1
|
Guo SJ, Shi YQ, Zheng YN, Liu H, Zheng YL. The Voltage-Gated Calcium Channel α2δ Subunit in Neuropathic Pain. Mol Neurobiol 2025; 62:2561-2572. [PMID: 39136907 DOI: 10.1007/s12035-024-04424-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/06/2024] [Indexed: 01/28/2025]
Abstract
Neuropathic pain (NP) is a chronic pain caused by injury or disease of the somatosensory nervous system, or it can be directly caused by disease. It often presents with clinical features like spontaneous pain, hyperalgesia, and dysesthesia. At present, voltage-gated calcium ion channels (VGCCs) are known to be closely related to the development of NP, especially the α2δ subunit. The α2δ subunit is a regulatory subunit of VGCCs. It exists mainly in the brain and peripheral nervous system, especially in nerve cells, and it plays a crucial part in regulating presynaptic and postsynaptic functions. Furthermore, the α2δ subunit influences neuronal excitation and pain signaling by promoting its expression and localization through binding to VGCC-related subunits. The α2δ subunit is widely used in the management of NP as a target of antiepileptic drugs gabapentin and pregabalin. Although drug therapy is one of the treatments for NP, its clinical application is limited due to the adverse reactions caused by drug therapy. Therefore, further research on the therapeutic target α2δ subunit is needed, and attempts are made to obtain an effective treatment for relieving NP without side effects. This review describes the current associated knowledge on the function of the α2δ subunit in perceiving and modulating NP.
Collapse
Affiliation(s)
- Sheng-Jie Guo
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China
| | - Yu-Qin Shi
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China
| | - Ya-Nan Zheng
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China
| | - Hui Liu
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China
| | - Yi-Li Zheng
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China.
- Department of Rehabilitation Medicine, Shanghai Shangti Orthopaedic Hospital, Shanghai, China.
| |
Collapse
|
2
|
Lauerer RJ, Lerche H. Voltage-gated calcium channels in genetic epilepsies. J Neurochem 2024; 168:3853-3871. [PMID: 37822150 PMCID: PMC11591408 DOI: 10.1111/jnc.15983] [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: 07/08/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
Voltage-gated calcium channels (VGCC) are abundant in the central nervous system and serve a broad spectrum of functions, either directly in cellular excitability or indirectly to regulate Ca2+ homeostasis. Ca2+ ions act as one of the main connections in excitation-transcription coupling, muscle contraction and excitation-exocytosis coupling, including synaptic transmission. In recent years, many genes encoding VGCCs main α or additional auxiliary subunits have been associated with epilepsy. This review sums up the current state of knowledge on disease mechanisms and provides guidance on disease-specific therapies where applicable.
Collapse
Affiliation(s)
- Robert J. Lauerer
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| |
Collapse
|
3
|
Salib AMN, Crane MJ, Lee SH, Wainger BJ, Jamieson AM, Lipscombe D. Interleukin-1α links peripheral Ca V2.2 channel activation to rapid adaptive increases in heat sensitivity in skin. Sci Rep 2024; 14:9051. [PMID: 38643253 PMCID: PMC11032389 DOI: 10.1038/s41598-024-59424-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/10/2024] [Indexed: 04/22/2024] Open
Abstract
Neurons have the unique capacity to adapt output in response to changes in their environment. Within seconds, sensory nerve endings can become hypersensitive to stimuli in response to potentially damaging events. The underlying behavioral response is well studied, but several of the key signaling molecules that mediate sensory hypersensitivity remain unknown. We previously discovered that peripheral voltage-gated CaV2.2 channels in nerve endings in skin are essential for the rapid, transient increase in sensitivity to heat, but not to mechanical stimuli, that accompanies intradermal capsaicin. Here we report that the cytokine interleukin-1α (IL-1α), an alarmin, is necessary and sufficient to trigger rapid heat and mechanical hypersensitivity in skin. Of 20 cytokines screened, only IL-1α was consistently detected in hind paw interstitial fluid in response to intradermal capsaicin and, similar to behavioral sensitivity to heat, IL-1α levels were also dependent on peripheral CaV2.2 channel activity. Neutralizing IL-1α in skin significantly reduced capsaicin-induced changes in hind paw sensitivity to radiant heat and mechanical stimulation. Intradermal IL-1α enhances behavioral responses to stimuli and, in culture, IL-1α enhances the responsiveness of Trpv1-expressing sensory neurons. Together, our data suggest that IL-1α is the key cytokine that underlies rapid and reversible neuroinflammatory responses in skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI, 02912, USA
| | - Meredith J Crane
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Sang Hun Lee
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Brian J Wainger
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Amanda M Jamieson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Diane Lipscombe
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
4
|
Salib AMN, Crane MJ, Lee SH, Wainger BJ, Jamieson AM, Lipscombe D. Interleukin-1α links peripheral Ca V2.2 channel activation to rapid adaptive increases in heat sensitivity in skin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.17.572072. [PMID: 38585803 PMCID: PMC10996502 DOI: 10.1101/2023.12.17.572072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Neurons have the unique capacity to adapt output in response to changes in their environment. Within seconds, sensory nerve endings can become hypersensitive to stimuli in response to potentially damaging events. The underlying behavioral response is well studied, but several of the key signaling molecules that mediate sensory hypersensitivity remain unknown. We previously discovered that peripheral voltage-gated CaV2.2 channels in nerve endings in skin are essential for the rapid, transient increase in sensitivity to heat, but not to mechanical stimuli, that accompanies intradermal capsaicin. Here we report that the cytokine interleukin-1α (IL-1α), an alarmin, is necessary and sufficient to trigger rapid heat and mechanical hypersensitivity in skin. Of 20 cytokines screened, only IL-1α was consistently detected in hind paw interstitial fluid in response to intradermal capsaicin and, similar to behavioral sensitivity to heat, IL-1α levels were also dependent on peripheral CaV2.2 channel activity. Neutralizing IL-1α in skin significantly reduced capsaicin-induced changes in hind paw sensitivity to radiant heat and mechanical stimulation. Intradermal IL-1α enhances behavioral responses to stimuli and, in culture, IL-1α enhances the responsiveness of Trpv1-expressing sensory neurons. Together, our data suggest that IL-1α is the key cytokine that underlies rapid and reversible neuroinflammatory responses in skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Meredith J Crane
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Sang Hun Lee
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Brian J Wainger
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amanda M Jamieson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Diane Lipscombe
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| |
Collapse
|
5
|
Pham T, Hussein T, Calis D, Bischof H, Skrabak D, Cruz Santos M, Maier S, Spähn D, Kalina D, Simonsig S, Ehinger R, Groschup B, Knipper M, Plesnila N, Ruth P, Lukowski R, Matt L. BK channels sustain neuronal Ca 2+ oscillations to support hippocampal long-term potentiation and memory formation. Cell Mol Life Sci 2023; 80:369. [PMID: 37989805 PMCID: PMC10663188 DOI: 10.1007/s00018-023-05016-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/25/2023] [Accepted: 10/24/2023] [Indexed: 11/23/2023]
Abstract
Mutations of large conductance Ca2+- and voltage-activated K+ channels (BK) are associated with cognitive impairment. Here we report that CA1 pyramidal neuron-specific conditional BK knock-out (cKO) mice display normal locomotor and anxiety behavior. They do, however, exhibit impaired memory acquisition and retrieval in the Morris Water Maze (MWM) when compared to littermate controls (CTRL). In line with cognitive impairment in vivo, electrical and chemical long-term potentiation (LTP) in cKO brain slices were impaired in vitro. We further used a genetically encoded fluorescent K+ biosensor and a Ca2+-sensitive probe to observe cultured hippocampal neurons during chemical LTP (cLTP) induction. cLTP massively reduced intracellular K+ concentration ([K+]i) while elevating L-Type Ca2+ channel- and NMDA receptor-dependent Ca2+ oscillation frequencies. Both, [K+]i decrease and Ca2+ oscillation frequency increase were absent after pharmacological BK inhibition or in cells lacking BK. Our data suggest that L-Type- and NMDAR-dependent BK-mediated K+ outflow significantly contributes to hippocampal LTP, as well as learning and memory.
Collapse
Affiliation(s)
- Thomas Pham
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Tamara Hussein
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Dila Calis
- Department of Otolaryngology, Head and Neck Surgery, Molecular Physiology of Hearing, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany
| | - Helmut Bischof
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - David Skrabak
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Melanie Cruz Santos
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Selina Maier
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - David Spähn
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Daniel Kalina
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Stefanie Simonsig
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Rebekka Ehinger
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Bernhard Groschup
- Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
| | - Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Molecular Physiology of Hearing, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany
| | - Nikolaus Plesnila
- Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Lucas Matt
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
6
|
Antunes FTT, Campos MM, Carvalho VDPR, da Silva Junior CA, Magno LAV, de Souza AH, Gomez MV. Current Drug Development Overview: Targeting Voltage-Gated Calcium Channels for the Treatment of Pain. Int J Mol Sci 2023; 24:ijms24119223. [PMID: 37298174 DOI: 10.3390/ijms24119223] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 06/12/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) are targeted to treat pain conditions. Since the discovery of their relation to pain processing control, they are investigated to find new strategies for better pain control. This review provides an overview of naturally based and synthetic VGCC blockers, highlighting new evidence on the development of drugs focusing on the VGCC subtypes as well as mixed targets with pre-clinical and clinical analgesic effects.
Collapse
Affiliation(s)
- Flavia Tasmin Techera Antunes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Maria Martha Campos
- Programa de Pós-Graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre 90619-900, RS, Brazil
| | | | | | - Luiz Alexandre Viana Magno
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade Ciências Médicas de Minas Gerais (FCMMG), Belo Horizonte 30110-005, MG, Brazil
| | - Alessandra Hubner de Souza
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade Ciências Médicas de Minas Gerais (FCMMG), Belo Horizonte 30110-005, MG, Brazil
| | | |
Collapse
|
7
|
Caminski ES, Antunes FTT, Souza IA, Dallegrave E, Zamponi GW. Regulation of N-type calcium channels by nociceptin receptors and its possible role in neurological disorders. Mol Brain 2022; 15:95. [PMID: 36434658 PMCID: PMC9700961 DOI: 10.1186/s13041-022-00982-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
Activation of nociceptin opioid peptide receptors (NOP, a.k.a. opioid-like receptor-1, ORL-1) by the ligand nociceptin/orphanin FQ, leads to G protein-dependent regulation of Cav2.2 (N-type) voltage-gated calcium channels (VGCCs). This typically causes a reduction in calcium currents, triggering changes in presynaptic calcium levels and thus neurotransmission. Because of the widespread expression patterns of NOP and VGCCs across multiple brain regions, the dorsal horn of the spinal cord, and the dorsal root ganglia, this results in the alteration of numerous neurophysiological features. Here we review the regulation of N-type calcium channels by the NOP-nociceptin system in the context of neurological conditions such as anxiety, addiction, and pain.
