1
|
Milanick W, Li J, Thomas CI, Al-Yaari M, Guerrero-Given D, Kamasawa N, Young SM. Presynaptic α 2δs specify synaptic gain, not synaptogenesis, in the mammalian brain. Neuron 2025:S0896-6273(25)00296-X. [PMID: 40367942 DOI: 10.1016/j.neuron.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 03/13/2025] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
The α2δs are a family of extracellular synaptic molecules that are auxiliary subunits of voltage-gated Ca2+ channel (CaV) complexes. They are linked to brain disorders and are drug targets. The α2δs are implicated in controlling synapse development and function through distinct CaV-dependent and CaV-independent pathways. However, the mechanisms of action remain enigmatic since synapses contain mixtures of α2δ isoforms in the pre- and postsynaptic compartments. We developed a triple conditional knockout mouse model and demonstrated the combined selective presynaptic ablation of α2δs in vivo in a developing mammalian glutamatergic synapse. We identified presynaptic α2δs as positive regulators of Munc13-1 levels, an essential neurotransmitter release protein. We found that mammalian synapse development, presynaptic CaV2.1 organization, and the transsynaptic alignment of presynaptic release sites and postsynaptic glutamate receptors are independent of presynaptic α2δs. Therefore, our results define presynaptic α2δ regulatory roles and suggest a new α2δ role in controlling synaptic strength and plasticity.
Collapse
Affiliation(s)
- William Milanick
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA 52242, USA
| | - Jianing Li
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Connon I Thomas
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Mohammed Al-Yaari
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Debbie Guerrero-Given
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Naomi Kamasawa
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Samuel M Young
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pediatrics, Department of Pharmacology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
2
|
Kim H, Moon S, Kim M, Oh H, Park J, Kim S, Yoo T, Kim JY, Kim Y, Kim YM, Choi YK. Upregulation of astrocytic mitochondrial functions via Korean red ginseng-induced CREB-BK α-HIF-1 α axis through L-type Ca 2+ channel subunits α1C and β4. J Cereb Blood Flow Metab 2025:271678X251332760. [PMID: 40314209 PMCID: PMC12048403 DOI: 10.1177/0271678x251332760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/28/2025] [Accepted: 03/20/2025] [Indexed: 05/03/2025]
Abstract
Korean red ginseng extract (KRGE) enhances astrocytic functions through hypoxia-inducible factor-1α (HIF-1α). Astrocytic Ca2+ influx through L-type Ca2+ channels (LTCCs) facilitates neurovascular communication, while the large-conductance Ca2+- and voltage-activated K+ (BK) channel mediates K+ efflux for vasodilation. However, the role of LTCC subunits in KRGE-mediated BKα and HIF-1α expression in astrocytes remains unclear. This study aimed to investigate the effects of KRGE on LTCC subunits, cytosolic Ca2+ influx, and BKα and HIF-1α induction in human astrocytes. The levels of BKα, LTCCs, and HIF-1α were analyzed in KRGE-treated mouse brain tissue using immunohistochemistry. Human astrocytes treated with an LTCC agonist exhibited increased BKα and HIF-1α protein levels. Similarly, KRGE increased the levels of LTCC subunits α1 C and β4, cytosolic Ca2+ influx, BKα, and HIF-1α. Moreover, knockdown of either α1 C or β4 attenuated KRGE-induced increases in Ca2+ influx and HIF-1α levels. Notably, their combined knockdown synergistically reduced KRGE-induced increases in BKα levels, mitochondrial mass, ATP production, and O2 consumption. The corpus callosum astrocytes of KRGE-treated mice exhibited increased levels of α1 C and β4, BKα, HIF-1α, and cAMP-response element binding protein (CREB). Collectively, these findings suggest that KRGE induced astrocytic BKα and HIF-1α expression via LTCC-mediated Ca2+ influx and subsequent CREB activation.
Collapse
Affiliation(s)
- Hyungsu Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Sunhong Moon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Minsu Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Hyungkeun Oh
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Jinhong Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Suji Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Taehyung Yoo
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Ji-Yoon Kim
- Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Yonghee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Yoon Kyung Choi
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Chen Z, Minor DL. Electrosome assembly: Structural insights from high voltage-activated calcium channel (CaV)-chaperone interactions. Biochem Soc Trans 2025; 53:BST20240422. [PMID: 39912874 DOI: 10.1042/bst20240422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/18/2024] [Accepted: 01/09/2025] [Indexed: 02/07/2025]
Abstract
Ion channels are multicomponent complexes (termed here as"electrosomes") that conduct the bioelectrical signals required for life. It has been appreciated for decades that assembly is critical for proper channel function, but knowledge of the factors that undergird this important process has been lacking. Although there are now exemplar structures of representatives of most major ion channel classes, there has been no direct structural information to inform how these complicated, multipart complexes are put together or whether they interact with chaperone proteins that aid in their assembly. Recent structural characterization of a complex of the endoplasmic membrane protein complex (EMC) chaperone and a voltage-gated calcium channel (CaV) assembly intermediate comprising the pore-forming CaVα1 and cytoplasmic CaVβ subunits offers the first structural view into the assembly of a member of the largest ion channel class, the voltagegated ion channel (VGIC) superfamily. The structure shows how the EMC remodels the CaVα1/CaVβ complex through a set of rigid body movements for handoff to the extracellular CaVα2δ subunit to complete channel assembly in a process that involves intersubunit coordination of a divalent cation and ordering of CaVα1 elements. These findings set a new framework for deciphering the structural underpinnings of ion channel biogenesis that has implications for understanding channel function, how drugs and disease mutations act, and for investigating how other membrane proteins may engage the ubiquitous EMC chaperone.
Collapse
Affiliation(s)
- Zhou Chen
- Cardiovascular Research Institute, University of California-San Francisco, San Francisco, CA 94158-9001, U.S.A
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California-San Francisco, San Francisco, CA 94158-9001, U.S.A
- Department of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California-San Francisco, San Francisco, CA 94158-9001, U.S.A
- California Institute for Quantitative Biomedical Research, University of California-San Francisco, San Francisco, CA 94158-9001, U.S.A
- Kavli Institute for Fundamental Neuroscience, University of California-San Francisco, San Francisco, CA 94158-9001, U.S.A
- Molecular Biophysics and Integrated Bio-imaging Division Lawrence Berkeley National Laboratory, Berkeley, CA 94720 CA 94720, U.S.A
| |
Collapse
|
4
|
Haddad S, Hessenberger M, Ablinger C, Eibl C, Stanika R, Campiglio M, Obermair GJ. Autism-Linked Mutations in α 2δ-1 and α 2δ-3 Reduce Protein Membrane Expression but Affect Neither Calcium Channels nor Trans-Synaptic Signaling. Pharmaceuticals (Basel) 2024; 17:1608. [PMID: 39770450 PMCID: PMC11677996 DOI: 10.3390/ph17121608] [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: 10/17/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND α2δ proteins regulate membrane trafficking and biophysical properties of voltage-gated calcium channels. Moreover, they modulate axonal wiring, synapse formation, and trans-synaptic signaling. Several rare missense variants in CACNA2D1 (coding for α2δ-1) and CACNA2D3 (coding for α2δ-3) genes were identified in patients with autism spectrum disorder (ASD). However, the pathogenicity of these variants is not known, and the molecular mechanism by which α2δ proteins may contribute to the pathophysiology of autism is, as of today, not understood. Therefore, in this study we functionally characterized two heterozygous missense variants in α2δ-1 (p.R351T) and α2δ-3 (p.A275T), previously identified in patients with ASD. METHODS Electrophysiological recordings in transfected tsA201 cells were used to study specific channel-dependent functions of mutated α2δ proteins. Membrane expression, presynaptic targeting, and trans-synaptic signaling of mutated α2δ proteins were studied upon expression in murine cultured hippocampal neurons. RESULTS Homologous expression of both mutated α2δ proteins revealed a strongly reduced membrane expression and synaptic localization compared to the corresponding wild type α2δ proteins. Moreover, the A275T mutation in α2δ-3 resulted in an altered glycosylation pattern upon heterologous expression. However, neither of the mutations compromised the biophysical properties of postsynaptic L-type (CaV1.2 and CaV1.3) and presynaptic P/Q-type (CaV2.1) channels when co-expressed in tsA201 cells. Furthermore, presynaptic expression of p.R351T in the α2δ-1 splice variant lacking exon 23 did not affect trans-synaptic signaling to postsynaptic GABAA receptors. CONCLUSIONS Our data provide evidence that the pathophysiological mechanisms of ASD-causing mutations of α2δ proteins may not involve their classical channel-dependent and trans-synaptic functions. Alternatively, these mutations may induce subtle changes in synapse formation or neuronal network function, highlighting the need for future α2δ protein-linked disease models.
Collapse
Affiliation(s)
- Sabrin Haddad
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Manuel Hessenberger
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| | - Cornelia Ablinger
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Clarissa Eibl
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| | - Ruslan Stanika
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| | - Marta Campiglio
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Gerald J. Obermair
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| |
Collapse
|
5
|
Zurek NA, Thiyagarajan S, Ehsanian R, Goins AE, Goyal S, Shilling M, Lambert CG, Westlund KN, Alles SRA. Machine Learning Elucidates Electrophysiological Properties Predictive of Multi- and Single-Firing Human and Mouse Dorsal Root Ganglia Neurons. eNeuro 2024; 11:ENEURO.0248-24.2024. [PMID: 39299808 PMCID: PMC11457269 DOI: 10.1523/eneuro.0248-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/08/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024] Open
Abstract
Human and mouse dorsal root ganglia (hDRG and mDRG) neurons are important tools in understanding the molecular and electrophysiological mechanisms that underlie nociception and drive pain behaviors. One of the simplest differences in firing phenotypes is that neurons are single-firing (exhibit only one action potential) or multi-firing (exhibit 2 or more action potentials). To determine if single- and multi-firing hDRG neurons exhibit differences in intrinsic properties, firing phenotypes, and AP waveform properties, and if these properties could be used to predict multi-firing, we measured 22 electrophysiological properties by whole-cell patch-clamp electrophysiology of 94 hDRG neurons from six male and four female donors. We then analyzed the data using several machine learning models to determine if these properties could be used to predict multi-firing. We used 1,000 iterations of Monte Carlo cross-validation to split the data into different train and test sets and tested the logistic regression, k-nearest neighbors, random forest, support vector classifier, and XGBoost machine learning models. All models tested had a >80% accuracy on average, with support vector classifier, and XGBoost performing the best. We found that several properties correlated with multi-firing hDRG neurons and together could be used to predict multi-firing neurons in hDRG including a long decay time, a low rheobase, and long first spike latency. We also found that the hDRG models were able to predict multi-firing with 90% accuracy in mDRG neurons. Understanding these properties could be beneficial in the elucidation of targets on peripheral sensory neurons related to pain.
