short-term blockade of the synaptogenic effects of astrocyte-secreted TSPs with gabapentin (GBP) after injury suppresses the new excitatory connectivity and epileptogenesis for at least 2 weeks. Results reveal that aberrant circuit rewiring is progressive in vivo and provide further rationale for prophylactic anti-epileptogenic use of gabapentinoids following cortical trauma.
Collapse
Affiliation(s)
- D K Takahashi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Sha Jin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - D A Prince
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
44
|
Chincholkar M. Analgesic mechanisms of gabapentinoids and effects in experimental pain models: a narrative review. Br J Anaesth 2018; 120:1315-1334. [DOI: 10.1016/j.bja.2018.02.066] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 02/07/2018] [Accepted: 03/14/2018] [Indexed: 12/17/2022] Open
|
45
|
Kusuyama K, Tachibana T, Yamanaka H, Okubo M, Yoshiya S, Noguchi K. Upregulation of calcium channel alpha-2-delta-1 subunit in dorsal horn contributes to spinal cord injury-induced tactile allodynia. Spine J 2018; 18:1062-1069. [PMID: 29355786 DOI: 10.1016/j.spinee.2018.01.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/08/2017] [Accepted: 01/10/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Spinal cord injury (SCI) commonly results not only in motor paralysis but also in the emergence of neuropathic pain (NeuP), both of which can impair the quality of life for patients with SCI. In the clinical field, it is well known that pregabalin, which binds to the voltage-gated calcium channel alpha-2-delta-1 (α2δ-1) subunit has therapeutic effects on NeuP after SCI. A previous study has demonstrated that SCI increased α2δ-1 in the L4-L6 dorsal spinal cord of SCI rats by Western blot analysis and that the increase of α2δ-1 was correlated with tactile allodynia of the hind paw. However, the detailed feature of an increase in α2δ-1 protein in the spinal dorsal horn and the mechanism of pregabalin effect on SCI-induced NeuP have not been fully examined. PURPOSE This study aimed to examine the detailed distribution of α2δ-1 expression in the lumbar spinal cord after thoracic SCI in rats and the correlation of the therapeutic effect of pregabalin in SCI rats. STUDY DESIGN Male Sprague-Dawley rats underwent thoracic (T10) spinal cord contusion injury using the IH impactor device. Spinal cord injury rats received pregabalin (30 mg/kg) once a day for 2 weeks over a 4-week period after SCI. METHODS The mechanical threshold in the rat hind paw was measured over 4 weeks. Alpha-2-delta-1 expression in the lumbar spinal cord and in the dorsal root ganglion (DRG) was analyzed using immunohistochemistry and in situ hybridization histochemistry. RESULTS A significant reduction of the withdrawal threshold of mechanical stimuli to the hind paw was observed for 2 weeks and continued at least 4 weeks after SCI. In the control rats, expression of α2δ-1 immunoreactivity was detected mainly in laminae I and II in the lumbar dorsal horn. Thoracic SCI significantly increased α2δ-1 immunoreactivity in laminae I and II in the lumbar dorsal horn 4 weeks after SCI; however, thoracic SCI did not affect the expression of α2δ-1 mRNA in the L4 and L5 DRGs. Meanwhile, the signal intensity of α2δ-1 mRNAs in the lumbar spinal cord increased from Day 7 and continued for at least 28 days after SCI. Cellular analysis showed that SCI increased the number of α2δ-1-expressing cells in laminae I and II. The tactile allodynia of the hind paw in the SCI rats was reversed after pregabalin treatment and was maintained for 21 days. This administration of pregabalin decreased the α2δ-1 immunoreactivity significantly in the lumbar dorsal horn of thoracic SCI rats at 28 days after SCI. CONCLUSIONS The present study results suggest that an increase of α2δ-1 in the L4 and L5 dorsal horns after thoracic SCI is derived from the increase in the expression in lumbar spinal neurons. This increase may be involved in the development of NeuP in the hind paws and the therapeutic effect of pregabalin on central NeuP after SCI.