Collapse
Affiliation(s)
- Emanuelle Sistherenn Caminski
- grid.412344.40000 0004 0444 6202Graduate Program in Health Sciences, Laboratory of Research in Toxicology (LAPETOX), Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS Brazil
| | - Flavia Tasmin Techera Antunes
- grid.22072.350000 0004 1936 7697Department of Clinical Neurosciences, University of Calgary, Calgary, AB Canada ,grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| | - Ivana Assis Souza
- grid.22072.350000 0004 1936 7697Department of Clinical Neurosciences, University of Calgary, Calgary, AB Canada ,grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| | - Eliane Dallegrave
- grid.412344.40000 0004 0444 6202Graduate Program in Health Sciences, Laboratory of Research in Toxicology (LAPETOX), Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS Brazil
| | - Gerald W. Zamponi
- grid.22072.350000 0004 1936 7697Department of Clinical Neurosciences, University of Calgary, Calgary, AB Canada ,grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| |
Collapse
|
8
|
Protein kinase Cγ in cerebellar Purkinje cells regulates Ca 2+-activated large-conductance K + channels and motor coordination. Proc Natl Acad Sci U S A 2022; 119:2113336119. [PMID: 35145028 PMCID: PMC8851492 DOI: 10.1073/pnas.2113336119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2021] [Indexed: 11/18/2022] Open
Abstract
The cerebellum, the site where protein kinase C (PKC) was discovered, contains the highest amount of PKCγ in the central nervous system. PKCγ in the cerebellum is exclusively confined to Purkinje cells (PCs), sole outputs from the cerebellar cortex. Systemic PKCγ-knockout mice show impaired motor coordination; however, the cause of motor defects remains unknown. Here we show that activation of PKCγ suppresses the Ca2+-activated large-conductance K+ (BK) channels located along the PC dendrites. A consequential increase in the membrane resistance attenuates electrical signal decay during propagation, resulting in an altered complex spike waveform. Our results suggest that synaptically activated PKCγ in PCs plays a critical role in motor coordination by negative modulation of BK currents. The cerebellum, the site where protein kinase C (PKC) was first discovered, contains the highest amount of PKC in the central nervous system, with PKCγ being the major isoform. Systemic PKCγ-knockout (KO) mice showed impaired motor coordination and deficient pruning of surplus climbing fibers (CFs) from developing cerebellar Purkinje cells (PCs). However, the physiological significance of PKCγ in the mature cerebellum and the cause of motor incoordination remain unknown. Using adeno-associated virus vectors targeting PCs, we showed that impaired motor coordination was restored by re-expression of PKCγ in mature PKCγ-KO mouse PCs in a kinase activity–dependent manner, while normal motor coordination in mature Prkcgfl/fl mice was impaired by the Cre-dependent removal of PKCγ from PCs. Notably, the rescue or removal of PKCγ from mature PKCγ-KO or Prkcgfl/fl mice, respectively, did not affect the CF innervation profile of PCs, suggesting the presence of a mechanism distinct from multiple CF innervation of PCs for the motor defects in PKCγ-deficient mice. We found marked potentiation of Ca2+-activated large-conductance K+ (BK) channel currents in PKCγ-deficient mice, as compared to wild-type mice, which decreased the membrane resistance, resulting in attenuation of the electrical signal during the propagation and significant alterations of the complex spike waveform. These changes in PKCγ-deficient mice were restored by the rescue of PKCγ or pharmacological suppression of BK channels. Our results suggest that PKCγ is a critical regulator that negatively modulates BK currents in PCs, which significantly influences PC output from the cerebellar cortex and, eventually, motor coordination.
Collapse
|
9
|
Voltage-dependent Ca V3.2 and Ca V2.2 channels in nociceptive pathways. Pflugers Arch 2022; 474:421-434. [PMID: 35043234 DOI: 10.1007/s00424-022-02666-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 10/19/2022]
Abstract
Noxious stimuli like cold, heat, pH change, tissue damage, and inflammation depolarize a membrane of peripheral endings of specialized nociceptive neurons which eventually results in the generation of an action potential. The electrical signal is carried along a long axon of nociceptive neurons from peripheral organs to soma located in dorsal root ganglions and further to the dorsal horn of the spinal cord where it is transmitted through a chemical synapse and is carried through the spinal thalamic tract into the brain. Two subtypes of voltage-activated calcium play a major role in signal transmission: a low voltage-activated CaV3.2 channel and a high voltage-activated CaV2.2 channel. The CaV3.2 channel contributes mainly to the signal conductance along nociceptive neurons while the principal role of the CaV2.2 channel is in the synaptic transmission at the dorsal horn. Both channels contribute to the signal initiation at peripheral nerve endings. This review summarizes current knowledge about the expression and distribution of these channels in a nociceptive pathway, the regulation of their expression and gating during pain pathology, and their suitability as targets for pharmacological therapy.
Collapse
|
10
|
Dong Y, Gao Y, Xu S, Wang Y, Yu Z, Li Y, Li B, Yuan T, Yang B, Zhang XC, Jiang D, Huang Z, Zhao Y. Closed-state inactivation and pore-blocker modulation mechanisms of human Ca V2.2. Cell Rep 2021; 37:109931. [PMID: 34731621 DOI: 10.1016/j.celrep.2021.109931] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/13/2021] [Accepted: 10/11/2021] [Indexed: 11/19/2022] Open
Abstract
N-type voltage-gated calcium (CaV) channels mediate Ca2+ influx at presynaptic terminals in response to action potentials and play vital roles in synaptogenesis, release of neurotransmitters, and nociceptive transmission. Here, we elucidate a cryo-electron microscopy (cryo-EM) structure of the human CaV2.2 complex in apo, ziconotide-bound, and two CaV2.2-specific pore blockers-bound states. The second voltage-sensing domain (VSD) is captured in a resting-state conformation, trapped by a phosphatidylinositol 4,5-bisphosphate (PIP2) molecule, which is distinct from the other three VSDs of CaV2.2, as well as activated VSDs observed in previous structures of CaV channels. This structure reveals the molecular basis for the unique inactivation process of CaV2.2 channels, in which the intracellular gate formed by S6 helices is closed and a W-helix from the domain II-III linker stabilizes closed-state inactivation. The structures of this inactivated, drug-bound complex lay a solid foundation for developing new state-dependent blockers for treatment of chronic pain.
Collapse
Affiliation(s)
- Yanli Dong
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiwei Gao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yuhang Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhuoya Yu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yue Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bin Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tian Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Bei Yang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuejun Cai Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Daohua Jiang
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China.
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China; IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| | - Yan Zhao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
11
|
McCarthy CI, Chou-Freed C, Rodríguez SS, Yaneff A, Davio C, Raingo J. Constitutive activity of dopamine receptor type 1 (D1R) increases CaV2.2 currents in PFC neurons. J Gen Physiol 2021; 152:151624. [PMID: 32259196 PMCID: PMC7201881 DOI: 10.1085/jgp.201912492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/14/2020] [Accepted: 03/12/2020] [Indexed: 01/19/2023] Open
Abstract
Alterations in dopamine receptor type 1 (D1R) density are associated with cognitive deficits of aging and schizophrenia. In the prefrontal cortex (PFC), D1R plays a critical role in the regulation of working memory, which is impaired in these cognitive deficit states, but the cellular events triggered by changes in D1R expression remain unknown. A previous report demonstrated that interaction between voltage-gated calcium channel type 2.2 (CaV2.2) and D1R stimulates CaV2.2 postsynaptic surface location in medial PFC pyramidal neurons. Here, we show that in addition to the occurrence of the physical receptor-channel interaction, constitutive D1R activity mediates up-regulation of functional CaV2.2 surface density. We performed patch-clamp experiments on transfected HEK293T cells and wild-type C57BL/6 mouse brain slices, as well as imaging experiments and cAMP measurements. We found that D1R coexpression led to ∼60% increase in CaV2.2 currents in HEK293T cells. This effect was occluded by preincubation with a D1/D5R inverse agonist, chlorpromazine, and by replacing D1R with a D1R mutant lacking constitutive activity. Moreover, D1R-induced increase in CaV2.2 currents required basally active Gs protein, as well as D1R-CaV2.2 interaction. In mice, intraperitoneal administration of chlorpromazine reduced native CaV currents’ sensitivity to ω-conotoxin-GVIA and their size by ∼49% in layer V/VI pyramidal neurons from medial PFC, indicating a selective effect on CaV2.2. Additionally, we found that reducing D1/D5R constitutive activity correlates with a decrease in the agonist-induced D1/D5R inhibitory effect on native CaV currents. Our results could be interpreted as a stimulatory effect of D1R constitutive activity on the number of CaV2.2 channels available for dopamine-mediated modulation. Our results contribute to the understanding of the physiological role of D1R constitutive activity and may explain the noncanonical postsynaptic distribution of functional CaV2.2 in PFC neurons.
Collapse
Affiliation(s)
- Clara Inés McCarthy
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology, Universidad Nacional de La Plata, Consejo de Investigaciones Científicas y Técnicas, Comisión de Investigaciones de la Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Cambria Chou-Freed
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology, Universidad Nacional de La Plata, Consejo de Investigaciones Científicas y Técnicas, Comisión de Investigaciones de la Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Silvia Susana Rodríguez
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology, Universidad Nacional de La Plata, Consejo de Investigaciones Científicas y Técnicas, Comisión de Investigaciones de la Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Agustín Yaneff
- Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos Davio
- Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jesica Raingo
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology, Universidad Nacional de La Plata, Consejo de Investigaciones Científicas y Técnicas, Comisión de Investigaciones de la Provincia de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
12
|
Garg J, Lakhani A, Dave V. Effects of the Involvement of Calcium Channels on Neuronal Hyperexcitability Related to Alzheimer’s Disease: A Computational Model. NEUROPHYSIOLOGY+ 2021. [DOI: 10.1007/s11062-021-09890-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
13
|
Gezalian MM, Mangiacotti L, Rajput P, Sparrow N, Schlick K, Lahiri S. Cerebrovascular and neurological perspectives on adrenoceptor and calcium channel modulating pharmacotherapies. J Cereb Blood Flow Metab 2021; 41:693-706. [PMID: 33210576 PMCID: PMC7983505 DOI: 10.1177/0271678x20972869] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 02/02/2023]
Abstract
Adrenoceptor and calcium channel modulating medications are widely used in clinical practice for acute neurological and systemic conditions. It is generally assumed that the cerebrovascular effects of these drugs mirror that of their systemic effects - and this is reflected in how these medications are currently used in clinical practice. However, recent research suggests that there are distinct cerebrovascular-specific effects of these medications that are related to the unique characteristics of the cerebrovascular anatomy including the regional heterogeneity in density and distribution of adrenoceptor subtypes and calcium channels along the cerebrovasculature. In this review, we critically evaluate existing basic science and clinical research to discuss known and putative interactions between adrenoceptor and calcium channel modulating pharmacotherapies, the neurovascular unit, and cerebrovascular anatomy. In doing so, we provide a rationale for selecting vasoactive medications based on lesion location and lay a foundation for future investigations that will define neuroprotective paradigms of adrenoceptor and calcium channel modulating therapies to improve neurological outcomes in acute neurological and systemic disorders.