Collapse
Affiliation(s)
- Nesia A Zurek
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Sherwin Thiyagarajan
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Reza Ehsanian
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Aleyah E Goins
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Sachin Goyal
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Mark Shilling
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Christophe G Lambert
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131
| | - Karin N Westlund
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| |
Collapse
|
6
|
Medeiros AT, Gratz SJ, Delgado A, Ritt JT, O'Connor-Giles KM. Ca 2+ channel and active zone protein abundance intersects with input-specific synapse organization to shape functional synaptic diversity. eLife 2024; 12:RP88412. [PMID: 39291956 PMCID: PMC11410372 DOI: 10.7554/elife.88412] [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: 09/19/2024] Open
Abstract
Synaptic heterogeneity is a hallmark of nervous systems that enables complex and adaptable communication in neural circuits. To understand circuit function, it is thus critical to determine the factors that contribute to the functional diversity of synapses. We investigated the contributions of voltage-gated calcium channel (VGCC) abundance, spatial organization, and subunit composition to synapse diversity among and between synapses formed by two closely related Drosophila glutamatergic motor neurons with distinct neurotransmitter release probabilities (Pr). Surprisingly, VGCC levels are highly predictive of heterogeneous Pr among individual synapses of either low- or high-Pr inputs, but not between inputs. We find that the same number of VGCCs are more densely organized at high-Pr synapses, consistent with tighter VGCC-synaptic vesicle coupling. We generated endogenously tagged lines to investigate VGCC subunits in vivo and found that the α2δ-3 subunit Straightjacket along with the CAST/ELKS active zone (AZ) protein Bruchpilot, both key regulators of VGCCs, are less abundant at high-Pr inputs, yet positively correlate with Pr among synapses formed by either input. Consistently, both Straightjacket and Bruchpilot levels are dynamically increased across AZs of both inputs when neurotransmitter release is potentiated to maintain stable communication following glutamate receptor inhibition. Together, these findings suggest a model in which VGCC and AZ protein abundance intersects with input-specific spatial and molecular organization to shape the functional diversity of synapses.
Collapse
Affiliation(s)
- Audrey T Medeiros
- Neuroscience Graduate Training Program, Brown University, Providence, United States
| | - Scott J Gratz
- Department of Neuroscience, Brown University, Providence, United States
| | - Ambar Delgado
- Department of Neuroscience, Brown University, Providence, United States
| | - Jason T Ritt
- Department of Neuroscience, Brown University, Providence, United States
- Carney Institute for Brain Science, Brown University, Providence, United States
| | - Kate M O'Connor-Giles
- Neuroscience Graduate Training Program, Brown University, Providence, United States
- Department of Neuroscience, Brown University, Providence, United States
- Carney Institute for Brain Science, Brown University, Providence, United States
| |
Collapse
|
7
|
Chen WF, Chuang JMJ, Yang SN, Chen NF, Bhattacharya M, Liu HT, Dhama K, Chakraborty C, Wen ZH. Gene expression profiling and the isocitrate dehydrogenase mutational landscape of temozolomide‑resistant glioblastoma. Oncol Lett 2024; 28:378. [PMID: 38939621 PMCID: PMC11209862 DOI: 10.3892/ol.2024.14511] [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: 09/21/2023] [Accepted: 04/09/2024] [Indexed: 06/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain cancer that occurs more frequently than other brain tumors. The present study aimed to reveal a novel mechanism of temozolomide resistance in GBM using bioinformatics and wet lab analyses, including meta-Z analysis, Kaplan-Meier survival analysis, protein-protein interaction (PPI) network establishment, cluster analysis of co-expressed gene networks, and hierarchical clustering of upregulated and downregulated genes. Next-generation sequencing and quantitative PCR analyses revealed downregulated [tyrosine kinase with immunoglobulin and epidermal growth factor homology domains 1 (TIE1), calcium voltage-gated channel auxiliary subunit α2Δ1 (CACNA2D1), calpain 6 (CAPN6) and a disintegrin and metalloproteinase with thrombospondin motifs 6 (ADAMTS6)] and upregulated [serum amyloid (SA)A1, SAA2, growth differentiation factor 15 (GDF15) and ubiquitin specific peptidase 26 (USP26)] genes. Different statistical models were developed for these genes using the Z-score for P-value conversion, and Kaplan-Meier plots were constructed using several patient cohorts with brain tumors. The highest number of nodes was observed in the PPI network was for ADAMTS6 and TIE1. The PPI network model for all genes contained 35 nodes and 241 edges. Immunohistochemical staining was performed using isocitrate dehydrogenase (IDH)-wild-type or IDH-mutant GBM samples from patients and a significant upregulation of TIE1 (P<0.001) and CAPN6 (P<0.05) protein expression was demonstrated in IDH-mutant GBM in comparison with IDH-wild-type GBM. Structural analysis revealed an IDH-mutant model demonstrating the mutant residues (R132, R140 and R172). The findings of the present study will help the future development of novel biomarkers and therapeutics for brain tumors.
Collapse
Affiliation(s)
- Wu-Fu Chen
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| | - Jimmy Ming-Jung Chuang
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
| | - San-Nan Yang
- Department of Pediatrics, E-DA Hospital, School of Medicine, College of Medicine I-Shou University, Kaohsiung 82445, Taiwan, R.O.C
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan, R.O.C
| | - Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan, R.O.C
- Center for General Education, Cheng Shiu University, Kaohsiung 833301, Taiwan, R.O.C
| | | | - Hsin-Tzu Liu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970374, Taiwan, R.O.C
| | - Kuldeep Dhama
- Division of Pathology, Indian Council of Agriculture Research-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| |
Collapse
|
8
|
Kissinger ST, O'neil E, Li B, Johnson KW, Krajewski JL, Kato AS. Distinctive Neurophysiological Signatures of Analgesia after Inflammatory Pain in the ACC of Freely Moving Mice. J Neurosci 2024; 44:e2231232024. [PMID: 38755005 PMCID: PMC11255429 DOI: 10.1523/jneurosci.2231-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/11/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Preclinical assessments of pain have often relied upon behavioral measurements and anesthetized neurophysiological recordings. Current technologies enabling large-scale neural recordings, however, have the potential to unveil quantifiable pain signals in conscious animals for preclinical studies. Although pain processing is distributed across many brain regions, the anterior cingulate cortex (ACC) is of particular interest in isolating these signals given its suggested role in the affective ("unpleasant") component of pain. Here, we explored the utility of the ACC toward preclinical pain research using head-mounted miniaturized microscopes to record calcium transients in freely moving male mice expressing genetically encoded calcium indicator 6f (GCaMP6f) under the Thy1 promoter. We verified the expression of GCaMP6f in excitatory neurons and found no intrinsic behavioral differences in this model. Using a multimodal stimulation paradigm across naive, pain, and analgesic conditions, we found that while ACC population activity roughly scaled with stimulus intensity, single-cell representations were highly flexible. We found only low-magnitude increases in population activity after complete Freund's adjuvant (CFA) and insufficient evidence for the existence of a robust nociceptive ensemble in the ACC. However, we found a temporal sharpening of response durations and generalized increases in pairwise neural correlations in the presence of the mechanistically distinct analgesics gabapentin or ibuprofen after (but not before) CFA-induced inflammatory pain. This increase was not explainable by changes in locomotion alone. Taken together, these results highlight challenges in isolating distinct pain signals among flexible representations in the ACC but suggest a neurophysiological hallmark of analgesia after pain that generalizes to at least two analgesics.
Collapse
Affiliation(s)
- Samuel T Kissinger
- Lilly Research Laboratories, Department of Neuroscience, Indianapolis, Indiana 46285
| | - Estefania O'neil
- Lilly Research Laboratories, Department of Neuroscience, Indianapolis, Indiana 46285
| | - Baolin Li
- Lilly Research Laboratories, Department of Neuroscience, Indianapolis, Indiana 46285
| | - Kirk W Johnson
- Lilly Research Laboratories, Department of Neuroscience, Indianapolis, Indiana 46285
| | - Jeffrey L Krajewski
- Lilly Research Laboratories, Department of Neuroscience, Indianapolis, Indiana 46285
| | - Akihiko S Kato
- Lilly Research Laboratories, Department of Neuroscience, Indianapolis, Indiana 46285
| |
Collapse
|
9
|
Zurek NA, Thiyagarajan S, Ehsanian R, Goins AE, Goyal S, Shilling M, Lambert CG, Westlund KN, Alles SRA. Machine learning elucidates electrophysiological properties predictive of multi- and single-firing human and mouse dorsal root ganglia neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597213. [PMID: 38895314 PMCID: PMC11185744 DOI: 10.1101/2024.06.03.597213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Human and mouse dorsal root ganglia (hDRG and mDRG) neurons are important tools in understanding the molecular and electrophysiological mechanisms that underlie nociception and drive pain behaviors. One of the simplest differences in firing phenotypes is that neurons are single-firing (exhibit only one action potential) or multi-firing (exhibit 2 or more action potentials). To determine if single- and multi-firing hDRG exhibit differences in intrinsic properties, firing phenotypes, and AP waveform properties, and if these properties could be used to predict multi-firing, we measured 22 electrophysiological properties by whole-cell patch-clamp electrophysiology of 94 hDRG neurons from 6 male and 4 female donors. We then analyzed the data using several machine learning models to determine if these properties could be used to predict multi-firing. We used 1000 iterations of Monte Carlo Cross Validation to split the data into different train and test sets and tested the Logistic Regression, k-Nearest Neighbors, Random Forest, Supported Vector Classification, and XGBoost machine learning models. All models tested had a greater than 80% accuracy on average, with Supported Vector Classification and XGBoost performing the best. We found that several properties correlated with multi-firing hDRG neurons and together could be used to predict multi-firing neurons in hDRG including a long decay time, a low rheobase, and long first spike latency. We also found that the hDRG models were able to predict multi-firing with 90% accuracy in mDRG. Targeting the neuronal properties that lead to multi-firing could elucidate better targets for treatment of chronic pain.