Collapse
Affiliation(s)
- Kazuki Kusuyama
- Department of Orthopaedic Surgery, Hyogo College of Medicine, Mukogawa-cho 1-1, Nishinomiya, Hyogo 663-8501, Japan; Department of Anatomy and Neuroscience, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Toshiya Tachibana
- Department of Orthopaedic Surgery, Hyogo College of Medicine, Mukogawa-cho 1-1, Nishinomiya, Hyogo 663-8501, Japan.
| | - Hiroki Yamanaka
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Masamichi Okubo
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Shinichi Yoshiya
- Department of Orthopaedic Surgery, Hyogo College of Medicine, Mukogawa-cho 1-1, Nishinomiya, Hyogo 663-8501, Japan
| | - Koichi Noguchi
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
46
|
Gong N, Park J, Luo ZD. Injury-induced maladaptation and dysregulation of calcium channel α 2 δ subunit proteins and its contribution to neuropathic pain development. Br J Pharmacol 2018; 175:2231-2243. [PMID: 28646556 PMCID: PMC5980513 DOI: 10.1111/bph.13930] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/05/2017] [Accepted: 06/12/2017] [Indexed: 01/12/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) play important roles in physiological functions including the modulation of neurotransmitter release, neuronal network activities, intracellular signalling pathways and gene expression. Some pathological conditions, including nerve injuries, can cause the dysregulation of VGCCs and their subunits. This in turn can lead to a functional maladaptation of VGCCs and their subunits, which can contribute to the development of disorders such as pain sensations. This review has summarized recent findings related to maladaptive changes in the dysregulated VGCC α2 δ1 subunit (Cav α2 δ1 ) with a focus on exploring the mechanisms underlying the contribution of Cav α2 δ1 to pain signal transduction. At least under neuropathic pain conditions, the dysregulated Cav α2 δ1 can modulate VGCC functions as well as other plasticity changes. The latter includes abnormal excitatory synaptogenesis resulting from its interactions with injury-induced extracellular matrix glycoprotein molecule thrombospondins, which is independent of the VGCC functions. Blocking Cav α2 δ1 with gabapentinoids can reverse neuropathic pain significantly with relatively mild side effects, but only in a small population of neuropathic pain patients due to reasons yet to be explored. There are emerging data suggesting that early preventive treatment with gabapentinoids can prevent aberrant excitatory synapse formation and the development of chronic pain. If these findings are confirmed clinically, this could be an attractive approach for neuropathic pain management. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Nian Gong
- Department of Anesthesiology & Perioperative CareSchool of Medicine, University of California IrvineIrvineCAUSA
| | - John Park
- Department of Pharmacology, School of MedicineUniversity of California IrvineIrvineCAUSA
| | - Z David Luo
- Department of Anesthesiology & Perioperative CareSchool of Medicine, University of California IrvineIrvineCAUSA
- Department of Pharmacology, School of MedicineUniversity of California IrvineIrvineCAUSA
| |
Collapse
|
47
|
Patel R, Montagut‐Bordas C, Dickenson AH. Calcium channel modulation as a target in chronic pain control. Br J Pharmacol 2018; 175:2173-2184. [PMID: 28320042 PMCID: PMC5980588 DOI: 10.1111/bph.13789] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/03/2017] [Accepted: 03/05/2017] [Indexed: 01/13/2023] Open
Abstract
Neuropathic pain remains poorly treated for large numbers of patients, and little progress has been made in developing novel classes of analgesics. To redress this issue, ziconotide (Prialt™) was developed and approved as a first-in-class synthetic version of ω-conotoxin MVIIA, a peptide blocker of Cav 2.2 channels. Unfortunately, the impracticalities of intrathecal delivery, low therapeutic index and severe neurological side effects associated with ziconotide have restricted its use to exceptional circumstances. Ziconotide exhibits no state or use-dependent block of Cav 2.2 channels; activation state-dependent blockers were hypothesized to circumvent the side effects of state-independent blockers by selectively targeting high-frequency firing of nociceptive neurones in chronic pain states, thus alleviating aberrant pain but not affecting normal sensory transduction. Unfortunately, numerous drugs, including state-dependent calcium channel blockers, have displayed efficacy in preclinical models but have subsequently been disappointing in clinical trials. In recent years, it has become more widely acknowledged that trans-aetiological sensory profiles exist amongst chronic pain patients and may indicate similar underlying mechanisms and drug sensitivities. Heterogeneity amongst patients, a reliance on stimulus-evoked endpoints in preclinical studies and a failure to utilize translatable endpoints, all are likely to have contributed to negative clinical trial results. We provide an overview of how electrophysiological and operant-based assays provide insight into sensory and affective aspects of pain in animal models and how these may relate to chronic pain patients in order to improve the bench-to-bedside translation of calcium channel modulators. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Ryan Patel