Collapse
Affiliation(s)
- Michael M Gezalian
- Departments of Neurology and Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Luigi Mangiacotti
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Padmesh Rajput
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nicklaus Sparrow
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Konrad Schlick
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shouri Lahiri
- Departments of Neurology, Neurosurgery, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
14
|
Hasan MM, Ragnarsson L, Cardoso FC, Lewis RJ. Transfection methods for high-throughput cellular assays of voltage-gated calcium and sodium channels involved in pain. PLoS One 2021; 16:e0243645. [PMID: 33667217 PMCID: PMC7935312 DOI: 10.1371/journal.pone.0243645] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 11/25/2020] [Indexed: 11/24/2022] Open
Abstract
Chemical transfection is broadly used to transiently transfect mammalian cells, although often associated with cellular stress and membrane instability, which imposes challenges for most cellular assays, including high-throughput (HT) assays. In the current study, we compared the effectiveness of calcium phosphate, FuGENE and Lipofectamine 3000 to transiently express two key voltage-gated ion channels critical in pain pathways, CaV2.2 and NaV1.7. The expression and function of these channels were validated using two HT platforms, the Fluorescence Imaging Plate Reader FLIPRTetra and the automated patch clamp QPatch 16X. We found that all transfection methods tested demonstrated similar effectiveness when applied to FLIPRTetra assays. Lipofectamine 3000-mediated transfection produced the largest peak currents for automated patch clamp QPatch assays. However, the FuGENE-mediated transfection was the most effective for QPatch assays as indicated by the superior number of cells displaying GΩ seal formation in whole-cell patch clamp configuration, medium to large peak currents, and higher rates of accomplished assays for both CaV2.2 and NaV1.7 channels. Our findings can facilitate the development of HT automated patch clamp assays for the discovery and characterization of novel analgesics and modulators of pain pathways, as well as assisting studies examining the pharmacology of mutated channels.
Collapse
Affiliation(s)
- Md. Mahadhi Hasan
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Lotten Ragnarsson
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Fernanda C. Cardoso
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
- * E-mail: (FCC); (RJL)
| | - Richard J. Lewis
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
- * E-mail: (FCC); (RJL)
| |
Collapse
|
15
|
Gandini MA, Zamponi GW. Voltage‐gated calcium channel nanodomains: molecular composition and function. FEBS J 2021; 289:614-633. [DOI: 10.1111/febs.15759] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Maria A. Gandini
- Department of Physiology and Pharmacology Alberta Children’s Hospital Research Institute Hotchkiss Brain Institute Cumming School of Medicine University of 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 AB Canada
| |
Collapse
|
16
|
Hasan MM, Starobova H, Mueller A, Vetter I, Lewis RJ. Subcutaneous ω-Conotoxins Alleviate Mechanical Pain in Rodent Models of Acute Peripheral Neuropathy. Mar Drugs 2021; 19:106. [PMID: 33670311 PMCID: PMC7917901 DOI: 10.3390/md19020106] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 11/16/2022] Open
Abstract
The peripheral effects of ω-conotoxins, selective blockers of N-type voltage-gated calcium channels (CaV2.2), have not been characterised across different clinically relevant pain models. This study examines the effects of locally administered ω-conotoxin MVIIA, GVIA, and CVIF on mechanical and thermal paw withdrawal threshold (PWT) in postsurgical pain (PSP), cisplatin-induced neuropathy (CisIPN), and oxaliplatin-induced neuropathy (OIPN) rodent models. Intraplantar injection of 300, 100 and 30 nM MVIIA significantly (p < 0.0001, p < 0.0001, and p < 0.05, respectively) alleviated mechanical allodynia of mice in PSP model compared to vehicle control group. Similarly, intraplantar injection of 300, 100, and 30 nM MVIIA (p < 0.0001, p < 0.01, and p < 0.05, respectively), and 300 nM and 100 nM GVIA (p < 0.0001 and p < 0.05, respectively) significantly increased mechanical thresholds of mice in OIPN model. The ED50 of GVIA and MVIIA in OIPN was found to be 1.8 pmol/paw and 0.8 pmol/paw, respectively. However, none of the ω-conotoxins were effective in a mouse model of CisIPN. The intraplantar administration of 300 nM GVIA, MVIIA, and CVIF did not cause any locomotor side effects. The intraplantar administration of MVIIA can alleviate incision-induced mechanical allodynia, and GVIA and MVIIA effectively reduce OIPN associated mechanical pain, without locomotor side effects, in rodent models. In contrast, CVIF was inactive in these pain models, suggesting it is unable to block a subset of N-type voltage-gated calcium channels associated with nociceptors in the skin.
Collapse
Affiliation(s)
- Md. Mahadhi Hasan
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (M.M.H.); (H.S.); (A.M.); (I.V.)
| | - Hana Starobova
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (M.M.H.); (H.S.); (A.M.); (I.V.)
| | - Alexander Mueller
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (M.M.H.); (H.S.); (A.M.); (I.V.)
| | - Irina Vetter
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (M.M.H.); (H.S.); (A.M.); (I.V.)
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Richard J. Lewis
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (M.M.H.); (H.S.); (A.M.); (I.V.)
| |
Collapse
|
17
|
Lacinova L, Mallmann RT, Jurkovičová-Tarabová B, Klugbauer N. Modulation of voltage-gated Ca V2.2 Ca 2+ channels by newly identified interaction partners. Channels (Austin) 2020; 14:380-392. [PMID: 33006503 PMCID: PMC7567506 DOI: 10.1080/19336950.2020.1831328] [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] [Indexed: 11/30/2022] Open
Abstract
Voltage-gated Ca2+ channels are typically integrated in a complex network of protein-protein-interactions, also referred to as Ca2+ channel nanodomains. Amongst the neuronal CaV2 channel family, CaV2.2 is of particular importance due to its general role for signal transmission from the periphery to the central nervous system, but also due to its significance for pain perception. Thus, CaV2.2 is an ideal target candidate to search for pharmacological inhibitors but also for novel modulatory interactors. In this review we summarize the last years findings of our intense screenings and characterization of the six CaV2.2 interaction partners, tetraspanin-13 (TSPAN-13), reticulon 1 (RTN1), member 1 of solute carrier family 38 (SLC38), prostaglandin D2 synthase (PTGDS), transmembrane protein 223 (TMEM223), and transmembrane BAX inhibitor motif 3 (Grina/TMBIM3) containing protein. Each protein shows a unique way of channel modulation as shown by extensive electrophysiological studies. Amongst the newly identified interactors, Grina/TMBIM3 is most striking due to its modulatory effect which is rather comparable to G-protein regulation.
Collapse
Affiliation(s)
- Lubica Lacinova
- Center of Bioscience, - Institute for Molecular Physiology and Genetics , Bratislava, Slovakia.,Faculty of Natural Sciences, University of Ss. Cyril and Methodius , Trnava, Slovakia
| | - Robert Theodor Mallmann
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Fakultät für Medizin, Albert-Ludwigs-Universität Freiburg , Freiburg, Germany
| | | | - Norbert Klugbauer
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Fakultät für Medizin, Albert-Ludwigs-Universität Freiburg , Freiburg, Germany.,Center for Basics in NeuroModulation (Neuromodul Basics), Albert-Ludwigs-Universität Freiburg , Freiburg, Germany
| |
Collapse
|
18
|
Presynaptic L-Type Ca 2+ Channels Increase Glutamate Release Probability and Excitatory Strength in the Hippocampus during Chronic Neuroinflammation. J Neurosci 2020; 40:6825-6841. [PMID: 32747440 DOI: 10.1523/jneurosci.2981-19.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 06/18/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation is involved in the pathogenesis of several neurologic disorders, including epilepsy. Both changes in the input/output functions of synaptic circuits and cell Ca2+ dysregulation participate in neuroinflammation, but their impact on neuron function in epilepsy is still poorly understood. Lipopolysaccharide (LPS), a toxic byproduct of bacterial lysis, has been extensively used to stimulate inflammatory responses both in vivo and in vitro LPS stimulates Toll-like receptor 4, an important mediator of the brain innate immune response that contributes to neuroinflammation processes. Although we report that Toll-like receptor 4 is expressed in both excitatory and inhibitory mouse hippocampal neurons (both sexes), its chronic stimulation by LPS induces a selective increase in the excitatory synaptic strength, characterized by enhanced synchronous and asynchronous glutamate release mechanisms. This effect is accompanied by a change in short-term plasticity with decreased facilitation, decreased post-tetanic potentiation, and increased depression. Quantal analysis demonstrated that the effects of LPS on excitatory transmission are attributable to an increase in the probability of release associated with an overall increased expression of L-type voltage-gated Ca2+ channels that, at presynaptic terminals, abnormally contributes to evoked glutamate release. Overall, these changes contribute to the excitatory/inhibitory imbalance that scales up neuronal network activity under inflammatory conditions. These results provide new molecular clues for treating hyperexcitability of hippocampal circuits associated with neuroinflammation in epilepsy and other neurologic disorders.SIGNIFICANCE STATEMENT Neuroinflammation is thought to have a pathogenetic role in epilepsy, a disorder characterized by an imbalance between excitation/inhibition. Fine adjustment of network excitability and regulation of synaptic strength are both implicated in the homeostatic maintenance of physiological levels of neuronal activity. Here, we focused on the effects of chronic neuroinflammation induced by lipopolysaccharides on hippocampal glutamatergic and GABAergic synaptic transmission. Our results show that, on chronic stimulation with lipopolysaccharides, glutamatergic, but not GABAergic, neurons exhibit an enhanced synaptic strength and changes in short-term plasticity because of an increased glutamate release that results from an anomalous contribution of L-type Ca2+ channels to neurotransmitter release.
Collapse
|
19
|
Bjørn-Yoshimoto WE, Ramiro IBL, Yandell M, McIntosh JM, Olivera BM, Ellgaard L, Safavi-Hemami H. Curses or Cures: A Review of the Numerous Benefits Versus the Biosecurity Concerns of Conotoxin Research. Biomedicines 2020; 8:E235. [PMID: 32708023 PMCID: PMC7460000 DOI: 10.3390/biomedicines8080235] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 01/18/2023] Open
Abstract
Conotoxins form a diverse group of peptide toxins found in the venom of predatory marine cone snails. Decades of conotoxin research have provided numerous measurable scientific and societal benefits. These include their use as a drug, diagnostic agent, drug leads, and research tools in neuroscience, pharmacology, biochemistry, structural biology, and molecular evolution. Human envenomations by cone snails are rare but can be fatal. Death by envenomation is likely caused by a small set of toxins that induce muscle paralysis of the diaphragm, resulting in respiratory arrest. The potency of these toxins led to concerns regarding the potential development and use of conotoxins as biological weapons. To address this, various regulatory measures have been introduced that limit the use and access of conotoxins within the research community. Some of these regulations apply to all of the ≈200,000 conotoxins predicted to exist in nature of which less than 0.05% are estimated to have any significant toxicity in humans. In this review we provide an overview of the many benefits of conotoxin research, and contrast these to the perceived biosecurity concerns of conotoxins and research thereof.