Collapse
|
10
|
Upshaw WC, Soileau LG, Storey NR, Perkinson KA, Luther PM, Spillers NJ, Robinson CL, Miller BC, Ahmadzadeh S, Viswanath O, Shekoohi S, Kaye AD. An extract of phase II and III trials on recent developments in managing neuropathic pain syndromes: diabetic peripheral neuropathy, trigeminal neuralgia, and postherpetic neuralgia. Expert Opin Emerg Drugs 2024; 29:103-112. [PMID: 38410863 DOI: 10.1080/14728214.2024.2323193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/21/2024] [Indexed: 02/28/2024]
Abstract
INTRODUCTION Neuropathic pain (NP) conditions involve lesions to the somatosensory nervous system leading to chronic and debilitating pain. Many patients suffering from NP utilize pharmacological treatments with various drugs that seek to reduce pathologic neuronal states. However, many of these drugs show poor efficacy as well as cause significant adverse effects. Because of this, there is a major need for the development of safer and more efficacious drugs to treat NP. AREAS COVERED In this review, we analyzed current treatments being developed for a variety of NP conditions. Specifically, we sought drugs in phase II/III clinical trials with indications for NP conditions. Various databases were searched including Google Scholar, PubMed, and clinicaltrials.gov. EXPERT OPINION All the mentioned targets for treatments of NP seem to be promising alternatives for existing treatments that often possess poor side effect profiles for patients. However, gene therapy potentially offers the unique ability to inject a plasmid containing growth factors leading to nerve growth and repair. Because of this, gene therapy appears to be the most intriguing new treatment for NP.
Collapse
Affiliation(s)
- William C Upshaw
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | - Lenise G Soileau
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | - Nicholas R Storey
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | | | - Patrick M Luther
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | - Noah J Spillers
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, USA
| | - Christopher L Robinson
- Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA, USA
| | - Benjamin C Miller
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, USA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, USA
| | - Omar Viswanath
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, USA
- Creighton University School of Medicine, Phoenix, AZ, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, USA
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, USA
| |
Collapse
|
11
|
Medeiros AT, Gratz S, Delgado A, Ritt J, O’Connor-Giles KM. Ca 2+ channel and active zone protein abundance intersects with input-specific synapse organization to shape functional synaptic diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.02.535290. [PMID: 37034654 PMCID: PMC10081318 DOI: 10.1101/2023.04.02.535290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Synaptic heterogeneity is a hallmark of nervous systems that enables complex and adaptable communication in neural circuits. To understand circuit function, it is thus critical to determine the factors that contribute to the functional diversity of synapses. We investigated the contributions of voltage-gated calcium channel (VGCC) abundance, spatial organization, and subunit composition to synapse diversity among and between synapses formed by two closely related Drosophila glutamatergic motor neurons with distinct neurotransmitter release probabilities (Pr). Surprisingly, VGCC levels are highly predictive of heterogeneous Pr among individual synapses of either low- or high-Pr inputs, but not between inputs. We find that the same number of VGCCs are more densely organized at high-Pr synapses, consistent with tighter VGCC-synaptic vesicle coupling. We generated endogenously tagged lines to investigate VGCC subunits in vivo and found that the α2δ-3 subunit Straightjacket along with the CAST/ELKS active zone (AZ) protein Bruchpilot, both key regulators of VGCCs, are less abundant at high-Pr inputs, yet positively correlate with Pr among synapses formed by either input. Consistently, both Straightjacket and Bruchpilot levels are dynamically increased across AZs of both inputs when neurotransmitter release is potentiated to maintain stable communication following glutamate receptor inhibition. Together, these findings suggest a model in which VGCC and AZ protein abundance intersects with input-specific spatial and molecular organization to shape the functional diversity of synapses.
Collapse
Affiliation(s)
- A. T. Medeiros
- Neuroscience Graduate Training Program, Brown University, Providence, RI
| | - S.J. Gratz
- Department of Neuroscience, Brown University, Providence, RI
| | - A. Delgado
- Department of Neuroscience, Brown University, Providence, RI
| | - J.T. Ritt
- Department of Neuroscience, Brown University, Providence, RI
- Carney Institute for Brain Science, Brown University, Providence, RI
| | - Kate M. O’Connor-Giles
- Neuroscience Graduate Training Program, Brown University, Providence, RI
- Department of Neuroscience, Brown University, Providence, RI
- Carney Institute for Brain Science, Brown University, Providence, RI
| |
Collapse
|
12
|
Varadi G. Mechanism of Analgesia by Gabapentinoid Drugs: Involvement of Modulation of Synaptogenesis and Trafficking of Glutamate-Gated Ion Channels. J Pharmacol Exp Ther 2024; 388:121-133. [PMID: 37918854 DOI: 10.1124/jpet.123.001669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
Gabapentinoids have clinically been used for treating epilepsy, neuropathic pain, and several other neurologic disorders for >30 years; however, the definitive molecular mechanism responsible for their therapeutic actions remained uncertain. The conventional pharmacological observation regarding their efficacy in chronic pain modulation is the weakening of glutamate release at presynaptic terminals in the spinal cord. While the α2/δ-1 subunit of voltage-gated calcium channels (VGCCs) has been identified as the primary drug receptor for gabapentinoids, the lack of consistent effect of this drug class on VGCC function is indicative of a minor role in regulating this ion channel's activity. The current review targets the efficacy and mechanism of gabapentinoids in treating chronic pain. The discovery of interaction of α2/δ-1 with thrombospondins established this protein as a major synaptogenic neuronal receptor for thrombospondins. Other findings identified α2/δ-1 as a powerful regulator of N-methyl-D-aspartate receptor (NMDA) and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) by potentiating the synaptic expression, a putative pathophysiological mechanism of neuropathic pain. Further, the interdependent interactions between thrombospondin and α2/δ-1 contribute to chronic pain states, while gabapentinoid ligands efficaciously reverse such pain conditions. Gabapentin normalizes and even blocks NMDAR and AMPAR synaptic targeting and activity elicited by nerve injury. SIGNIFICANCE STATEMENT: Gabapentinoid drugs are used to treat various neurological conditions including chronic pain. In chronic pain states, gene expression of cacnα2/δ-1 and thrombospondins are upregulated and promote aberrant excitatory synaptogenesis. The complex trait of protein associations that involve interdependent interactions between α2/δ-1 and thrombospondins, further, association of N-methyl-D-aspartate receptor and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor with the C-tail of α2/δ-1, constitutes a macromolecular signaling complex that forms the crucial elements for the pharmacological mode of action of gabapentinoids.
Collapse
|
13
|
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
|
14
|
Blankenship HE, Carter KA, Cassidy NT, Markiewicz AN, Thellmann MI, Sharpe AL, Freeman WM, Beckstead MJ. VTA dopamine neurons are hyperexcitable in 3xTg-AD mice due to casein kinase 2-dependent SK channel dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.16.567486. [PMID: 38014232 PMCID: PMC10680865 DOI: 10.1101/2023.11.16.567486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Alzheimer's disease (AD) patients exhibit neuropsychiatric symptoms that extend beyond classical cognitive deficits, suggesting involvement of subcortical areas. Here, we investigated the role of midbrain dopamine (DA) neurons in AD using the amyloid + tau-driven 3xTg-AD mouse model. We found deficits in reward-based operant learning in AD mice, suggesting possible VTA DA neuron dysregulation. Physiological assessment revealed hyperexcitability and disrupted firing in DA neurons caused by reduced activity of small-conductance calcium-activated potassium (SK) channels. RNA sequencing from contents of single patch-clamped DA neurons (Patch-seq) identified up-regulation of the SK channel modulator casein kinase 2 (CK2). Pharmacological inhibition of CK2 restored SK channel activity and normal firing patterns in 3xTg-AD mice. These findings shed light on a complex interplay between neuropsychiatric symptoms and subcortical circuits in AD, paving the way for novel treatment strategies.
Collapse
Affiliation(s)
- Harris E Blankenship
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation
- Department of Physiology, University of Oklahoma Health Sciences Center
| | - Kelsey A Carter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation
| | - Nina T Cassidy
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation
| | | | | | - Amanda L Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center
| | - Willard M Freeman
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Michael J Beckstead
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| |
Collapse
|
15
|
Ślęczkowska M, Misra K, Santoro S, Gerrits MM, Hoeijmakers JGJ. Ion Channel Genes in Painful Neuropathies. Biomedicines 2023; 11:2680. [PMID: 37893054 PMCID: PMC10604193 DOI: 10.3390/biomedicines11102680] [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: 09/06/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Neuropathic pain (NP) is a typical symptom of peripheral nerve disorders, including painful neuropathy. The biological mechanisms that control ion channels are important for many cell activities and are also therapeutic targets. Disruption of the cellular mechanisms that govern ion channel activity can contribute to pain pathophysiology. The voltage-gated sodium channel (VGSC) is the most researched ion channel in terms of NP; however, VGSC impairment is detected in only <20% of painful neuropathy patients. Here, we discuss the potential role of the other peripheral ion channels involved in sensory signaling (transient receptor potential cation channels), neuronal excitation regulation (potassium channels), involuntary action potential generation (hyperpolarization-activated cyclic nucleotide-gated channels), thermal pain (anoctamins), pH modulation (acid sensing ion channels), and neurotransmitter release (calcium channels) related to pain and their prospective role as therapeutic targets for painful neuropathy.
Collapse
Affiliation(s)
- Milena Ślęczkowska
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands;
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Kaalindi Misra
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, INSPE, 20132 Milan, Italy; (K.M.); (S.S.)
| | - Silvia Santoro
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, INSPE, 20132 Milan, Italy; (K.M.); (S.S.)
| | - Monique M. Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands;
| | - Janneke G. J. Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
16
|
Reyes Fernandez PC, Wright CS, Farach-Carson MC, Thompson WR. Examining Mechanisms for Voltage-Sensitive Calcium Channel-Mediated Secretion Events in Bone Cells. Calcif Tissue Int 2023; 113:126-142. [PMID: 37261463 PMCID: PMC11008533 DOI: 10.1007/s00223-023-01097-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
In addition to their well-described functions in cell excitability, voltage-sensitive calcium channels (VSCCs) serve a critical role in calcium (Ca2+)-mediated secretion of pleiotropic paracrine and endocrine factors, including those produced in bone. Influx of Ca2+ through VSCCs activates intracellular signaling pathways to modulate a variety of cellular processes that include cell proliferation, differentiation, and bone adaptation in response to mechanical stimuli. Less well understood is the role of VSCCs in the control of bone and calcium homeostasis mediated through secreted factors. In this review, we discuss the various functions of VSCCs in skeletal cells as regulators of Ca2+ dynamics and detail how these channels might control the release of bioactive factors from bone cells. Because VSCCs are druggable, a better understanding of the multiple functions of these channels in the skeleton offers the opportunity for developing new therapies to enhance and maintain bone and to improve systemic health.
