1
|
Dai F, Hu C, Li X, Zhang Z, Wang H, Zhou W, Wang J, Geng Q, Dong Y, Tang C. Cav3.2 channel regulates cerebral ischemia/reperfusion injury: a promising target for intervention. Neural Regen Res 2024; 19:2480-2487. [PMID: 38526284 PMCID: PMC11090426 DOI: 10.4103/1673-5374.390966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/05/2023] [Accepted: 10/25/2023] [Indexed: 03/26/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202419110-00028/figure1/v/2024-03-08T184507Z/r/image-tiff Calcium influx into neurons triggers neuronal death during cerebral ischemia/reperfusion injury. Various calcium channels are involved in cerebral ischemia/reperfusion injury. Cav3.2 channel is a main subtype of T-type calcium channels. T-type calcium channel blockers, such as pimozide and mibefradil, have been shown to prevent cerebral ischemia/reperfusion injury-induced brain injury. However, the role of Cav3.2 channels in cerebral ischemia/reperfusion injury remains unclear. Here, in vitro and in vivo models of cerebral ischemia/reperfusion injury were established using middle cerebral artery occlusion in mice and high glucose hypoxia/reoxygenation exposure in primary hippocampal neurons. The results showed that Cav3.2 expression was significantly upregulated in injured hippocampal tissue and primary hippocampal neurons. We further established a Cav3.2 gene-knockout mouse model of cerebral ischemia/reperfusion injury. Cav3.2 knockout markedly reduced infarct volume and brain water content, and alleviated neurological dysfunction after cerebral ischemia/reperfusion injury. Additionally, Cav3.2 knockout attenuated cerebral ischemia/reperfusion injury-induced oxidative stress, inflammatory response, and neuronal apoptosis. In the hippocampus of Cav3.2-knockout mice, calcineurin overexpression offset the beneficial effect of Cav3.2 knockout after cerebral ischemia/reperfusion injury. These findings suggest that the neuroprotective function of Cav3.2 knockout is mediated by calcineurin/nuclear factor of activated T cells 3 signaling. Findings from this study suggest that Cav3.2 could be a promising target for treatment of cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Feibiao Dai
- Graduate School, Wannan Medical College, Wuhu, Anhui Province, China
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Chengyun Hu
- Graduate School, Wannan Medical College, Wuhu, Anhui Province, China
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Xue Li
- Graduate School, Wannan Medical College, Wuhu, Anhui Province, China
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Zhetao Zhang
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Hongtao Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Wanjun Zhou
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Jiawu Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Qingtian Geng
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Yongfei Dong
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Chaoliang Tang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| |
Collapse
|
2
|
Cai H, Chen S, Sun Y, Zheng T, Liu Y, Tao J, Zhang Y. Interleukin-22 receptor 1-mediated stimulation of T-type Ca 2+ channels enhances sensory neuronal excitability through the tyrosine-protein kinase Lyn-dependent PKA pathway. Cell Commun Signal 2024; 22:307. [PMID: 38831315 PMCID: PMC11145867 DOI: 10.1186/s12964-024-01688-6] [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: 03/31/2024] [Accepted: 05/28/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Interleukin 24 (IL-24) has been implicated in the nociceptive signaling. However, direct evidence and the precise molecular mechanism underlying IL-24's role in peripheral nociception remain unclear. METHODS Using patch clamp recording, molecular biological analysis, immunofluorescence labeling, siRNA-mediated knockdown approach and behavior tests, we elucidated the effects of IL-24 on sensory neuronal excitability and peripheral pain sensitivity mediated by T-type Ca2+ channels (T-type channels). RESULTS IL-24 enhances T-type channel currents (T-currents) in trigeminal ganglion (TG) neurons in a reversible and dose-dependent manner, primarily by activating the interleukin-22 receptor 1 (IL-22R1). Furthermore, we found that the IL-24-induced T-type channel response is mediated through tyrosine-protein kinase Lyn, but not its common downstream target JAK1. IL-24 application significantly activated protein kinase A; this effect was independent of cAMP and prevented by Lyn antagonism. Inhibition of PKA prevented the IL-24-induced T-current response, whereas inhibition of protein kinase C or MAPK kinases had no effect. Functionally, IL-24 increased TG neuronal excitability and enhanced pain sensitivity to mechanical stimuli in mice, both of which were suppressed by blocking T-type channels. In a trigeminal neuropathic pain model induced by chronic constriction injury of the infraorbital nerve, inhibiting IL-22R1 signaling alleviated mechanical allodynia, which was reversed by blocking T-type channels or knocking down Cav3.2. CONCLUSION Our findings reveal that IL-24 enhances T-currents by stimulating IL-22R1 coupled to Lyn-dependent PKA signaling, leading to TG neuronal hyperexcitability and pain hypersensitivity. Understanding the mechanism of IL-24/IL-22R1 signaling in sensory neurons may pave the way for innovative therapeutic strategies in pain management.