Collapse
Affiliation(s)
- Walden E. Bjørn-Yoshimoto
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; (W.E.B.-Y.); (I.B.L.R.)
| | - Iris Bea L. Ramiro
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; (W.E.B.-Y.); (I.B.L.R.)
| | - Mark Yandell
- Eccles Institute of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA;
- Utah Center for Genetic Discovery, University of Utah, Salt Lake City, UT 84112, USA
| | - J. Michael McIntosh
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA; (J.M.M.); (B.M.O.)
- George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT 84148, USA
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA
| | - Baldomero M. Olivera
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA; (J.M.M.); (B.M.O.)
| | - Lars Ellgaard
- Department of Biology, Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, 2200 Copenhagen N, Denmark;
| | - Helena Safavi-Hemami
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; (W.E.B.-Y.); (I.B.L.R.)
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA; (J.M.M.); (B.M.O.)
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
20
|
Targeting the CaVα-CaVβ interaction yields an antagonist of the N-type CaV2.2 channel with broad antinociceptive efficacy. Pain 2020; 160:1644-1661. [PMID: 30933958 DOI: 10.1097/j.pain.0000000000001524] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Inhibition of voltage-gated calcium (CaV) channels is a potential therapy for many neurological diseases including chronic pain. Neuronal CaV1/CaV2 channels are composed of α, β, γ and α2δ subunits. The β subunits of CaV channels are cytoplasmic proteins that increase the surface expression of the pore-forming α subunit of CaV. We targeted the high-affinity protein-protein interface of CaVβ's pocket within the CaVα subunit. Structure-based virtual screening of 50,000 small molecule library docked to the β subunit led to the identification of 2-(3,5-dimethylisoxazol-4-yl)-N-((4-((3-phenylpropyl)amino)quinazolin-2-yl)methyl)acetamide (IPPQ). This small molecule bound to CaVβ and inhibited its coupling with N-type voltage-gated calcium (CaV2.2) channels, leading to a reduction in CaV2.2 currents in rat dorsal root ganglion sensory neurons, decreased presynaptic localization of CaV2.2 in vivo, decreased frequency of spontaneous excitatory postsynaptic potentials and miniature excitatory postsynaptic potentials, and inhibited release of the nociceptive neurotransmitter calcitonin gene-related peptide from spinal cord. IPPQ did not target opioid receptors nor did it engage inhibitory G protein-coupled receptor signaling. IPPQ was antinociceptive in naive animals and reversed allodynia and hyperalgesia in models of acute (postsurgical) and neuropathic (spinal nerve ligation, chemotherapy- and gp120-induced peripheral neuropathy, and genome-edited neuropathy) pain. IPPQ did not cause akinesia or motor impairment, a common adverse effect of CaV2.2 targeting drugs, when injected into the brain. IPPQ, a quinazoline analog, represents a novel class of CaV2.2-targeting compounds that may serve as probes to interrogate CaVα-CaVβ function and ultimately be developed as a nonopioid therapeutic for chronic pain.
Collapse
|
21
|
Zhou Y, Cai S, Gomez K, Wijeratne EMK, Ji Y, Bellampalli SS, Luo S, Moutal A, Gunatilaka AAL, Khanna R. 1-O-Acetylgeopyxin A, a derivative of a fungal metabolite, blocks tetrodotoxin-sensitive voltage-gated sodium, calcium channels and neuronal excitability which correlates with inhibition of neuropathic pain. Mol Brain 2020; 13:73. [PMID: 32393368 PMCID: PMC7216607 DOI: 10.1186/s13041-020-00616-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/04/2020] [Indexed: 01/03/2023] Open
Abstract
Chronic pain can be the result of an underlying disease or condition, medical treatment, inflammation, or injury. The number of persons experiencing this type of pain is substantial, affecting upwards of 50 million adults in the United States. Pharmacotherapy of most of the severe chronic pain patients includes drugs such as gabapentinoids, re-uptake blockers and opioids. Unfortunately, gabapentinoids are not effective in up to two-thirds of this population and although opioids can be initially effective, their long-term use is associated with multiple side effects. Therefore, there is a great need to develop novel non-opioid alternative therapies to relieve chronic pain. For this purpose, we screened a small library of natural products and their derivatives in the search for pharmacological inhibitors of voltage-gated calcium and sodium channels, which are outstanding molecular targets due to their important roles in nociceptive pathways. We discovered that the acetylated derivative of the ent-kaurane diterpenoid, geopyxin A, 1-O-acetylgeopyxin A, blocks voltage-gated calcium and tetrodotoxin-sensitive voltage-gated sodium channels but not tetrodotoxin-resistant sodium channels in dorsal root ganglion (DRG) neurons. Consistent with inhibition of voltage-gated sodium and calcium channels, 1-O-acetylgeopyxin A reduced reduce action potential firing frequency and increased firing threshold (rheobase) in DRG neurons. Finally, we identified the potential of 1-O-acetylgeopyxin A to reverse mechanical allodynia in a preclinical rat model of HIV-induced sensory neuropathy. Dual targeting of both sodium and calcium channels may permit block of nociceptor excitability and of release of pro-nociceptive transmitters. Future studies will harness the core structure of geopyxins for the generation of antinociceptive drugs.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Clinical Laboratory, the First Hospital of Jilin University, Changchun, 130021, China
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Song Cai
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Kimberly Gomez
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - E M Kithsiri Wijeratne
- Southwest Center for Natural Products Research, School of Natural Resources & the Environment, College of Agriculture & Life Sciences, The University of Arizona, Tucson, AZ, 85724, USA
| | - Yingshi Ji
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Shreya S Bellampalli
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Shizhen Luo
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - A A Leslie Gunatilaka
- Southwest Center for Natural Products Research, School of Natural Resources & the Environment, College of Agriculture & Life Sciences, The University of Arizona, Tucson, AZ, 85724, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA.
- Neuroscience Graduate Interdisciplinary Program, College of Medicine, Tucson, AZ, 85724, USA.
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, 85724, USA.
| |
Collapse
|
22
|
Calcium Signaling in Neurons and Glial Cells: Role of Cav1 channels. Neuroscience 2019; 421:95-111. [DOI: 10.1016/j.neuroscience.2019.09.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 11/18/2022]
|
23
|
Pitake S, Middleton LJ, Abdus-Saboor I, Mishra SK. Inflammation Induced Sensory Nerve Growth and Pain Hypersensitivity Requires the N-Type Calcium Channel Cav2.2. Front Neurosci 2019; 13:1009. [PMID: 31607850 PMCID: PMC6761232 DOI: 10.3389/fnins.2019.01009] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/05/2019] [Indexed: 12/26/2022] Open
Abstract
Voltage-gated calcium channels (VGCCs) are important mediators of pain hypersensitivity during inflammatory states, but their role in sensory nerve growth remains underexplored. Here, we assess the role of the N-type calcium channel Cav2.2 in the complete Freund’s adjuvant (CFA) model of inflammatory pain. We demonstrate with in situ hybridization and immunoblotting, an increase in Cav2.2 expression after hind paw CFA injection in sensory neurons that respond to thermal stimuli, but not in two different mechanosensitive neuronal populations. Further, Cav2.2 upregulation post-CFA correlates with thermal but not mechanical hyperalgesia in behaving mice, and this hypersensitivity is blocked with a specific Cav2.2 inhibitor. Voltage clamp recordings reveal a significant increase in Cav2.2 currents post-CFA, while current clamp analyses demonstrate a significant increase in action potential frequency. Moreover, CFA-induced sensory nerve growth, which involves the extracellular signal-related kinase (ERK1/2) signaling pathway and likely contributes to inflammation-induced hyperalgesia, was blocked with the Cav2.2 inhibitor. Together, this work uncovers a role for Cav2.2 during inflammation, demonstrating that VGCC activity can promote thermal hyperalgesia through both changes in firing rates of sensory neurons as well as promotion of new neurite outgrowth.
Collapse
Affiliation(s)
- Saumitra Pitake
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| | - Leah J Middleton
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| | - Ishmail Abdus-Saboor
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| | - Santosh K Mishra
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,The W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, United States.,Program in Genetics, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
24
|
Yu H, Shin SM, Xiang H, Chao D, Cai Y, Xu H, Khanna R, Pan B, Hogan QH. AAV-encoded Ca V2.2 peptide aptamer CBD3A6K for primary sensory neuron-targeted treatment of established neuropathic pain. Gene Ther 2019; 26:308-323. [PMID: 31118475 DOI: 10.1038/s41434-019-0082-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/25/2019] [Accepted: 03/20/2019] [Indexed: 12/17/2022]
Abstract
Transmission of pain signals from primary sensory neurons to secondary neurons of the central nervous system is critically dependent on presynaptic voltage-gated calcium channels. Calcium channel-binding domain 3 (CBD3), derived from the collapsin response mediator protein 2 (CRMP2), is a peptide aptamer that is effective in blocking N-type voltage-gated calcium channel (CaV2.2) activity. We previously reported that recombinant adeno-associated virus (AAV)-mediated restricted expression of CBD3 affixed to enhanced green fluorescent protein (EGFP) in primary sensory neurons prevents the development of cutaneous mechanical hypersensitivity in a rat neuropathic pain model. In this study, we tested whether this strategy is effective in treating established pain. We constructed AAV6-EGFP-CBD3A6K (AAV6-CBD3A6K) expressing a fluorescent CBD3A6K (replacing A to K at position 6 of CBD3 peptide), which is an optimized variant of the parental CBD3 peptide that is a more potent blocker of CaV2.2. Delivery of AAV6-CBD3A6K into lumbar (L) 4 and 5 dorsal root ganglia (DRG) of rats 2 weeks following tibial nerve injury (TNI) induced transgene expression in neurons of these DRG and their axonal projections, accompanied by attenuation of pain behavior. We additionally observed that the increased CaV2.2α1b immunoreactivity in the ipsilateral spinal cord dorsal horn and DRG following TNI was significantly normalized by AAV6-CBD3A6K treatment. Finally, the increased neuronal activity in the ipsilateral dorsal horn that developed after TNI was reduced by AAV6-CBD3A6K treatment. Collectively, these results indicate that DRG-restricted AAV6 delivery of CBD3A6K is an effective analgesic molecular strategy for the treatment of established neuropathic pain.
Collapse
Affiliation(s)
- Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA. .,Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA.
| | - Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.,Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA
| | - Hongfei Xiang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.,Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, 266000, Qingdao, PR China
| | - Dongman Chao
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Yongsong Cai
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.,Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, PR China
| | - Hao Xu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.,Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, 266000, Qingdao, PR China
| | - Rajesh Khanna
- Departments of Pharmacology, Neuroscience and Anesthesiology, College of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.,Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA
| |
Collapse
|
25
|
Presynaptic Calcium Channels. Int J Mol Sci 2019; 20:ijms20092217. [PMID: 31064106 PMCID: PMC6539076 DOI: 10.3390/ijms20092217] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/22/2019] [Accepted: 04/26/2019] [Indexed: 12/27/2022] Open
Abstract
Presynaptic Ca2+ entry occurs through voltage-gated Ca2+ (CaV) channels which are activated by membrane depolarization. Depolarization accompanies neuronal firing and elevation of Ca2+ triggers neurotransmitter release from synaptic vesicles. For synchronization of efficient neurotransmitter release, synaptic vesicles are targeted by presynaptic Ca2+ channels forming a large signaling complex in the active zone. The presynaptic CaV2 channel gene family (comprising CaV2.1, CaV2.2, and CaV2.3 isoforms) encode the pore-forming α1 subunit. The cytoplasmic regions are responsible for channel modulation by interacting with regulatory proteins. This article overviews modulation of the activity of CaV2.1 and CaV2.2 channels in the control of synaptic strength and presynaptic plasticity.