Collapse
Affiliation(s)
- Perla C Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Christian S Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX, 77005, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
17
|
Chen Z, Mondal A, Abderemane-Ali F, Jang S, Niranjan S, Montaño JL, Zaro BW, Minor DL. EMC chaperone-Ca V structure reveals an ion channel assembly intermediate. Nature 2023; 619:410-419. [PMID: 37196677 PMCID: PMC10896479 DOI: 10.1038/s41586-023-06175-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 05/05/2023] [Indexed: 05/19/2023]
Abstract
Voltage-gated ion channels (VGICs) comprise multiple structural units, the assembly of which is required for function1,2. Structural understanding of how VGIC subunits assemble and whether chaperone proteins are required is lacking. High-voltage-activated calcium channels (CaVs)3,4 are paradigmatic multisubunit VGICs whose function and trafficking are powerfully shaped by interactions between pore-forming CaV1 or CaV2 CaVα1 (ref. 3), and the auxiliary CaVβ5 and CaVα2δ subunits6,7. Here we present cryo-electron microscopy structures of human brain and cardiac CaV1.2 bound with CaVβ3 to a chaperone-the endoplasmic reticulum membrane protein complex (EMC)8,9-and of the assembled CaV1.2-CaVβ3-CaVα2δ-1 channel. These structures provide a view of an EMC-client complex and define EMC sites-the transmembrane (TM) and cytoplasmic (Cyto) docks; interaction between these sites and the client channel causes partial extraction of a pore subunit and splays open the CaVα2δ-interaction site. The structures identify the CaVα2δ-binding site for gabapentinoid anti-pain and anti-anxiety drugs6, show that EMC and CaVα2δ interactions with the channel are mutually exclusive, and indicate that EMC-to-CaVα2δ hand-off involves a divalent ion-dependent step and CaV1.2 element ordering. Disruption of the EMC-CaV complex compromises CaV function, suggesting that the EMC functions as a channel holdase that facilitates channel assembly. Together, the structures reveal a CaV assembly intermediate and EMC client-binding sites that could have wide-ranging implications for the biogenesis of VGICs and other membrane proteins.
Collapse
Affiliation(s)
- Zhou Chen
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Abhisek Mondal
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Fayal Abderemane-Ali
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Seil Jang
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Sangeeta Niranjan
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - José L Montaño
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Balyn W Zaro
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, USA.
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
| |
Collapse
|
18
|
Ramírez A, Ogonaga-Borja I, Acosta B, Chiliquinga AJ, de la Garza J, Gariglio P, Ocádiz-Delgado R, Bañuelos C, Camacho J. Ion Channels and Personalized Medicine in Gynecological Cancers. Pharmaceuticals (Basel) 2023; 16:800. [PMID: 37375748 DOI: 10.3390/ph16060800] [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: 04/15/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Targeted therapy against cancer plays a key role in delivering safer and more efficient treatments. In the last decades, ion channels have been studied for their participation in oncogenic processes because their aberrant expression and/or function have been associated with different types of malignancies, including ovarian, cervical, and endometrial cancer. The altered expression or function of several ion channels have been associated with tumor aggressiveness, increased proliferation, migration, invasion, and metastasis of cancer cells and with poor prognosis in gynecological cancer patients. Most ion channels are integral membrane proteins easily accessible by drugs. Interestingly, a plethora of ion channel blockers have demonstrated anticancer activity. Consequently, some ion channels have been proposed as oncogenes, cancer, and prognostic biomarkers, as well as therapeutic targets in gynecological cancers. Here, we review the association of ion channels with the properties of cancer cells in these tumors, which makes them very promising candidates to be exploited in personalized medicine. The detailed analysis of the expression pattern and function of ion channels could help to improve the clinical outcomes in gynecological cancer patients.
Collapse
Affiliation(s)
- Ana Ramírez
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Calzada Universidad 14418, Parque Industrial Internacional, Tijuana 22390, Mexico
| | - Ingrid Ogonaga-Borja
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Av. 17 de Julio 5-21, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Brenda Acosta
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Av. 17 de Julio 5-21, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Andrea Jazmín Chiliquinga
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Av. 17 de Julio 5-21, Ibarra 100105, Ecuador
| | - Jaime de la Garza
- Unidad de Oncología Torácica y Laboratorio de Medicina Personalizada, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Ciudad de Mexico14080, Mexico
| | - Patricio Gariglio
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Rodolfo Ocádiz-Delgado
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Cecilia Bañuelos
- Programa Transdisciplinario en Desarrollo Científico y Tecnológico para la Sociedad, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Javier Camacho
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| |
Collapse
|
19
|
Kozai D, Numoto N, Nishikawa K, Kamegawa A, Kawasaki S, Hiroaki Y, Irie K, Oshima A, Hanzawa H, Shimada K, Kitano Y, Fujiyoshi Y. Recognition mechanism of a novel gabapentinoid drug, mirogabalin, for recombinant human α 2δ1, a voltage-gated calcium channel subunit. J Mol Biol 2023; 435:168049. [PMID: 36933823 DOI: 10.1016/j.jmb.2023.168049] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/22/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023]
Abstract
Mirogabalin is a novel gabapentinoid drug with a hydrophobic bicyclo substituent on the γ-aminobutyric acid moiety that targets the voltage-gated calcium channel subunit α2δ1. Here, to reveal the mirogabalin recognition mechanisms of α2δ1, we present structures of recombinant human α2δ1 with and without mirogabalin analyzed by cryo-electron microscopy. These structures show the binding of mirogabalin to the previously reported gabapentinoid binding site, which is the extracellular dCache_1 domain containing a conserved amino acid binding motif. A slight conformational change occurs around the residues positioned close to the hydrophobic group of mirogabalin. Mutagenesis binding assays identified that residues in the hydrophobic interaction region, in addition to several amino acid binding motif residues around the amino and carboxyl groups of mirogabalin, are critical for mirogabalin binding. The A215L mutation introduced to decrease the hydrophobic pocket volume predictably suppressed mirogabalin binding and promoted the binding of another ligand, L-Leu, with a smaller hydrophobic substituent than mirogabalin. Alterations of residues in the hydrophobic interaction region of α2δ1 to those of the α2δ2, α2δ3, and α2δ4 isoforms, of which α2δ3 and α2δ4 are gabapentin-insensitive, suppressed the binding of mirogabalin. These results support the importance of hydrophobic interactions in α2δ1 ligand recognition.
Collapse
Affiliation(s)
- Daisuke Kozai
- Cellular and Structural Physiology Institute (CeSPI), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Japan Biological Informatics Consortium, 2-4-32 Aomi, Koto-ku, Tokyo 135-0063, Japan; Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8501, Japan.
| | - Nobutaka Numoto
- Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8501, Japan.
| | - Kouki Nishikawa
- CeSPIA Inc., 2-1-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan; Joint Research Course for Advanced Biomolecular Characterization, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan.
| | - Akiko Kamegawa
- Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8501, Japan; CeSPIA Inc., 2-1-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan.
| | - Shohei Kawasaki
- Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan.
| | - Yoko Hiroaki
- Cellular and Structural Physiology Institute (CeSPI), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Japan Biological Informatics Consortium, 2-4-32 Aomi, Koto-ku, Tokyo 135-0063, Japan.
| | - Katsumasa Irie
- Cellular and Structural Physiology Institute (CeSPI), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.
| | - Atsunori Oshima
- Cellular and Structural Physiology Institute (CeSPI), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Institute for Glyco-core Research (iGCORE), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.
| | - Hiroyuki Hanzawa
- Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan.
| | - Kousei Shimada
- Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan.
| | - Yutaka Kitano
- Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan.
| | - Yoshinori Fujiyoshi
- Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8501, Japan; CeSPIA Inc., 2-1-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
20
|
Newmyer S, Ssemadaali MA, Radhakrishnan H, Javitz HS, Bhatnagar P. Electrically regulated cell-based intervention for viral infections. Bioeng Transl Med 2023; 8:e10434. [PMID: 36925710 PMCID: PMC10013824 DOI: 10.1002/btm2.10434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/09/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
This work reports on an engineered cell that-when electrically stimulated-synthesizes a desired protein, that is, ES-Biofactory. The platform has been used to express interferon (IFN)-β as a universal antiviral protein. Compelling evidence indicates the inevitability of new pandemics and drives the need for a pan-viral intervention that may be quickly deployed while more specific vaccines are in development. Toward this goal, a fast-growing mammalian cell (Chassis) has been engineered with multiple synthetic elements. These include-(1) a voltage-gated Ca2+ channel (Voltage-Sensor) that, upon sensing the electric field, activates the (2) Ca2+-mediated signaling pathway (Actuator) to upregulate (3) IFN-β, via an engineered antiviral transgene (Effector), that is, ES-Biofactory➔IFN-β. The antiviral effects of the ES-Biofactory➔IFN-β have been validated on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected cells. The irradiated ES-Biofactory, that does not exhibit oncogenic capacity, continues to exert antiviral effect. The resulting ES-Biofactory➔IFN-β uses a novel signaling pathway that, unlike the natural IFN synthesis pathway, is not subject to viral interference. Once clinically validated, the ES-Biofactory will be a universal antiviral cell therapy that can be immediately deployed in the event of an outbreak. The platform may also be useful in treating other diseases including cancer and autoimmune disorders.
Collapse
Affiliation(s)
- Sherri Newmyer
- Biosciences DivisionSRI InternationalMenlo ParkCaliforniaUSA
| | | | | | | | | |
Collapse
|
21
|
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: 5] [Impact Index Per Article: 2.5] [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
|
22
|
Cunningham KL, Littleton JT. Mechanisms controlling the trafficking, localization, and abundance of presynaptic Ca 2+ channels. Front Mol Neurosci 2023; 15:1116729. [PMID: 36710932 PMCID: PMC9880069 DOI: 10.3389/fnmol.2022.1116729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/26/2022] [Indexed: 01/14/2023] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) mediate Ca2+ influx to trigger neurotransmitter release at specialized presynaptic sites termed active zones (AZs). The abundance of VGCCs at AZs regulates neurotransmitter release probability (Pr ), a key presynaptic determinant of synaptic strength. Given this functional significance, defining the processes that cooperate to establish AZ VGCC abundance is critical for understanding how these mechanisms set synaptic strength and how they might be regulated to control presynaptic plasticity. VGCC abundance at AZs involves multiple steps, including channel biosynthesis (transcription, translation, and trafficking through the endomembrane system), forward axonal trafficking and delivery to synaptic terminals, incorporation and retention at presynaptic sites, and protein recycling. Here we discuss mechanisms that control VGCC abundance at synapses, highlighting findings from invertebrate and vertebrate models.
Collapse
Affiliation(s)
- Karen L. Cunningham
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | | |
Collapse
|
23
|
Abstract
The CACNA1C gene encodes the pore-forming subunit of the CaV1.2 L-type Ca2+ channel, a critical component of membrane physiology in multiple tissues, including the heart, brain, and immune system. As such, mutations altering the function of these channels have the potential to impact a wide array of cellular functions. The first mutations identified within CACNA1C were shown to cause a severe, multisystem disorder known as Timothy syndrome (TS), which is characterized by neurodevelopmental deficits, long-QT syndrome, life-threatening cardiac arrhythmias, craniofacial abnormalities, and immune deficits. Since this initial description, the number and variety of disease-associated mutations identified in CACNA1C have grown tremendously, expanding the range of phenotypes observed in affected patients. CACNA1C channelopathies are now known to encompass multisystem phenotypes as described in TS, as well as more selective phenotypes where patients may exhibit predominantly cardiac or neurological symptoms. Here, we review the impact of genetic mutations on CaV1.2 function and the resultant physiological consequences.