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | | | - Anthony H Dickenson
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
48
|
|
49
|
Kato AS, Witkin JM. Protein complexes as psychiatric and neurological drug targets. Biochem Pharmacol 2018; 151:263-281. [PMID: 29330067 DOI: 10.1016/j.bcp.2018.01.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/05/2018] [Indexed: 12/25/2022]
Abstract
The need for improved medications for psychiatric and neurological disorders is clear. Difficulties in finding such drugs demands that all strategic means be utilized for their invention. The discovery of forebrain specific AMPA receptor antagonists, which selectively block the specific combinations of principal and auxiliary subunits present in forebrain regions but spare targets in the cerebellum, was recently disclosed. This discovery raised the possibility that other auxiliary protein systems could be utilized to help identify new medicines. Discussion of the TARP-dependent AMPA receptor antagonists has been presented elsewhere. Here we review the diversity of protein complexes of neurotransmitter receptors in the nervous system to highlight the broad range of protein/protein drug targets. We briefly outline the structural basis of protein complexes as drug targets for G-protein-coupled receptors, voltage-gated ion channels, and ligand-gated ion channels. This review highlights heterodimers, subunit-specific receptor constructions, multiple signaling pathways, and auxiliary proteins with an emphasis on the later. We conclude that the use of auxiliary proteins in chemical compound screening could enhance the detection of specific, targeted drug searches and lead to novel and improved medicines for psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Akihiko S Kato
- Neuroscience Discovery, Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA.
| | - Jeffrey M Witkin
- Neuroscience Discovery, Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
50
|
Bonnet U, Scherbaum N. How addictive are gabapentin and pregabalin? A systematic review. Eur Neuropsychopharmacol 2017; 27:1185-1215. [PMID: 28988943 DOI: 10.1016/j.euroneuro.2017.08.430] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/16/2017] [Accepted: 08/20/2017] [Indexed: 12/21/2022]
Abstract
In the last ten years, gabapentin and pregabalin have been becoming dispensed broadly and sold on black markets, thereby, exposing millions to potential side-effects. Meanwhile, several pharmacovigilance-databases have warned for potential abuse liabilities and overdose fatalities in association with both gabapentinoids. To evaluate their addiction risk in more detail, we conducted a systematic review on PubMed/Scopus and included 106 studies. We did not find convincing evidence of a vigorous addictive power of gabapentinoids which is primarily suggested from their limited rewarding properties, marginal notes on relapses, and the very few cases with gabapentinoid-related behavioral dependence symptoms (ICD-10) in patients without a prior abuse history (N=4). In support, there was no publication about people who sought treatment for the use of gabapentinoids. Pregabalin appeared to be somewhat more addictive than gabapentin regarding the magnitude of behavioral dependence symptoms, transitions from prescription to self-administration, and the durability of the self-administrations. The principal population at risk for addiction of gabapentinoids consists of patients with other current or past substance use disorders (SUD), mostly opioid and multi-drug users, who preferred pregabalin. Pure overdoses of gabapentinoids appeared to be relative safe but can become lethal (pregabalin > gabapentin) in mixture with other psychoactive drugs, especially opioids again and sedatives. Based upon these results, we compared the addiction risks of gabapentin and pregabalin with those of traditional psychoactive substances and recommend that in patients with a history of SUD, gabapentinoids should be avoided or if indispensable, administered with caution by using a strict therapeutic and prescription monitoring.
Collapse
Affiliation(s)
- U Bonnet
- Department of Psychiatry, Psychotherapy, and Psychosomatic Medicine, Evangelisches Krankenhaus Castrop-Rauxel, Academic Teaching Hospital of the University of Duisburg-Essen, Grutholzallee 21, D-44577 Castrop-Rauxel, Germany; LVR-Hospital Essen, Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Duisburg-Essen, D-45147 Essen, Germany.
| | - N Scherbaum
- LVR-Hospital Essen, Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Duisburg-Essen, D-45147 Essen, Germany
| |
Collapse
|