Collapse
|
26
|
Ablation of α 2δ-1 inhibits cell-surface trafficking of endogenous N-type calcium channels in the pain pathway in vivo. Proc Natl Acad Sci U S A 2018; 115:E12043-E12052. [PMID: 30487217 PMCID: PMC6305000 DOI: 10.1073/pnas.1811212115] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Neuronal N-type (CaV2.2) voltage-gated calcium channels are important at the first synapse in the pain pathway. In this study, we have characterized a knockin mouse containing CaV2.2 with an extracellular HA tag to determine the localization of CaV2.2 in primary afferent pain pathways. These endogenous channels have been visualized at the plasma membrane and rigorously quantified in vivo. We examined the effect of ablation of the calcium channel auxiliary subunit α2δ-1 (the target of gabapentinoids) on CaV2.2 distribution. We found preferential cell-surface localization of CaV2.2 in DRG nociceptor neuron cell bodies was lost, accompanied by a dramatic reduction at dorsal horn terminals, but no effect on distribution of other spinal cord synaptic markers. The auxiliary α2δ calcium channel subunits play key roles in voltage-gated calcium channel function. Independent of this, α2δ-1 has also been suggested to be important for synaptogenesis. Using an epitope-tagged knockin mouse strategy, we examined the effect of α2δ-1 on CaV2.2 localization in the pain pathway in vivo, where CaV2.2 is important for nociceptive transmission and α2δ-1 plays a critical role in neuropathic pain. We find CaV2.2 is preferentially expressed on the plasma membrane of calcitonin gene-related peptide-positive small nociceptors. This is paralleled by strong presynaptic expression of CaV2.2 in the superficial spinal cord dorsal horn. EM-immunogold localization shows CaV2.2 predominantly in active zones of glomerular primary afferent terminals. Genetic ablation of α2δ-1 abolishes CaV2.2 cell-surface expression in dorsal root ganglion neurons and dramatically reduces dorsal horn expression. There was no effect of α2δ-1 knockout on other dorsal horn pre- and postsynaptic markers, indicating the primary afferent pathways are not otherwise affected by α2δ-1 ablation.
Collapse
|
27
|
Voltage-Dependent Calcium Channels, Calcium Binding Proteins, and Their Interaction in the Pathological Process of Epilepsy. Int J Mol Sci 2018; 19:ijms19092735. [PMID: 30213136 PMCID: PMC6164075 DOI: 10.3390/ijms19092735] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 01/08/2023] Open
Abstract
As an important second messenger, the calcium ion (Ca2+) plays a vital role in normal brain function and in the pathophysiological process of different neurodegenerative diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), and epilepsy. Ca2+ takes part in the regulation of neuronal excitability, and the imbalance of intracellular Ca2+ is a trigger factor for the occurrence of epilepsy. Several anti-epileptic drugs target voltage-dependent calcium channels (VDCCs). Intracellular Ca2+ levels are mainly controlled by VDCCs located in the plasma membrane, the calcium-binding proteins (CBPs) inside the cytoplasm, calcium channels located on the intracellular calcium store (particular the endoplasmic reticulum/sarcoplasmic reticulum), and the Ca2+-pumps located in the plasma membrane and intracellular calcium store. So far, while many studies have established the relationship between calcium control factors and epilepsy, the mechanism of various Ca2+ regulatory factors in epileptogenesis is still unknown. In this paper, we reviewed the function, distribution, and alteration of VDCCs and CBPs in the central nervous system in the pathological process of epilepsy. The interaction of VDCCs with CBPs in the pathological process of epilepsy was also summarized. We hope this review can provide some clues for better understanding the mechanism of epileptogenesis, and for the development of new anti-epileptic drugs targeting on VDCCs and CBPs.
Collapse
|
28
|
Zhou WZ, Zhao TY, Wang ZY, Lu GY, Zhang SZ, Zhang C, Wu N, Li J. Synergistic antinociception between ZC88, an N-type voltage-dependent calcium channel blocker, and ibuprofen in mouse models of visceral and somatic inflammatory pain. Eur J Pain 2018; 23:46-56. [PMID: 29978517 DOI: 10.1002/ejp.1281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2018] [Indexed: 11/06/2022]
Abstract
BACKGROUND A combination of analgesic agents with different mechanisms can induce additive or synergistic analgesia. The N-type voltage-dependent calcium channel (N-VDCC) is a novel therapeutic target for pain control. In addition to providing effective pain relief when used alone, N-VDCC blockers produce synergistic analgesia when used in combination with opiates. However, the interaction between N-VDCC blockers and nonsteroidal anti-inflammatory drugs (NSAIDs) remains unclear. METHODS Using isobolographic analysis and composite additive curve analysis, the antinociceptive interaction between ZC88, a selective N-VDCC blocker and ibuprofen, a classical NSAID, was investigated in two mouse models of visceral and somatic inflammatory pain. RESULTS In the acetic acid writhing test, both ZC88 (10.5-42 mg/kg, intraperitoneally) and ibuprofen (50-200 mg/kg, orally) produced dose-dependent antinociception, with ED50 values of 27.2 and 100.5 mg/kg, respectively. ZC88 in combination with ibuprofen (ZC88 + ibuprofen) also induced significant antinociception, and isobolographic analysis revealed a synergistic interaction at 50% effect level. The experimental ED50 (ED50 mix ) of this combination (34.5 mg/kg) was significantly lower than the theoretical ED50 (ED50 add ; 63.8 mg/kg). Additionally, composite additive curve analysis displayed synergistic interaction at other effect levels. In the formalin test, ZC88 or ibuprofen alone significantly reduced late-phase rather than early-phase pain, with ED50 values of 31.3 and 123.9 mg/kg, respectively. Similarly, both isobolographic analysis and composite additive curve analysis revealed synergistic antinociception of ZC88 + ibuprofen (40.6 mg/kg of ED50 mix vs. 77.6 mg/kg of ED50 add ). CONCLUSION ZC88 in combination with ibuprofen produces synergistic antinociception in mouse models of somatic and visceral inflammatory pain. SIGNIFICANCE Because ZC88 + ibuprofen achieves the same antinociceptive effect at lower doses, the use of this combination could result in fewer dose-related untoward effects. The potentiation of ZC88 on ibuprofen-induced antinociception indicates that N-VDCC blocker has potential benefit to treat severe inflammatory pain.
Collapse
Affiliation(s)
- W-Z Zhou
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | - T-Y Zhao
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | - Z-Y Wang
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | - G-Y Lu
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | - S-Z Zhang
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | - C Zhang
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | - N Wu
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| | - J Li
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, China
| |
Collapse
|
29
|
Abstract
Neuropathic pain represents a significant and mounting burden on patients and society at large. Management of neuropathic pain, however, is both intricate and challenging, exacerbated by the limited quantity and quality of clinically available treatments. On this stage, dysfunctional voltage-gated ion channels, especially the presynaptic N-type voltage-gated calcium channel (VGCC) (Cav2.2) and the tetrodotoxin-sensitive voltage-gated sodium channel (VGSC) (Nav1.7), underlie the pathophysiology of neuropathic pain and serve as high profile therapeutic targets. Indirect regulation of these channels holds promise for the treatment of neuropathic pain. In this review, we focus on collapsin response mediator protein 2 (CRMP2), a protein with emergent roles in voltage-gated ion channel trafficking and discuss the therapeutic potential of targetting this protein.
Collapse
|
30
|
Presynaptic calcium channels. Neurosci Res 2018; 127:33-44. [DOI: 10.1016/j.neures.2017.09.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 07/13/2017] [Accepted: 08/23/2017] [Indexed: 12/30/2022]
|
31
|
Garcia-Rill E, Mahaffey S, Hyde JR, Urbano FJ. Bottom-up gamma maintenance in various disorders. Neurobiol Dis 2018; 128:31-39. [PMID: 29353013 DOI: 10.1016/j.nbd.2018.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/02/2018] [Accepted: 01/10/2018] [Indexed: 11/30/2022] Open
Abstract
Maintained gamma band activity is a key element of higher brain function, participating in perception, executive function, and memory. The pedunculopontine nucleus (PPN), as part of the reticular activating system (RAS), is a major source of the "bottom-up" flow of gamma activity to higher regions. However, interruption of gamma band activity is associated with a number of neurological and psychiatric disorders. This review will focus on the role of the PPN in activating higher regions to induce arousal and descending pathways to modulate posture and locomotion. As such, PPN deep brain stimulation (DBS) can not only help regulate arousal and stepping, but continuous application may help maintain necessary levels of gamma band activity for a host of other brain processes. We will explore the potential future applications of PPN DBS for a number of disorders that are characterized by disturbances in gamma band maintenance.
Collapse
Affiliation(s)
- E Garcia-Rill
- Center for Translational Neuroscience, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - S Mahaffey
- Center for Translational Neuroscience, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - F J Urbano
- IFIBYNE (CONICET-UBA), DFBMC, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
32
|
Zhang D, Tu H, Cao L, Zheng H, Muelleman RL, Wadman MC, Li YL. Reduced N-Type Ca 2+ Channels in Atrioventricular Ganglion Neurons Are Involved in Ventricular Arrhythmogenesis. J Am Heart Assoc 2018; 7:JAHA.117.007457. [PMID: 29335317 PMCID: PMC5850164 DOI: 10.1161/jaha.117.007457] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Attenuated cardiac vagal activity is associated with ventricular arrhythmogenesis and related mortality in patients with chronic heart failure. Our recent study has shown that expression of N‐type Ca2+ channel α‐subunits (Cav2.2‐α) and N‐type Ca2+ currents are reduced in intracardiac ganglion neurons from rats with chronic heart failure. Rat intracardiac ganglia are divided into the atrioventricular ganglion (AVG) and sinoatrial ganglion. Ventricular myocardium receives projection of neuronal terminals only from the AVG. In this study we tested whether a decrease in N‐type Ca2+ channels in AVG neurons contributes to ventricular arrhythmogenesis. Methods and Results Lentiviral Cav2.2‐α shRNA (2 μL, 2×107 pfu/mL) or scrambled shRNA was in vivo transfected into rat AVG neurons. Nontransfected sham rats served as controls. Using real‐time single‐cell polymerase chain reaction and reverse‐phase protein array, we found that in vivo transfection of Cav2.2‐α shRNA decreased expression of Cav2.2‐α mRNA and protein in rat AVG neurons. Whole‐cell patch‐clamp data showed that Cav2.2‐α shRNA reduced N‐type Ca2+ currents and cell excitability in AVG neurons. The data from telemetry electrocardiographic recording demonstrated that 83% (5 out of 6) of conscious rats with Cav2.2‐α shRNA transfection had premature ventricular contractions (P<0.05 versus 0% of nontransfected sham rats or scrambled shRNA‐transfected rats). Additionally, an index of susceptibility to ventricular arrhythmias, inducibility of ventricular arrhythmias evoked by programmed electrical stimulation, was higher in rats with Cav2.2‐α shRNA transfection compared with nontransfected sham rats and scrambled shRNA‐transfected rats. Conclusions A decrease in N‐type Ca2+ channels in AVG neurons attenuates vagal control of ventricular myocardium, thereby initiating ventricular arrhythmias.