Collapse
Affiliation(s)
- Kevin G Herold
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - John W Hussey
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ivy E Dick
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
24
|
Patel S, Zissimopoulos S, Marchant JS. Endo-Lysosomal Two-Pore Channels and Their Protein Partners. Handb Exp Pharmacol 2023; 278:199-214. [PMID: 35902438 DOI: 10.1007/164_2022_601] [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: 04/28/2023]
Abstract
Two-pore channels are ion channels expressed on acidic organelles such as the various vesicles that constitute the endo-lysosomal system. They are permeable to Ca2+ and Na+ and activated by the second messenger NAADP as well as the phosphoinositide, PI(3,5)P2 and/or voltage. Here, we review the proteins that interact with these channels including recently identified NAADP receptors.
Collapse
Affiliation(s)
- Sandip Patel
- Department of Cell and Developmental Biology, University College London, London, UK.
| | | | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
25
|
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
|
26
|
Goyal S, Goyal S, Goins AE, Alles SR. Plant-derived natural products targeting ion channels for pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 13:100128. [PMID: 37151956 PMCID: PMC10160805 DOI: 10.1016/j.ynpai.2023.100128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/27/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023]
Abstract
Chronic pain affects approximately one-fifth of people worldwide and reduces quality of life and in some cases, working ability. Ion channels expressed along nociceptive pathways affect neuronal excitability and as a result modulate pain experience. Several ion channels have been identified and investigated as potential targets for new medicines for the treatment of a variety of human diseases, including chronic pain. Voltage-gated channels Na+ and Ca2+ channels, K+ channels, transient receptor potential channels (TRP), purinergic (P2X) channels and acid-sensing ion channels (ASICs) are some examples of ion channels exhibiting altered function or expression in different chronic pain states. Pharmacological approaches are being developed to mitigate dysregulation of these channels as potential treatment options. Since natural compounds of plant origin exert promising biological and pharmacological properties and are believed to possess less adverse effects compared to synthetic drugs, they have been widely studied as treatments for chronic pain for their ability to alter the functional activity of ion channels. A literature review was conducted using Medline, Google Scholar and PubMed, resulted in listing 79 natural compounds/extracts that are reported to interact with ion channels as part of their analgesic mechanism of action. Most in vitro studies utilized electrophysiological techniques to study the effect of natural compounds on ion channels using primary cultures of dorsal root ganglia (DRG) neurons. In vivo studies concentrated on different pain models and were conducted mainly in mice and rats. Proceeding into clinical trials will require further study to develop new, potent and specific ion channel modulators of plant origin.
Collapse
Affiliation(s)
- Sachin Goyal
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87106, USA
| | - Shivali Goyal
- School of Pharmacy, Abhilashi University, Chail Chowk, Mandi, HP 175045, India
| | - Aleyah E. Goins
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87106, USA
| | - Sascha R.A. Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87106, USA
- Corresponding author.
| |
Collapse
|
27
|
Reyes Fernandez PC, Wright CS, Warden SJ, Hum J, Farach-Carson MC, Thompson WR. Effects of Gabapentin and Pregabalin on Calcium Homeostasis: Implications for Physical Rehabilitation of Musculoskeletal Tissues. Curr Osteoporos Rep 2022; 20:365-378. [PMID: 36149592 PMCID: PMC10108402 DOI: 10.1007/s11914-022-00750-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 01/30/2023]
Abstract
PURPOSE OF REVIEW In this review, we discuss the mechanism of action of gabapentinoids and the potential consequences of long-term treatment with these drugs on the musculoskeletal system. RECENT FINDINGS Gabapentinoids, such as gabapentin (GBP) and pregabalin (PGB) were designed as antiepileptic reagents and are now commonly used as first-line treatment for neuropathic pain and increasingly prescribed off-label for other pain disorders such as migraines and back pain. GBP and PGB exert their analgesic actions by selectively binding the α2δ1 auxiliary subunit of voltage-sensitive calcium channels, thereby inhibiting channel function. Numerous tissues express the α2δ1 subunit where GBP and PGB can alter calcium-mediated signaling events. In tissues such as bone, muscle, and cartilage, α2δ1 has important roles in skeletal formation, mechanosensation, and normal tissue function/repair that may be affected by chronic use of gabapentinoids. Long-term use of gabapentinoids is associated with detrimental musculoskeletal outcomes, including increased fracture risk. Therefore, understanding potential complications is essential for clinicians to guide appropriate treatments.
Collapse
Affiliation(s)
- Perla C Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Christian S Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Stuart J Warden
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Julia Hum
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
- College of Osteopathic Medicine, Marian University, Indianapolis, IN, 4622, 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 Human Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- College of Osteopathic Medicine, Marian University, Indianapolis, IN, 4622, USA.
- Department of Anatomy and Cell Biology, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
28
|
Depleted Calcium Stores and Increased Calcium Entry in Rod Photoreceptors of the Cacna2d4 Mouse Model of Cone-Rod Dystrophy RCD4. Int J Mol Sci 2022; 23:ijms232113080. [DOI: 10.3390/ijms232113080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Unidentified pathogenetic mechanisms and genetic and clinical heterogeneity represent critical factors hindering the development of treatments for inherited retinal dystrophies. Frameshift mutations in Cacna2d4, which codes for an accessory subunit of voltage-gated calcium channels (VGCC), cause cone-rod dystrophy RCD4 in patients, but the underlying mechanisms remain unknown. To define its pathogenetic mechanisms, we investigated the impact of a Cacna2d4 frameshift mutation on the electrophysiological profile and calcium handling of mouse rod photoreceptors by patch-clamp recordings and calcium imaging, respectively. In mutant (MUT) rods, the dysregulation of calcium handling extends beyond the reduction in calcium entry through VGCC and surprisingly involves internal calcium stores’ depletion and upregulation of calcium entry via non-selective cationic channels (CSC). The similar dependence of CSC on basal calcium levels in WT and MUT rods suggests that the primary defect in MUT rods lies in defective calcium stores. Calcium stores’ depletion, leading to upregulated calcium and sodium influx via CSC, represents a novel and, so far, unsuspected consequence of the Cacna2d4 mutation. Blocking CSC may provide a novel strategy to counteract the well-known pathogenetic mechanisms involved in rod demise, such as the reticulum stress response and calcium and sodium overload due to store depletion.
Collapse
|
29
|
Aguiar F, Rhana P, Bloise E, Rodrigues ALP, Ferreira E. L-type voltage-dependent Ca2+ channels expression involved in pre-neoplastic transformation of breast cancer. SURGICAL AND EXPERIMENTAL PATHOLOGY 2022. [DOI: 10.1186/s42047-022-00117-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Intracellular Ca2+ levels can modulate several cellular functions, including proliferation and other processes found altered in neoplastic cells. Helping to maintain Ca2+ homeostasis, L-type voltage-dependent Ca2+ channels had its expression identified in neoplasias, including breast cancer. Invasive breast carcinoma of no special type, the most common classification of breast cancer, has ductal hyperplasia and ductal carcinoma in situ as its possible non-obligate precursors. This channel’s role in breast cancer development from these precursors has not been investigated. Evaluate protein expression and subcellular localization of CaV1.1, CaV1.2, and CaV1.3 in mammary epithelium without alteration and neoplastic and non-neoplastic ductal proliferative lesions through immunohistochemistry was the aim of this investigation.
Methods
In the present study, CaV1.1, CaV1.2, and CaV1.3 protein expression was evaluated by immunohistochemistry in breast without alteration and in proliferative non-neoplastic and neoplastic ductal epithelial lesions of the human breast.
Results
It was observed that CaV1.3 presented a reduction in nuclear expression at neoplastic lesions, in addition to an increase in cytoplasmic CaV1.1 expression. The analyses of membrane immunostaining showed that CaV1.2 and CaV1.3 had an increase of expression as the lesions progressed in the stages leading to invasive carcinomas.
Conclusions
Changes in protein expression and subcellular localization of these channels during the progression stages indicate that they may be involved in neoplastic transformation.
Collapse
|
30
|
Li X, Wang B, Yu N, Yang L, Nan C, Sun Z, Guo L, Zhao Z. Gabapentin Alleviates Brain Injury in Intracerebral Hemorrhage Through Suppressing Neuroinflammation and Apoptosis. Neurochem Res 2022; 47:3063-3075. [PMID: 35809188 DOI: 10.1007/s11064-022-03657-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 11/29/2022]
Abstract
Neuroinflammation plays an important role in brain tissue injury during intracerebral hemorrhage. Gabapentin can reduce inflammation and oxidative stress through inhibiting nuclear factor κB (NFκB) signals. Here, we showed that gabapentin reduced brain tissue injury in ICH through suppressing NFκB-mediated neuroinflammation. ICH was induced by injecting collagenase IV into the right striatum of Sprague-Dawley rats. PC12 and BV2 cells injury induced by Hemin were used to simulate ICH in vitro. Inflammation and apoptosis were assessed in rat brain tissue and in vitro cells. The neurobehavioral scores were significantly decreased in ICH rats compared with sham rats. Phosphorylated IκB-α and cleaved caspase3, and apoptosis rate were significantly higher in tissue surrounding the hematoma than in brain tissues from rats subjected to sham surgery. Furthermore, serum IL-6 levels in ICH rats were higher than in sham rats. Gabapentin treatment significantly improved the behavioral scores, decreased levels of phosphorylated IκB-α and cleaved caspase3, apoptosis rate, and serum IL-6 level in ICH rats. Hemin-treated BV2 cells displayed higher levels of phosphorylated IκB-α, cleaved caspase3, and IL-6 in the supernatant compared with vehicle-treated cells. Hemin treatment induced a significantly lower level of peroxisome proliferator-activated receptor γ (PPARγ) in BV2 cells. BV2-PC12 co-culture cells treated by hemin displayed higher levels of cleaved caspase3 in PC12 cells. Furthermore, gabapentin treatment could reduce these effects induced by hemin and the protective effects of gabapentin were significantly attenuated by PPARγ inhibitor. Therefore, gabapentin may reduce inflammation and apoptosis induced by the ICH through PPARγ-NFκB pathway.