Collapse
Affiliation(s)
- Dongze Zhang
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Huiyin Tu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Liang Cao
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE.,Department of Cardiac surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Hong Zheng
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE
| | - Robert L Muelleman
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Michael C Wadman
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE .,Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
33
|
Suarez LM, Solis O, Aguado C, Lujan R, Moratalla R. L-DOPA Oppositely Regulates Synaptic Strength and Spine Morphology in D1 and D2 Striatal Projection Neurons in Dyskinesia. Cereb Cortex 2018; 26:4253-4264. [PMID: 27613437 PMCID: PMC5066835 DOI: 10.1093/cercor/bhw263] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/01/2016] [Indexed: 12/21/2022] Open
Abstract
Dopamine depletion in Parkinson's disease (PD) produces dendritic spine loss in striatal medium spiny neurons (MSNs) and increases their excitability. However, the synaptic changes that occur in MSNs in PD, in particular those induced by chronic L-3,4-dihydroxyphenylalanine (L-DOPA) treatment, are still poorly understood. We exposed BAC-transgenic D1-tomato and D2-eGFP mice to PD and dyskinesia model paradigms, enabling cell type-specific assessment of changes in synaptic physiology and morphology. The distinct fluorescence markers allowed us to identify D1 and D2 MSNs for analysis using intracellular sharp electrode recordings, electron microscopy, and 3D reconstructions with single-cell Lucifer Yellow injections. Dopamine depletion induced spine pruning in both types of MSNs, affecting mushroom and thin spines equally. Dopamine depletion also increased firing rate in both D1- and D2-MSNs, but reduced evoked-EPSP amplitude selectively in D2-MSNs. L-DOPA treatment that produced dyskinesia differentially affected synaptic properties in D1- and D2-MSNs. In D1-MSNs, spine density remained reduced but the remaining spines were enlarged, with bigger heads and larger postsynaptic densities. These morphological changes were accompanied by facilitation of action potential firing triggered by synaptic inputs. In contrast, although L-DOPA restored the number of spines in D2-MSNs, it resulted in shortened postsynaptic densities. These changes in D2-MSNs correlated with a decrease in synaptic transmission. Our findings indicate that L-DOPA-induced dyskinesia is associated with abnormal spine morphology, modified synaptic transmission, and altered EPSP-spike coupling, with distinct effects in D1- and D2-MSNs.
Collapse
Affiliation(s)
- Luz M Suarez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, 28002 Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| | - Oscar Solis
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, 28002 Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| | - Carolina Aguado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Dept. Ciencias Medicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Campus Biosanitario, Albacete, Spain, Spain
| | - Rafael Lujan
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Dept. Ciencias Medicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Campus Biosanitario, Albacete, Spain, Spain
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, 28002 Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| |
Collapse
|
34
|
N'Gouemo P. Voltage-Sensitive Calcium Channels in the Brain: Relevance to Alcohol Intoxication and Withdrawal. Handb Exp Pharmacol 2018; 248:263-280. [PMID: 29500720 DOI: 10.1007/164_2018_93] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Voltage-sensitive Ca2+ (CaV) channels are the primary route of depolarization-induced Ca2+ entry in neurons and other excitable cells, leading to an increase in intracellular Ca2+ concentration ([Ca2+]i). The resulting increase in [Ca2+]i activates a wide range of Ca2+-dependent processes in neurons, including neurotransmitter release, gene transcription, activation of Ca2+-dependent enzymes, and activation of certain K+ channels and chloride channels. In addition to their key roles under physiological conditions, CaV channels are also an important target of alcohol, and alcohol-induced changes in Ca2+ signaling can disturb neuronal homeostasis, Ca2+-mediated gene transcription, and the function of neuronal circuits, leading to various neurological and/or neuropsychiatric symptoms and disorders, including alcohol withdrawal induced-seizures and alcoholism.
Collapse
Affiliation(s)
- Prosper N'Gouemo
- Department of Pediatrics, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
35
|
Buonarati OR, Henderson PB, Murphy GG, Horne MC, Hell JW. Proteolytic processing of the L-type Ca 2+ channel alpha 11.2 subunit in neurons. F1000Res 2017; 6:1166. [PMID: 28781760 PMCID: PMC5531164 DOI: 10.12688/f1000research.11808.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/20/2017] [Indexed: 09/29/2023] Open
Abstract
Background: The L-type Ca2+ channel Cav1.2 is a prominent regulator of neuronal excitability, synaptic plasticity, and gene expression. The central element of Cav1.2 is the pore-forming α 11.2 subunit. It exists in two major size forms, whose molecular masses have proven difficult to precisely determine. Recent work suggests that α 11.2 is proteolytically cleaved between the second and third of its four pore-forming domains (Michailidis et al,. 2014). Methods: To better determine the apparent molecular masses (M R)of the α 11.2 size forms, extensive systematic immunoblotting of brain tissue as well as full length and C-terminally truncated α 11.2 expressed in HEK293 cells was conducted using six different region-specific antibodies against α 11.2. Results: The full length form of α 11.2 migrated, as expected, with an apparent M R of ~250 kDa. A shorter form of comparable prevalence with an apparent M R of ~210 kDa could only be detected in immunoblots probed with antibodies recognizing α 11.2 at an epitope 400 or more residues upstream of the C-terminus. Conclusions: The main two size forms of α 11.2 are the full length form and a shorter form, which lacks ~350 distal C-terminal residues. Midchannel cleavage as suggested by Michailidis et al. (2014) is at best minimal in brain tissue.
Collapse
Affiliation(s)
| | | | - Geoffrey G. Murphy
- Department of Molecular and Integrative Physiology, Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Mary C. Horne
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Johannes W. Hell
- Department of Pharmacology, University of California, Davis, CA, USA
| |
Collapse
|
36
|
Buonarati OR, Henderson PB, Murphy GG, Horne MC, Hell JW. Proteolytic processing of the L-type Ca 2+ channel alpha 11.2 subunit in neurons. F1000Res 2017; 6:1166. [PMID: 28781760 PMCID: PMC5531164 DOI: 10.12688/f1000research.11808.2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/14/2018] [Indexed: 12/30/2022] Open
Abstract
Background: The L-type Ca2+ channel Cav1.2 is a prominent regulator of neuronal excitability, synaptic plasticity, and gene expression. The central element of Cav1.2 is the pore-forming α 11.2 subunit. It exists in two major size forms, whose molecular masses have proven difficult to precisely determine. Recent work suggests that α 11.2 is proteolytically cleaved between the second and third of its four pore-forming domains (Michailidis et al,. 2014). Methods: To better determine the apparent molecular masses (M R)of the α 11.2 size forms, extensive systematic immunoblotting of brain tissue as well as full length and C-terminally truncated α 11.2 expressed in HEK293 cells was conducted using six different region-specific antibodies against α 11.2. Results: The full length form of α 11.2 migrated, as expected, with an apparent M R of ~250 kDa. A shorter form of comparable prevalence with an apparent M R of ~210 kDa could only be detected in immunoblots probed with antibodies recognizing α 11.2 at an epitope 400 or more residues upstream of the C-terminus. Conclusions: The main two size forms of α 11.2 are the full length form and a shorter form, which lacks ~350 distal C-terminal residues. Midchannel cleavage as suggested by Michailidis et al. (2014) is at best minimal in brain tissue.
Collapse
Affiliation(s)
| | | | - Geoffrey G. Murphy
- Department of Molecular and Integrative Physiology, Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Mary C. Horne
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Johannes W. Hell
- Department of Pharmacology, University of California, Davis, CA, USA
| |
Collapse
|
37
|
Chi CH, Tang CY, Pan CY. Calmodulin modulates the Ca 2+-dependent inactivation and expression level of bovine Ca V2.2 expressed in HEK293T cells. IBRO Rep 2017; 2:63-71. [PMID: 30135934 PMCID: PMC6084911 DOI: 10.1016/j.ibror.2017.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/27/2017] [Accepted: 03/10/2017] [Indexed: 01/31/2023] Open
Abstract
Ca2+ influx through voltage-gated Ca2+ channels (CaVs) at the plasma membrane is the major pathway responsible for the elevation of the intracellular Ca2+ concentration ([Ca2+]i), which activates various physiological activities. Calmodulin (CaM) is known to be involved in the Ca2+-dependent inactivation (CDI) of several types of CaVs; however, little is known about how CaM modulates CaV2.2. Here, we expressed CaV2.2 with CaM or CaM mutants with a Ca2+-binding deficiency in HEK293T cells and measured the currents to characterize the CDI. The results showed that CaV2.2 displayed a fast inactivation with Ca2+ but not Ba2+ as the charge carrier; when CaV2.2 was co-expressed with CaM mutants with a Ca2+-binding deficiency, the level of inactivation decreased. Using glutathione S-transferase-tagged CaM or CaM mutants as the bait, we found that CaM could interact with the intracellular C-terminal fragment of CaV2.2 in the presence or absence of Ca2+. However, CaM and its mutants could not interact with this fragment when mutations were generated in the conserved amino acid residues of the CaM-binding site. CaV2.2 with mutations in the CaM-binding site showed a greatly reduced current that could be rescued by CaM12 (Ca2+-binding deficiency at the N-lobe) overexpression; in addition, CaM12 enhanced the total expression level of CaV2.2, but the ratio of CaV2.2 present in the membrane to the total fraction remained unchanged. Together, our data suggest that CaM, with different Ca2+-binding abilities, modulates not only the inactivation of CaV2.2 but also its expression to regulate Ca2+-related physiological activities.