Collapse
Affiliation(s)
- Xiaopeng Li
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China.,Department of Neurosurgery, The First Hospital of Handan City, Handan, 056000, HeBei, China
| | - Bingqian Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China.,Department of Neurosurgery, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, 054000, HeBei, China
| | - Ning Yu
- Department of Anesthesiology and Intensive Care Unit, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, HeBei, China
| | - Liang Yang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China
| | - Chengrui Nan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China
| | - Zhimin Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China.,Department of Neurosurgery, The Third Hospital of Shijiazhuang City, Shijiazhuang, 050000, HeBei, China
| | - Lisi Guo
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijiazhuang, 050000, HeBei, China.
| |
Collapse
|
31
|
Wu J, Tan Y, Kang D, Yu J, Qi J, Wu J, Zhang M. Xiaoyu Jiangzhi capsule protects against heart failure via Ca2+/CaMKII signaling pathways in mice. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2022. [DOI: 10.1016/j.jtcms.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
32
|
Largeau B, Bordy R, Pasqualin C, Bredeloux P, Cracowski JL, Lengellé C, Gras-Champel V, Auffret M, Maupoil V, Jonville-Béra AP. Gabapentinoid-induced peripheral edema and acute heart failure: A translational study combining pharmacovigilance data and in vitro animal experiments. Biomed Pharmacother 2022; 149:112807. [PMID: 35303569 DOI: 10.1016/j.biopha.2022.112807] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/20/2022] [Accepted: 03/07/2022] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Gabapentinoids are ligands of the α2-δ subunit of voltage-gated calcium channels (Cav) that have been associated with a risk of peripheral edema and acute heart failure in connection with a potentially dual mechanism, vascular and cardiac. OBJECTIVES & METHODS All cases of peripheral edema or heart failure involving gabapentin or pregabalin reported to the French Pharmacovigilance Centers between January 1, 1994 and April 30, 2020 were included to describe their onset patterns (e.g., time to onset). Based on these data, we investigated the impact of gabapentinoids on the myogenic tone of rat third-order mesenteric arteries and on the electrophysiological properties of rat ventricular cardiomyocytes. RESULTS A total of 58 reports were included (gabapentin n = 5, pregabalin n = 53). The female-to-male ratio was 4:1 and the median age was 77 years (IQR 57-85, range 32-95). The median time to onset were 23 days (IQR 10-54) and 17 days (IQR 3-30) for non-cardiogenic edema and acute heart failure, respectively. Cardiogenic and non-cardiogenic peripheral edema occurred frequently after a dose escalation (27/45, 60%), and the course was rapidly favorable after discontinuation of gabapentinoid (median 7 days, IQR 5-13). On rat mesenteric arteries, gabapentinoids significantly decreased the myogenic tone to the same extent as verapamil and nifedipine. Acute application of gabapentinoids had no significant effect on Cav1.2 currents of ventricular cardiomyocytes. CONCLUSION Gabapentinoids can cause concentration-dependent peripheral edema of early onset. The primary mechanism of non-cardiogenic peripheral edema is vasodilatory edema secondary to altered myogenic tone, independent of Cav1.2 blockade under the experimental conditions tested.
Collapse
Affiliation(s)
- Bérenger Largeau
- CHRU de Tours, Service de Pharmacosurveillance, Centre Régional de Pharmacovigilance Centre-Val de Loire, Tours 37044, France.
| | - Romain Bordy
- Université de Tours, Transplantation, Immunologie et Inflammation (T2I) - EA4245, Tours 37044, France.
| | - Côme Pasqualin
- Université de Tours, Transplantation, Immunologie et Inflammation (T2I) - EA4245, Tours 37044, France.
| | - Pierre Bredeloux
- Université de Tours, Transplantation, Immunologie et Inflammation (T2I) - EA4245, Tours 37044, France.
| | - Jean-Luc Cracowski
- CHU Grenoble Alpes, Centre Régional de Pharmacovigilance et d'Information sur les Médicaments, Grenoble 38000, France; University of Grenoble HP2, INSERM, Grenoble, 38000, France.
| | - Céline Lengellé
- CHRU de Tours, Service de Pharmacosurveillance, Centre Régional de Pharmacovigilance Centre-Val de Loire, Tours 37044, France.
| | - Valérie Gras-Champel
- CHU d'Amiens, Service de Pharmacologie Clinique, Centre Régional de Pharmacovigilance d'Amiens, Amiens 80054, France.
| | - Marine Auffret
- Hospices Civils de Lyon, Service Hospitalo-Universitaire de Pharmacotoxicologie, Centre Régional de Pharmacovigilance, Lyon, France.
| | - Véronique Maupoil
- Université de Tours, Transplantation, Immunologie et Inflammation (T2I) - EA4245, Tours 37044, France.
| | - Annie-Pierre Jonville-Béra
- CHRU de Tours, Service de Pharmacosurveillance, Centre Régional de Pharmacovigilance Centre-Val de Loire, Tours 37044, France; Université de Tours, Université de Nantes, INSERM, methodS in Patients-centered outcomes and HEalth ResEarch (SPHERE) - UMR 1246, Tours 37044, France.
| |
Collapse
|
33
|
IP3-dependent Ca2+ signals are tightly controlled by Cavβ3, but not by Cavβ1, 2 and 4. Cell Calcium 2022; 104:102573. [DOI: 10.1016/j.ceca.2022.102573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/17/2022] [Accepted: 03/15/2022] [Indexed: 11/18/2022]
|
34
|
Research Progress on Neuroprotection of Insulin-like Growth Factor-1 towards Glutamate-Induced Neurotoxicity. Cells 2022; 11:cells11040666. [PMID: 35203315 PMCID: PMC8870287 DOI: 10.3390/cells11040666] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/05/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) and its binding proteins and receptors are widely expressed in the central nervous system (CNS), proposing IGF-1-induced neurotrophic actions in normal growth, development, and maintenance. However, while there is convincing evidence that the IGF-1 system has specific endocrine roles in the CNS, the concept is emerging that IGF-I might be also important in disorders such as ischemic stroke, brain trauma, Alzheimer’s disease, epilepsy, etc., by inducing neuroprotective effects towards glutamate-mediated excitotoxic signaling pathways. Research in rodent models has demonstrated rescue of pathophysiological and behavioral abnormalities when IGF-1 was administered by different routes, and several clinical studies have shown safety and promise of efficacy in neurological disorders of the CNS. Focusing on the relationship between IGF-1-induced neuroprotection and glutamate-induced excitatory neurotoxicity, this review addresses the research progress in the field, intending to provide a rationale for using IGF-I clinically to confer neuroprotective therapy towards neurological diseases with glutamate excitotoxicity as a common pathological pathway.
Collapse
|
35
|
Zheng YL, Su X, Chen YM, Guo JB, Song G, Yang Z, Chen PJ, Wang XQ. microRNA-Based Network and Pathway Analysis for Neuropathic Pain in Rodent Models. Front Mol Biosci 2022; 8:780730. [PMID: 35096965 PMCID: PMC8794747 DOI: 10.3389/fmolb.2021.780730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/27/2021] [Indexed: 12/03/2022] Open
Abstract
Neuropathic pain (NP) is poorly managed, and in-depth mechanisms of gene transcriptome alterations in NP pathogenesis are not yet fully understood. To determine microRNA-related molecular mechanisms of NP and their transcriptional regulation in NP, PubMed, Embase, Web of Science and CINAHL Complete (EBSCO) were searched from inception to April 2021. Commonly dysregulated miRNAs in NP were assessed. The putative targets of these miRNAs were determined using TargetScan, Funrich, Cytoscape and String database. A total of 133 literatures containing miRNA profiles studies and experimentally verify studies were included. Venn analysis, target gene prediction analysis and functional enrichment analysis indicated several miRNAs (miR-200b-3p, miR-96, miR-182, miR-183, miR-30b, miR-155 and miR-145) and their target genes involved in known relevant pathways for NP. Targets on transient receptor potential channels, voltage-gated sodium channels and voltage-gated calcium channels may be harnessed for pain relief. A further delineation of signal processing and modulation in neuronal ensembles is key to achieving therapeutic success in future studies.
Collapse
Affiliation(s)
- Yi-Li Zheng
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Xuan Su
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Yu-Meng Chen
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Jia-Bao Guo
- The Second School of Clinical Medical, Xuzhou Medical University, Xuzhou, China
| | - Ge Song
- Department of Rehabilitation Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Yang
- Department of Rehabilitation Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Pei-Jie Chen
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- *Correspondence: Pei-Jie Chen, ; Xue-Qiang Wang,
| | - Xue-Qiang Wang
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai Shangti Orthopaedic Hospital, Shanghai, China
- *Correspondence: Pei-Jie Chen, ; Xue-Qiang Wang,
| |
Collapse
|
36
|
Wright DJ, Hall NAL, Irish N, Man AL, Glynn W, Mould A, Angeles ADL, Angiolini E, Swarbreck D, Gharbi K, Tunbridge EM, Haerty W. Long read sequencing reveals novel isoforms and insights into splicing regulation during cell state changes. BMC Genomics 2022; 23:42. [PMID: 35012468 PMCID: PMC8744310 DOI: 10.1186/s12864-021-08261-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 12/15/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Alternative splicing is a key mechanism underlying cellular differentiation and a driver of complexity in mammalian neuronal tissues. However, understanding of which isoforms are differentially used or expressed and how this affects cellular differentiation remains unclear. Long read sequencing allows full-length transcript recovery and quantification, enabling transcript-level analysis of alternative splicing processes and how these change with cell state. Here, we utilise Oxford Nanopore Technologies sequencing to produce a custom annotation of a well-studied human neuroblastoma cell line SH-SY5Y, and to characterise isoform expression and usage across differentiation. RESULTS We identify many previously unannotated features, including a novel transcript of the voltage-gated calcium channel subunit gene, CACNA2D2. We show differential expression and usage of transcripts during differentiation identifying candidates for future research into state change regulation. CONCLUSIONS Our work highlights the potential of long read sequencing to uncover previously unknown transcript diversity and mechanisms influencing alternative splicing.