Collapse
Affiliation(s)
- Chih-Hung Chi
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Chih-Yung Tang
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan
| | - Chien-Yuan Pan
- Department of Life Science, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
38
|
Moutal A, Li W, Wang Y, Ju W, Luo S, Cai S, François-Moutal L, Perez-Miller S, Hu J, Dustrude ET, Vanderah TW, Gokhale V, Khanna M, Khanna R. Homology-guided mutational analysis reveals the functional requirements for antinociceptive specificity of collapsin response mediator protein 2-derived peptides. Br J Pharmacol 2017; 175:2244-2260. [PMID: 28161890 DOI: 10.1111/bph.13737] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE N-type voltage-gated calcium (Cav 2.2) channels are critical determinants of increased neuronal excitability and neurotransmission accompanying persistent neuropathic pain. Although Cav 2.2 channel antagonists are recommended as first-line treatment for neuropathic pain, calcium-current blocking gabapentinoids inadequately alleviate chronic pain symptoms and often exhibit numerous side effects. Collapsin response mediator protein 2 (CRMP2) targets Cav 2.2 channels to the sensory neuron membrane and allosterically modulates their function. A 15-amino-acid peptide (CBD3), derived from CRMP2, disrupts the functional protein-protein interaction between CRMP2 and Cav 2.2 channels to inhibit calcium influx, transmitter release and acute, inflammatory and neuropathic pain. Here, we have mapped the minimal domain of CBD3 necessary for its antinociceptive potential. EXPERIMENTAL APPROACH Truncated as well as homology-guided mutant versions of CBD3 were generated and assessed using depolarization-evoked calcium influx in rat dorsal root ganglion neurons, binding between CRMP2 and Cav 2.2 channels, whole-cell voltage clamp electrophysiology and behavioural effects in two models of experimental pain: post-surgical pain and HIV-induced sensory neuropathy induced by the viral glycoprotein 120. KEY RESULTS The first six amino acids within CBD3 accounted for all in vitro activity and antinociception. Spinal administration of a prototypical peptide (TAT-CBD3-L5M) reversed pain behaviours. Homology-guided mutational analyses of these six amino acids identified at least two residues, Ala1 and Arg4, as being critical for antinociception in two pain models. CONCLUSIONS AND IMPLICATIONS These results identify an antinociceptive scaffold core in CBD3 that can be used for development of low MW mimetics of CBD3. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Aubin Moutal
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Wennan Li
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Yue Wang
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Weina Ju
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China.,Department of Pharmacology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Shizhen Luo
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Song Cai
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | | | | | - Jackie Hu
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Erik T Dustrude
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Todd W Vanderah
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Vijay Gokhale
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - May Khanna
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA.,Neuroscience Graduate Interdisciplinary Program, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
39
|
Trafficking of neuronal calcium channels. Neuronal Signal 2017; 1:NS20160003. [PMID: 32714572 PMCID: PMC7373241 DOI: 10.1042/ns20160003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 01/20/2017] [Accepted: 01/19/2017] [Indexed: 12/18/2022] Open
Abstract
Neuronal voltage-gated calcium channels (VGCCs) serve complex yet essential physiological functions via their pivotal role in translating electrical signals into intracellular calcium elevations and associated downstream signalling pathways. There are a number of regulatory mechanisms to ensure a dynamic control of the number of channels embedded in the plasma membrane, whereas alteration of the surface expression of VGCCs has been linked to various disease conditions. Here, we provide an overview of the mechanisms that control the trafficking of VGCCs to and from the plasma membrane, and discuss their implication in pathophysiological conditions and their potential as therapeutic targets.
Collapse
|
40
|
Jiang HH, Dong FW, Zhou J, Hu JM, Yang J, Nian Y. Cav2.2 and Cav3.1 calcium channel inhibitors from Valeriana jatamansi Jones. RSC Adv 2017. [DOI: 10.1039/c7ra07327e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In China, the roots and rhizomes of Valeriana jatamansi Jones are traditionally used to treat gastrointestinal and rheumatic pain. In the present study, a number of N- and T-type calcium channel inhibitors were initially isolated and identified.
Collapse
Affiliation(s)
- He-Hai Jiang
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences
- Ion Channel Research and Drug Development Center
- Kunming Institute of Zoology
- Chinese Academy of Sciences
- Kunming 650223
| | - Fa-Wu Dong
- State Key Laboratory of Phytochemistry and Plant Resources in West China
- Kunming Institute of Botany
- Chinese Academy of Sciences
- Kunming 650201
- P. R. China
| | - Jun Zhou
- State Key Laboratory of Phytochemistry and Plant Resources in West China
- Kunming Institute of Botany
- Chinese Academy of Sciences
- Kunming 650201
- P. R. China
| | - Jiang-Miao Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China
- Kunming Institute of Botany
- Chinese Academy of Sciences
- Kunming 650201
- P. R. China
| | - Jian Yang
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences
- Ion Channel Research and Drug Development Center
- Kunming Institute of Zoology
- Chinese Academy of Sciences
- Kunming 650223
| | - Yin Nian
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences
- Ion Channel Research and Drug Development Center
- Kunming Institute of Zoology
- Chinese Academy of Sciences
- Kunming 650223
| |
Collapse
|
41
|
Swart T, Hurley MJ. Calcium Channel Antagonists as Disease-Modifying Therapy for Parkinson's Disease: Therapeutic Rationale and Current Status. CNS Drugs 2016; 30:1127-1135. [PMID: 27826740 DOI: 10.1007/s40263-016-0393-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Parkinson's disease is a disabling hypokinetic neurological movement disorder in which the aetiology is unknown in the majority of cases. Current pharmacological treatments, though effective at restoring movement, are only symptomatic and do nothing to slow disease progression. Electrophysiological, epidemiological and neuropathological studies have implicated CaV1.3 subtype calcium channels in the pathogenesis of the disorder, and drugs with some selectivity for this ion channel (brain-penetrant dihydropyridine calcium channel blockers) are neuroprotective in animal models of the disease. Dihydropyridines have been safely used for decades to treat hypertension and other cardiovascular disorders. A phase II clinical trial found that isradipine was safely tolerated by patients with Parkinson's disease, and a phase III trial is currently underway to determine whether treatment with isradipine is neuroprotective and therefore able to slow the progression of Parkinson's disease. This manuscript reviews the current information about the use of dihydropyridines as therapy for Parkinson's disease and discusses the possible mechanism of action of these drugs, highlighting CaV1.3 calcium channels as a potential therapeutic target for neuroprotection in Parkinson's disease.
Collapse
Affiliation(s)
- Tara Swart
- MIT Sloan Executive Education, Cambridge, MA, 02142, USA
| | - Michael J Hurley
- Neuroepidemiology and Ageing Research, School of Public Health, Imperial College London, Burlington Danes Building, Hammersmith Campus, DuCane Road, London, W12 0NN, UK.
| |
Collapse
|
42
|
Garcia-Rill E, D’Onofrio S, Mahaffey S. Bottom-up Gamma: the Pedunculopontine Nucleus and Reticular Activating System. TRANSLATIONAL BRAIN RHYTHMICITY 2016; 1:49-53. [PMID: 28691105 PMCID: PMC5497760 DOI: 10.15761/tbr.1000109] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Gamma rhythms have been proposed to promote the feed forward or "bottom-up" flow of information from lower to higher regions in the brain during perception. On the other hand, beta rhythms have been proposed to represent feed back or "top-down" influence from higher regions to lower. The pedunculopontine nucleus (PPN) has been implicated in sleep-wake control and arousal, and is part of the reticular activating system (RAS). This review describes the properties of the cells in this nucleus. These properties are unique, and perhaps it is the particular characteristics of these cells that allow the PPN to be involved in a host of functions and disorders. The fact that all PPN neurons fire maximally at gamma band frequency regardless of electrophysiological or transmitter type, make this an unusual cell group. In other regions, for example in the cortex, cells with such a property represent only a sub-population. More importantly, the fact that this cell group's functions are related to the capacity to generate coherent activity at a preferred natural frequency, gamma band, speaks volumes about how the PPN functions. We propose that "bottom-up" gamma band influence arises in the RAS and contributes to the build-up of the background of activity necessary for preconscious awareness and gamma activity at cortical levels.
Collapse
Affiliation(s)
- E. Garcia-Rill
- Center for Translational Neuroscience, Department of Neurobiology, University of Arkansas for Medical Sciences., Little Rock, AR
| | - S. D’Onofrio
- Center for Translational Neuroscience, Department of Neurobiology, University of Arkansas for Medical Sciences., Little Rock, AR
| | - S. Mahaffey
- Center for Translational Neuroscience, Department of Neurobiology, University of Arkansas for Medical Sciences., Little Rock, AR
| |
Collapse
|
43
|
Wormuth C, Lundt A, Henseler C, Müller R, Broich K, Papazoglou A, Weiergräber M. Review: Ca v2.3 R-type Voltage-Gated Ca 2+ Channels - Functional Implications in Convulsive and Non-convulsive Seizure Activity. Open Neurol J 2016; 10:99-126. [PMID: 27843503 PMCID: PMC5080872 DOI: 10.2174/1874205x01610010099] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/16/2016] [Accepted: 06/24/2016] [Indexed: 11/22/2022] Open
Abstract
Background: Researchers have gained substantial insight into mechanisms of synaptic transmission, hyperexcitability, excitotoxicity and neurodegeneration within the last decades. Voltage-gated Ca2+ channels are of central relevance in these processes. In particular, they are key elements in the etiopathogenesis of numerous seizure types and epilepsies. Earlier studies predominantly targeted on Cav2.1 P/Q-type and Cav3.2 T-type Ca2+ channels relevant for absence epileptogenesis. Recent findings bring other channels entities more into focus such as the Cav2.3 R-type Ca2+ channel which exhibits an intriguing role in ictogenesis and seizure propagation. Cav2.3 R-type voltage gated Ca2+ channels (VGCC) emerged to be important factors in the pathogenesis of absence epilepsy, human juvenile myoclonic epilepsy (JME), and cellular epileptiform activity, e.g. in CA1 neurons. They also serve as potential target for various antiepileptic drugs, such as lamotrigine and topiramate. Objective: This review provides a summary of structure, function and pharmacology of VGCCs and their fundamental role in cellular Ca2+ homeostasis. We elaborate the unique modulatory properties of Cav2.3 R-type Ca2+ channels and point to recent findings in the proictogenic and proneuroapoptotic role of Cav2.3 R-type VGCCs in generalized convulsive tonic–clonic and complex-partial hippocampal seizures and its role in non-convulsive absence like seizure activity. Conclusion: Development of novel Cav2.3 specific modulators can be effective in the pharmacological treatment of epilepsies and other neurological disorders.
Collapse
Affiliation(s)
- Carola Wormuth
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Andreas Lundt
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Christina Henseler
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Ralf Müller
- Department of Psychiatry and Psychotherapy, University of Cologne, Faculty of Medicine, Cologne, Germany
| | - Karl Broich
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Anna Papazoglou
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Marco Weiergräber
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| |
Collapse
|
44
|
Bock T, Stuart GJ. The Impact of BK Channels on Cellular Excitability Depends on their Subcellular Location. Front Cell Neurosci 2016; 10:206. [PMID: 27630543 PMCID: PMC5006691 DOI: 10.3389/fncel.2016.00206] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/16/2016] [Indexed: 12/04/2022] Open
Abstract
Large conductance calcium-activated potassium channels (or BK channels) fulfil a multitude of roles in the central nervous system. At the soma of many neuronal cell types they control the speed of action potential (AP) repolarization and therefore they can have an impact on neuronal excitability. Due to their presence in nerve terminals they also regulate transmitter release. BK channels have also been shown to be present in the dendrites of some neurons where they can regulate the magnitude and duration of dendritic spikes. Here, we investigate the impact of modulating the activation of BK channels at different locations on the cellular excitability of cortical layer 5 pyramidal neurons. We find that while somatic BK channels help to repolarize APs at the soma and mediate the fast after-hyperpolarization, dendritic BK channels are responsible for repolarization of dendritic calcium spikes and thereby regulate somatic AP burst firing. We found no evidence for a role of dendritic BK channels in the regulation of backpropagating AP amplitude or duration. These experiments highlight the diverse roles of BK channels in regulating neuronal excitability and indicate that their functional impact depends on their subcellular location.