Collapse
Affiliation(s)
- David J Wright
- Earlham Institute, Norwich Research Park, Norfolk, NR4 7UZ, UK
| | - Nicola A L Hall
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxfordshire, OX3 3JX, UK
- Oxford Health, NHS Foundation Trust, Oxford, Oxfordshire, OX3 7JX, UK
| | - Naomi Irish
- Earlham Institute, Norwich Research Park, Norfolk, NR4 7UZ, UK
| | - Angela L Man
- Earlham Institute, Norwich Research Park, Norfolk, NR4 7UZ, UK
| | - Will Glynn
- Earlham Institute, Norwich Research Park, Norfolk, NR4 7UZ, UK
| | - Arne Mould
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxfordshire, OX3 3JX, UK
- Oxford Health, NHS Foundation Trust, Oxford, Oxfordshire, OX3 7JX, UK
| | - Alejandro De Los Angeles
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxfordshire, OX3 3JX, UK
- Oxford Health, NHS Foundation Trust, Oxford, Oxfordshire, OX3 7JX, UK
| | - Emily Angiolini
- Earlham Institute, Norwich Research Park, Norfolk, NR4 7UZ, UK
| | - David Swarbreck
- Earlham Institute, Norwich Research Park, Norfolk, NR4 7UZ, UK
| | - Karim Gharbi
- Earlham Institute, Norwich Research Park, Norfolk, NR4 7UZ, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxfordshire, OX3 3JX, UK
- Oxford Health, NHS Foundation Trust, Oxford, Oxfordshire, OX3 7JX, UK
| | - Wilfried Haerty
- Earlham Institute, Norwich Research Park, Norfolk, NR4 7UZ, UK.
| |
Collapse
|
37
|
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: 50] [Impact Index Per Article: 16.7] [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
|
38
|
Dixon RE, Navedo MF, Binder MD, Santana LF. Mechanisms and Physiological Implications of Cooperative Gating of Ion Channels Clusters. Physiol Rev 2021; 102:1159-1210. [PMID: 34927454 DOI: 10.1152/physrev.00022.2021] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive Cav1.2 and Cav1.3 channels to obligatory dimeric assembly and gating of voltage-gated Nav1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pace-making activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.
Collapse
Affiliation(s)
- Rose Ellen Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| |
Collapse
|
39
|
Fernández A, Díaz JL, García M, Rodríguez-Escrich S, Lorente A, Enrech R, Dordal A, Portillo-Salido E, Porras M, Fernández B, Reinoso RF, Vela JM, Almansa C. Piperazinyl Bicyclic Derivatives as Selective Ligands of the α2δ-1 Subunit of Voltage-Gated Calcium Channels. ACS Med Chem Lett 2021; 12:1802-1809. [PMID: 34795870 DOI: 10.1021/acsmedchemlett.1c00416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/01/2021] [Indexed: 12/14/2022] Open
Abstract
The synthesis and pharmacological activities of a new series of piperazinyl quinazolin-4-(3H)-one derivatives acting toward the α2δ-1 subunit of voltage-gated calcium channels (Cavα2δ-1) are reported. Different positions of a micromolar HTS hit were explored, and best activities were obtained for compounds containing a small alkyl group in position 3 of the quinazolin-4-(3H)-one scaffold and a 3-methyl-piperazin-1-yl- or 3,5-dimethyl-piperazin-1-yl-butyl group in position 2. The activity was shown to reside in the R enantiomer of the chain in position 2, and several eutomers reached single digit nanomolar affinities. Final modification of the central scaffold to reduce lipophilicity provided the pyrido[4,3-d]pyrimidin-4(3H)-one 16RR, which showed high selectivity for Cavα2δ-1 versus Cavα2δ-2, probably linked to its improved analgesic efficacy-safety ratio in mice over pregabalin.
Collapse
Affiliation(s)
- Ariadna Fernández
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, 08028 Barcelona, Spain
| | - José Luis Díaz
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, 08028 Barcelona, Spain
| | - Mónica García
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, 08028 Barcelona, Spain
| | | | - Adriana Lorente
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, 08028 Barcelona, Spain
| | - Raquel Enrech
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, 08028 Barcelona, Spain
| | - Albert Dordal
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, 08028 Barcelona, Spain
| | | | - Mónica Porras
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, 08028 Barcelona, Spain
| | - Begoña Fernández
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, 08028 Barcelona, Spain
| | - Raquel F. Reinoso
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, 08028 Barcelona, Spain
| | - José Miguel Vela
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, 08028 Barcelona, Spain
| | - Carmen Almansa
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, 08028 Barcelona, Spain
| |
Collapse
|
40
|
Gomez K, Vargas-Parada A, Duran P, Sandoval A, Delgado-Lezama R, Khanna R, Felix R. L5-6 Spinal Nerve Ligation-induced Neuropathy Changes the Location and Function of Ca 2+ Channels and Cdk5 and Affects the Compound Action Potential in Adjacent Intact L4 Afferent Fibers. Neuroscience 2021; 471:20-31. [PMID: 34303780 PMCID: PMC8384716 DOI: 10.1016/j.neuroscience.2021.07.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/30/2021] [Accepted: 07/14/2021] [Indexed: 01/07/2023]
Abstract
Voltage-gated Ca2+ (CaV) channels regulate multiple cell processes, including neurotransmitter release, and have been associated with several pathological conditions, such as neuropathic pain. Cdk5, a neuron-specific kinase, may phosphorylate CaV channels, altering their functional expression. During peripheral nerve injury, upregulation of CaV channels and Cdk5 in the dorsal root ganglia (DRG) and the spinal cord, has been correlated with allodynia. We recently reported an increase in the amplitude of the C component of the compound action potential (cAP) of afferent fibers in animals with allodynia induced by L5-6 spinal nerve ligation (SNL), recorded in the corresponding dorsal roots. This was related to an increase in T-type (CaV3.2) channels generated by Cdk5-mediated phosphorylation. Here, we show that CaV channel functional expression is also altered in the L4 adjacent intact afferent fibers in rats with allodynia induced by L5-6 SNL. Western blot analysis showed that both Cdk5 and CaV3.2 total levels are not increased in the DRG L3-4, but their subcellular distribution changes by concentrating on the neuronal soma. Likewise, the Cdk5 inhibitor olomoucine affected the rapid and the slow C components of the cAP recorded in the dorsal roots. Patch-clamp recordings revealed an increase in T- and N-type currents recorded in the soma of acute isolated L3-4 sensory neurons after L5-6 SNL, which was prevented by olomoucine. These findings suggest changes in CaV channels location and function in L3-4 afferent fibers associated with Cdk5-mediated phosphorylation after L5-6 SNL, which may contribute to nerve injury-induced allodynia.
Collapse
Affiliation(s)
- Kimberly Gomez
- Department of Physiology, Biophysics and Neuroscience, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Alberto Vargas-Parada
- Department of Physiology, Biophysics and Neuroscience, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Paz Duran
- Department of Cell Biology, Cinvestav, Mexico City, Mexico
| | - Alejandro Sandoval
- School of Medicine FES Iztacala, National Autonomous University of Mexico (UNAM), Tlalnepantla, Mexico
| | - Rodolfo Delgado-Lezama
- Department of Physiology, Biophysics and Neuroscience, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Ricardo Felix
- Department of Cell Biology, Cinvestav, Mexico City, Mexico.
| |
Collapse
|
41
|
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
|
42
|
Weiss N, Zamponi GW. Opioid Receptor Regulation of Neuronal Voltage-Gated Calcium Channels. Cell Mol Neurobiol 2021; 41:839-847. [PMID: 32514826 PMCID: PMC11448596 DOI: 10.1007/s10571-020-00894-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 12/28/2022]
Abstract
Neuronal voltage-gated calcium channels play a pivotal role in the conversion of electrical signals into calcium entry into nerve endings that is required for the release of neurotransmitters. They are under the control of a number of cellular signaling pathways that serve to fine tune synaptic activities, including G-protein coupled receptors (GPCRs) and the opioid system. Besides modulating channel activity via activation of second messengers, GPCRs also physically associate with calcium channels to regulate their function and expression at the plasma membrane. In this mini review, we discuss the mechanisms by which calcium channels are regulated by classical opioid and nociceptin receptors. We highlight the importance of this regulation in the control of neuronal functions and their implication in the development of disease conditions. Finally, we present recent literature concerning the use of novel μ-opioid receptor/nociceptin receptor modulators and discuss their use as potential drug candidates for the treatment of pain.
Collapse
Affiliation(s)
- Norbert Weiss
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
43
|
Wang X, Burke SRA, Talmadge RJ, Voss AA, Rich MM. Depressed neuromuscular transmission causes weakness in mice lacking BK potassium channels. J Gen Physiol 2021; 152:151617. [PMID: 32243496 PMCID: PMC7201880 DOI: 10.1085/jgp.201912526] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/27/2020] [Accepted: 03/02/2020] [Indexed: 12/20/2022] Open
Abstract
Mice lacking functional large-conductance voltage- and Ca2+-activated K+ channels (BK channels) are viable but have motor deficits including ataxia and weakness. The cause of weakness is unknown. In this study, we discovered, in vivo, that skeletal muscle in mice lacking BK channels (BK−/−) was weak in response to nerve stimulation but not to direct muscle stimulation, suggesting a failure of neuromuscular transmission. Voltage-clamp studies of the BK−/− neuromuscular junction (NMJ) revealed a reduction in evoked endplate current amplitude and the frequency of spontaneous vesicle release compared with WT littermates. Responses to 50-Hz stimulation indicated a reduced probability of vesicle release in BK−/− mice, suggestive of lower presynaptic Ca2+ entry. Pharmacological block of BK channels in WT NMJs did not affect NMJ function, surprisingly suggesting that the reduced vesicle release in BK−/− NMJs was not due to loss of BK channel–mediated K+ current. Possible explanations for our data include an effect of BK channels on development of the NMJ, a role for BK channels in regulating presynaptic Ca2+ current or the effectiveness of Ca2+ in triggering release. Consistent with reduced Ca2+ entry or effectiveness of Ca2+ in triggering release, use of 3,4-diaminopyridine to widen action potentials normalized evoked release in BK−/− mice to WT levels. Intraperitoneal application of 3,4-diaminopyridine fully restored in vivo nerve-stimulated muscle force in BK−/− mice. Our work demonstrates that mice lacking BK channels have weakness due to a defect in vesicle release at the NMJ.
Collapse
Affiliation(s)
- Xueyong Wang
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH
| | - Steven R A Burke
- Department of Biological Sciences, Wright State University, Dayton, OH
| | - Robert J Talmadge
- Department of Biological Sciences, California State Polytechnic University, Pomona, Pomona, CA
| | - Andrew A Voss
- Department of Biological Sciences, Wright State University, Dayton, OH
| | - Mark M Rich
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH
| |
Collapse
|
44
|
Kessi M, Chen B, Peng J, Yan F, Yang L, Yin F. Calcium channelopathies and intellectual disability: a systematic review. Orphanet J Rare Dis 2021; 16:219. [PMID: 33985586 PMCID: PMC8120735 DOI: 10.1186/s13023-021-01850-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/04/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Calcium ions are involved in several human cellular processes including corticogenesis, transcription, and synaptogenesis. Nevertheless, the relationship between calcium channelopathies (CCs) and intellectual disability (ID)/global developmental delay (GDD) has been poorly investigated. We hypothesised that CCs play a major role in the development of ID/GDD and that both gain- and loss-of-function variants of calcium channel genes can induce ID/GDD. As a result, we performed a systematic review to investigate the contribution of CCs, potential mechanisms underlying their involvement in ID/GDD, advancements in cell and animal models, treatments, brain anomalies in patients with CCs, and the existing gaps in the knowledge. We performed a systematic search in PubMed, Embase, ClinVar, OMIM, ClinGen, Gene Reviews, DECIPHER and LOVD databases to search for articles/records published before March 2021. The following search strategies were employed: ID and calcium channel, mental retardation and calcium channel, GDD and calcium channel, developmental delay and calcium channel. MAIN BODY A total of 59 reports describing 159 cases were found in PubMed, Embase, ClinVar, and LOVD databases. Variations in ten calcium channel genes including CACNA1A, CACNA1C, CACNA1I, CACNA1H, CACNA1D, CACNA2D1, CACNA2D2, CACNA1E, CACNA1F, and CACNA1G were found to be associated with ID/GDD. Most variants exhibited gain-of-function effect. Severe to profound ID/GDD was observed more for the cases with gain-of-function variants as compared to those with loss-of-function. CACNA1E, CACNA1G, CACNA1F, CACNA2D2 and CACNA1A associated with more severe phenotype. Furthermore, 157 copy number variations (CNVs) spanning calcium genes were identified in DECIPHER database. The leading genes included CACNA1C, CACNA1A, and CACNA1E. Overall, the underlying mechanisms included gain- and/ or loss-of-function, alteration in kinetics (activation, inactivation) and dominant-negative effects of truncated forms of alpha1 subunits. Forty of the identified cases featured cerebellar atrophy. We identified only a few cell and animal studies that focused on the mechanisms of ID/GDD in relation to CCs. There is a scarcity of studies on treatment options for ID/GDD both in vivo and in vitro. CONCLUSION Our results suggest that CCs play a major role in ID/GDD. While both gain- and loss-of-function variants are associated with ID/GDD, the mechanisms underlying their involvement need further scrutiny.
Collapse
Affiliation(s)
- Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China
- Kilimanjaro Christian Medical University College, Moshi, Tanzania
- Mawenzi Regional Referral Hospital, Moshi, Tanzania
| | - Baiyu Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China
| | - Fangling Yan
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China.
| |
Collapse
|
45
|
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
|
46
|
Schöpf CL, Ablinger C, Geisler SM, Stanika RI, Campiglio M, Kaufmann WA, Nimmervoll B, Schlick B, Brockhaus J, Missler M, Shigemoto R, Obermair GJ. Presynaptic α 2δ subunits are key organizers of glutamatergic synapses. Proc Natl Acad Sci U S A 2021; 118:e1920827118. [PMID: 33782113 PMCID: PMC8040823 DOI: 10.1073/pnas.1920827118] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In nerve cells the genes encoding for α2δ subunits of voltage-gated calcium channels have been linked to synaptic functions and neurological disease. Here we show that α2δ subunits are essential for the formation and organization of glutamatergic synapses. Using a cellular α2δ subunit triple-knockout/knockdown model, we demonstrate a failure in presynaptic differentiation evidenced by defective presynaptic calcium channel clustering and calcium influx, smaller presynaptic active zones, and a strongly reduced accumulation of presynaptic vesicle-associated proteins (synapsin and vGLUT). The presynaptic defect is associated with the downscaling of postsynaptic AMPA receptors and the postsynaptic density. The role of α2δ isoforms as synaptic organizers is highly redundant, as each individual α2δ isoform can rescue presynaptic calcium channel trafficking and expression of synaptic proteins. Moreover, α2δ-2 and α2δ-3 with mutated metal ion-dependent adhesion sites can fully rescue presynaptic synapsin expression but only partially calcium channel trafficking, suggesting that the regulatory role of α2δ subunits is independent from its role as a calcium channel subunit. Our findings influence the current view on excitatory synapse formation. First, our study suggests that postsynaptic differentiation is secondary to presynaptic differentiation. Second, the dependence of presynaptic differentiation on α2δ implicates α2δ subunits as potential nucleation points for the organization of synapses. Finally, our results suggest that α2δ subunits act as transsynaptic organizers of glutamatergic synapses, thereby aligning the synaptic active zone with the postsynaptic density.
Collapse
Affiliation(s)
- Clemens L Schöpf
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Cornelia Ablinger
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Stefanie M Geisler
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
- Department of Pharmacology and Toxicology, University of Innsbruck, A-6020 Innsbruck, Austria
| | - Ruslan I Stanika
- Division of Physiology, Karl Landsteiner University of Health Sciences, A-3500 Krems, Austria
| | - Marta Campiglio
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Walter A Kaufmann
- Institute of Science and Technology Austria, A-3400 Klosterneuburg, Austria
| | - Benedikt Nimmervoll
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Bettina Schlick
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Johannes Brockhaus
- Institute of Anatomy and Molecular Neurobiology, Westfälische Wilhelms University, 48149 Münster, Germany
| | - Markus Missler
- Institute of Anatomy and Molecular Neurobiology, Westfälische Wilhelms University, 48149 Münster, Germany
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria, A-3400 Klosterneuburg, Austria
| | - Gerald J Obermair
- Institute of Physiology, Medical University of Innsbruck, A-6020 Innsbruck, Austria;
- Division of Physiology, Karl Landsteiner University of Health Sciences, A-3500 Krems, Austria
| |
Collapse
|
47
|
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: 15] [Impact Index Per Article: 3.8] [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
|
48
|
Striessnig J. Voltage-Gated Ca 2+-Channel α1-Subunit de novo Missense Mutations: Gain or Loss of Function - Implications for Potential Therapies. Front Synaptic Neurosci 2021; 13:634760. [PMID: 33746731 PMCID: PMC7966529 DOI: 10.3389/fnsyn.2021.634760] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
This review summarizes our current knowledge of human disease-relevant genetic variants within the family of voltage gated Ca2+ channels. Ca2+ channelopathies cover a wide spectrum of diseases including epilepsies, autism spectrum disorders, intellectual disabilities, developmental delay, cerebellar ataxias and degeneration, severe cardiac arrhythmias, sudden cardiac death, eye disease and endocrine disorders such as congential hyperinsulinism and hyperaldosteronism. A special focus will be on the rapidly increasing number of de novo missense mutations identified in the pore-forming α1-subunits with next generation sequencing studies of well-defined patient cohorts. In contrast to likely gene disrupting mutations these can not only cause a channel loss-of-function but can also induce typical functional changes permitting enhanced channel activity and Ca2+ signaling. Such gain-of-function mutations could represent therapeutic targets for mutation-specific therapy of Ca2+-channelopathies with existing or novel Ca2+-channel inhibitors. Moreover, many pathogenic mutations affect positive charges in the voltage sensors with the potential to form gating-pore currents through voltage sensors. If confirmed in functional studies, specific blockers of gating-pore currents could also be of therapeutic interest.
Collapse
Affiliation(s)
- Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
49
|
Young SM, Veeraraghavan P. Presynaptic voltage-gated calcium channels in the auditory brainstem. Mol Cell Neurosci 2021; 112:103609. [PMID: 33662542 DOI: 10.1016/j.mcn.2021.103609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/06/2021] [Accepted: 02/17/2021] [Indexed: 10/22/2022] Open
Abstract
Sound information encoding within the initial synapses in the auditory brainstem requires reliable and precise synaptic transmission in response to rapid and large fluctuations in action potential (AP) firing rates. The magnitude and location of Ca2+ entry through voltage-gated Ca2+ channels (CaV) in the presynaptic terminal are key determinants in triggering AP-mediated release. In the mammalian central nervous system (CNS), the CaV2.1 subtype is the critical subtype for CNS function, since it is the most efficient CaV2 subtype in triggering AP-mediated synaptic vesicle (SV) release. Auditory brainstem synapses utilize CaV2.1 to sustain fast and repetitive SV release to encode sound information. Therefore, understanding the presynaptic mechanisms that control CaV2.1 localization, organization and biophysical properties are integral to understanding auditory processing. Here, we review our current knowledge about the control of presynaptic CaV2 abundance and organization in the auditory brainstem and impact on the regulation of auditory processing.
Collapse
Affiliation(s)
- Samuel M Young
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Otolaryngology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | | |
Collapse
|
50
|
Geisler SM, Benedetti A, Schöpf CL, Schwarzer C, Stefanova N, Schwartz A, Obermair GJ. Phenotypic Characterization and Brain Structure Analysis of Calcium Channel Subunit α 2δ-2 Mutant (Ducky) and α 2δ Double Knockout Mice. Front Synaptic Neurosci 2021; 13:634412. [PMID: 33679366 PMCID: PMC7933509 DOI: 10.3389/fnsyn.2021.634412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/11/2021] [Indexed: 01/19/2023] Open
Abstract
Auxiliary α2δ subunits of voltage-gated calcium channels modulate channel trafficking, current properties, and synapse formation. Three of the four isoforms (α2δ-1, α2δ-2, and α2δ-3) are abundantly expressed in the brain; however, of the available knockout models, only α2δ-2 knockout or mutant mice display an obvious abnormal neurological phenotype. Thus, we hypothesize that the neuronal α2δ isoforms may have partially specific as well as redundant functions. To address this, we generated three distinct α2δ double knockout mouse models by crossbreeding single knockout (α2δ-1 and -3) or mutant (α2δ-2/ducky) mice. Here, we provide a first phenotypic description and brain structure analysis. We found that genotypic distribution of neonatal litters in distinct α2δ-1/-2, α2δ-1/-3, and α2δ-2/-3 breeding combinations did not conform to Mendel's law, suggesting premature lethality of single and double knockout mice. Notably, high occurrences of infant mortality correlated with the absence of specific α2δ isoforms (α2Δ-2 > α2δ-1 > α2δ-3), and was particularly observed in cages with behaviorally abnormal parenting animals of α2δ-2/-3 cross-breedings. Juvenile α2δ-1/-2 and α2δ-2/-3 double knockout mice displayed a waddling gate similar to ducky mice. However, in contrast to ducky and α2δ-1/-3 double knockout animals, α2δ-1/-2 and α2δ-2/-3 double knockout mice showed a more severe disease progression and highly impaired development. The observed phenotypes within the individual mouse lines may be linked to differences in the volume of specific brain regions. Reduced cortical volume in ducky mice, for example, was associated with a progressively decreased space between neurons, suggesting a reduction of total synaptic connections. Taken together, our findings show that α2δ subunits differentially regulate premature survival, postnatal growth, brain development, and behavior, suggesting specific neuronal functions in health and disease.
Collapse
Affiliation(s)
- Stefanie M. Geisler
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Ariane Benedetti
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Clemens L. Schöpf
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Christoph Schwarzer
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Arnold Schwartz
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Gerald J. Obermair
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
- Division Physiology, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| |
Collapse
|