Collapse
Affiliation(s)
- Tobias Bock
- Eccles Institute of Neuroscience and Australian Research Council Centre of Excellence for Integrative Brain Function, John Curtin School of Medical Research, Australian National University Canberra, ACT, Australia
| | - Greg J Stuart
- Eccles Institute of Neuroscience and Australian Research Council Centre of Excellence for Integrative Brain Function, John Curtin School of Medical Research, Australian National University Canberra, ACT, Australia
| |
Collapse
|
45
|
Alles SRA, Smith PA. The Anti-Allodynic Gabapentinoids: Myths, Paradoxes, and Acute Effects. Neuroscientist 2016; 23:40-55. [PMID: 27118808 DOI: 10.1177/1073858416628793] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The gabapentinoids (pregabalin and gabapentin) are first line treatments for neuropathic pain. They exert their actions by binding to the α2δ accessory subunits of voltage-gated Ca2+ channels. Because these subunits interact with critical aspects of the neurotransmitter release process, gabapentinoid binding prevents transmission in nociceptive pathways. Gabapentinoids also reduce plasma membrane expression of voltage-gated Ca2+ channels but this may have little direct bearing on their therapeutic actions. In animal models of neuropathic pain, gabapentinoids exert an anti-allodynic action within 30 minutes but most of their in vitro effects are 30-fold slower, taking at least 17 hours to develop. This difference may relate to increased levels of α2δ expression in the injured nervous system. Thus, in situations where α2δ is experimentally upregulated in vitro, gabapentinoids act within minutes to interrupt trafficking of α2δ subunits to the plasma membrane within nerve terminals. When α2δ is not up-regulated, gabapentinoids act slowly to interrupt trafficking of α2δ protein from cell bodies to nerve terminals. This improved understanding of the mechanism of gabapentinoid action is related to their slowly developing actions in neuropathic pain patients, to the concept that different processes underlie the onset and maintenance of neuropathic pain and to the use of gabapentinoids in management of postsurgical pain.
Collapse
Affiliation(s)
- Sascha R A Alles
- 1 Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Peter A Smith
- 1 Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
46
|
Dolphin AC. Voltage-gated calcium channels and their auxiliary subunits: physiology and pathophysiology and pharmacology. J Physiol 2016; 594:5369-90. [PMID: 27273705 PMCID: PMC5043047 DOI: 10.1113/jp272262] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Voltage‐gated calcium channels are essential players in many physiological processes in excitable cells. There are three main subdivisions of calcium channel, defined by the pore‐forming α1 subunit, the CaV1, CaV2 and CaV3 channels. For all the subtypes of voltage‐gated calcium channel, their gating properties are key for the precise control of neurotransmitter release, muscle contraction and cell excitability, among many other processes. For the CaV1 and CaV2 channels, their ability to reach their required destinations in the cell membrane, their activation and the fine tuning of their biophysical properties are all dramatically influenced by the auxiliary subunits that associate with them. Furthermore, there are many diseases, both genetic and acquired, involving voltage‐gated calcium channels. This review will provide a general introduction and then concentrate particularly on the role of auxiliary α2δ subunits in both physiological and pathological processes involving calcium channels, and as a therapeutic target.
![]()
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
47
|
Aviner B, Gradwohl G, Bliznyuk A, Grossman Y. Selective pressure modulation of synaptic voltage-dependent calcium channels-involvement in HPNS mechanism. J Cell Mol Med 2016; 20:1872-88. [PMID: 27273194 PMCID: PMC5020619 DOI: 10.1111/jcmm.12877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 03/21/2016] [Indexed: 11/28/2022] Open
Abstract
Exposure to hyperbaric pressure (HP) exceeding 100 msw (1.1 MPa) is known to cause a constellation of motor and cognitive impairments named high-pressure neurological syndrome (HPNS), considered to be the result of synaptic transmission alteration. Long periods of repetitive HP exposure could be an occupational risk for professional deep-sea divers. Previous studies have indicated the modulation of presynaptic Ca(2+) currents based on synaptic activity modified by HP. We have recently demonstrated that currents in genetically identified cellular voltage-dependent Ca(2+) channels (VDCCs), CaV 1.2 and CaV 3.2 are selectively affected by HP. This work further elucidates the HPNS mechanism by examining HP effect on Ca(2+) currents in neuronal VDCCs, CaV 2.2 and CaV 2.1, which are prevalent in presynaptic terminals, expressed in Xenopus oocytes. HP augmented the CaV 2.2 current amplitude, much less so in a channel variation containing an additional modulatory subunit, and had almost no effect on the CaV 2.1 currents. HP differentially affected the channels' kinetics. It is, therefore, suggested that HPNS signs and symptoms arise, at least in part, from pressure modulation of various VDCCs.
Collapse
Affiliation(s)
- Ben Aviner
- Department of Physiology and Neurobiology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Gideon Gradwohl
- Department of Physics, Jerusalem College of Technology, Jerusalem, Israel
| | - Alice Bliznyuk
- Department of Physiology and Neurobiology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yoram Grossman
- Department of Physiology and Neurobiology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
48
|
Chang Q, Martin LJ. Voltage-gated calcium channels are abnormal in cultured spinal motoneurons in the G93A-SOD1 transgenic mouse model of ALS. Neurobiol Dis 2016; 93:78-95. [PMID: 27151771 DOI: 10.1016/j.nbd.2016.04.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 04/01/2016] [Accepted: 04/29/2016] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive loss of motoneurons. Hyperexcitability and excitotoxicity have been implicated in the early pathogenesis of ALS. Studies addressing excitotoxic motoneuron death and intracellular Ca(2+) overload have mostly focused on Ca(2+) influx through AMPA glutamate receptors. However, intrinsic excitability of motoneurons through voltage-gated ion channels may also have a role in the neurodegeneration. In this study we examined the function and localization of voltage-gated Ca(2+) channels in cultured spinal cord motoneurons from mice expressing a mutant form of human superoxide dismutase-1 with a Gly93→Ala substitution (G93A-SOD1). Using whole-cell patch-clamp recordings, we showed that high voltage activated (HVA) Ca(2+) currents are increased in G93A-SOD1 motoneurons, but low voltage activated Ca(2+) currents are not affected. G93A-SOD1 motoneurons also have altered persistent Ca(2+) current mediated by L-type Ca(2+) channels. Quantitative single-cell RT-PCR revealed higher levels of Ca1a, Ca1b, Ca1c, and Ca1e subunit mRNA expression in G93A-SOD1 motoneurons, indicating that the increase of HVA Ca(2+) currents may result from upregulation of Ca(2+) channel mRNA expression in motoneurons. The localizations of the Ca1B N-type and Ca1D L-type Ca(2+) channels in motoneurons were examined by immunocytochemistry and confocal microscopy. G93A-SOD1 motoneurons had increased Ca1B channels on the plasma membrane of soma and dendrites. Ca1D channels are similar on the plasma membrane of soma and lower on the plasma membrane of dendrites of G93A-SOD1 motoneurons. Our study demonstrates that voltage-gated Ca(2+) channels have aberrant functions and localizations in ALS mouse motoneurons. The increased HVA Ca(2+) currents and PCCa current could contribute to early pathogenesis of ALS.
Collapse
Affiliation(s)
- Qing Chang
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, MD 21205, United States.
| | - Lee J Martin
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, MD 21205, United States; Department of Neuroscience, Johns Hopkins University School of Medicine, MD 21205, United States
| |
Collapse
|
49
|
Tay A, Kunze A, Murray C, Di Carlo D. Induction of Calcium Influx in Cortical Neural Networks by Nanomagnetic Forces. ACS NANO 2016; 10:2331-41. [PMID: 26805612 DOI: 10.1021/acsnano.5b07118] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Nanomagnetic force stimulation with ferromagnetic nanoparticles was found to trigger calcium influx in cortical neural networks without observable cytotoxicity. Stimulated neural networks showed an average of 20% increment in calcium fluorescence signals and a heightened frequency in calcium spiking. These effects were also confined spatially to areas with engineered high magnetic field gradients. Furthermore, blockage of N-type calcium channels inhibited the stimulatory effects of the nanomagnetic forces, suggesting the role of mechano-sensitive ion channels in mediating calcium influx.
Collapse
Affiliation(s)
- Andy Tay
- Department of Bioengineering, ‡California Nanosystems Institute, and §Jonsson Comprehensive Cancer Center, University of California , Los Angeles, California 90025, United States
| | - Anja Kunze
- Department of Bioengineering, ‡California Nanosystems Institute, and §Jonsson Comprehensive Cancer Center, University of California , Los Angeles, California 90025, United States
| | - Coleman Murray
- Department of Bioengineering, ‡California Nanosystems Institute, and §Jonsson Comprehensive Cancer Center, University of California , Los Angeles, California 90025, United States
| | - Dino Di Carlo
- Department of Bioengineering, ‡California Nanosystems Institute, and §Jonsson Comprehensive Cancer Center, University of California , Los Angeles, California 90025, United States
| |
Collapse
|
50
|
Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 2015; 67:821-70. [PMID: 26362469 PMCID: PMC4630564 DOI: 10.1124/pr.114.009654] [Citation(s) in RCA: 773] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium channels are required for many key functions in the body. In this review, the different subtypes of voltage-gated calcium channels are described and their physiologic roles and pharmacology are outlined. We describe the current uses of drugs interacting with the different calcium channel subtypes and subunits, as well as specific areas in which there is strong potential for future drug development. Current therapeutic agents include drugs targeting L-type Ca(V)1.2 calcium channels, particularly 1,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Ca(V)3) channels are a target of ethosuximide, widely used in absence epilepsy. The auxiliary subunit α2δ-1 is the therapeutic target of the gabapentinoid drugs, which are of value in certain epilepsies and chronic neuropathic pain. The limited use of intrathecal ziconotide, a peptide blocker of N-type (Ca(V)2.2) calcium channels, as a treatment of intractable pain, gives an indication that these channels represent excellent drug targets for various pain conditions. We describe how selectivity for different subtypes of calcium channels (e.g., Ca(V)1.2 and Ca(V)1.3 L-type channels) may be achieved in the future by exploiting differences between channel isoforms in terms of sequence and biophysical properties, variation in splicing in different target tissues, and differences in the properties of the target tissues themselves in terms of membrane potential or firing frequency. Thus, use-dependent blockers of the different isoforms could selectively block calcium channels in particular pathologies, such as nociceptive neurons in pain states or in epileptic brain circuits. Of important future potential are selective Ca(V)1.3 blockers for neuropsychiatric diseases, neuroprotection in Parkinson's disease, and resistant hypertension. In addition, selective or nonselective T-type channel blockers are considered potential therapeutic targets in epilepsy, pain, obesity, sleep, and anxiety. Use-dependent N-type calcium channel blockers are likely to be of therapeutic use in chronic pain conditions. Thus, more selective calcium channel blockers hold promise for therapeutic intervention.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Joerg Striessnig
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Alexandra Koschak
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Annette C Dolphin
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| |
Collapse
|