Collapse
Affiliation(s)
- Hua Cai
- Clinical Research Center of Neurological Disease, Department of Geriatrics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, P.R. China
| | - Siyu Chen
- Clinical Research Center of Neurological Disease, Department of Geriatrics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, P.R. China
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Suzhou Medical College of Soochow University, Suzhou, 215123, P.R. China
| | - Yufang Sun
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Suzhou Medical College of Soochow University, Suzhou, 215123, P.R. China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123, P.R. China
| | - Tingting Zheng
- Clinical Research Center of Neurological Disease, Department of Geriatrics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, P.R. China
| | - Yulu Liu
- Clinical Research Center of Neurological Disease, Department of Geriatrics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, P.R. China
| | - Jin Tao
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Suzhou Medical College of Soochow University, Suzhou, 215123, P.R. China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123, P.R. China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, 215123, P.R. China.
| | - Yuan Zhang
- Clinical Research Center of Neurological Disease, Department of Geriatrics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, P.R. China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123, P.R. China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, 215123, P.R. China.
| |
Collapse
|
3
|
Tiryaki ES, Arslan G, Günaydın C, Ayyıldız M, Ağar E. The role of HCN channels on the effects of T-type calcium channels and GABA A receptors in the absence epilepsy model of WAG/Rij rats. Pflugers Arch 2024; 476:337-350. [PMID: 38159130 DOI: 10.1007/s00424-023-02900-1] [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: 10/10/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
In this study we used ivabradine (IVA), a hyperpolarization-activated cyclic nucleotide-gated (HCN) channel blocker, to identify its effect on spike-wave discharges (SWDs); and aimed to determine the role of IVA on the effects of T-type calcium channel blocker NNC 55-0396, GABAA receptor agonist muscimol and antagonist bicuculline in male WAG/Rij rats. After tripolar electrodes for electrocorticogram (ECoG) recordings were placed on the WAG/Rij rats' skulls, 5, 10, and 20 mg/kg IVA were intraperitoneally administered for 7 consecutive days and ECoG recordings were obtained on days 0th, 3rd, 6th, and 7th for three hours before and after injections. While acute injection of 5, 10, and 20 mg/kg IVA did not affect the total number and the mean duration of SWDs, subacute administration (7 days) of IVA decreased the SWDs parameters 24 hours after the 7th injection. Interestingly, when IVA was administered again 24 hours after the 6th IVA injection, it increased the SWDs parameters. Western-blot analyses showed that HCN1 and HCN2 expressions decreased and HCN4 increased in the 5-month-old WAG/Rij rats compared to the 1-month-old WAG/Rij and 5-month-old native Wistar rats, while subacute IVA administration increased the levels of HCN1 and HCN2 channels, except HCN4. Subacute administration of IVA reduced the antiepileptic activity of NNC, while the proepileptic activity of muscimol and the antiepileptic activity of bicuculline were abolished. It might be suggested that subacute IVA administration reduces absence seizures by changing the HCN channel expressions in WAG/Rij rats, and this affects the T-type calcium channels and GABAA receptors.
Collapse
Affiliation(s)
- Emre Soner Tiryaki
- Department of Physiology, Faculty of Medicine, University of Ondokuz Mayıs, Samsun, Türkiye
| | - Gökhan Arslan
- Department of Physiology, Faculty of Medicine, University of Ondokuz Mayıs, Samsun, Türkiye.
| | - Caner Günaydın
- Department of Pharmacology, Faculty of Medicine, University of Samsun, Samsun, Türkiye
| | - Mustafa Ayyıldız
- Department of Physiology, Faculty of Medicine, University of Ondokuz Mayıs, Samsun, Türkiye
| | - Erdal Ağar
- Department of Physiology, Faculty of Medicine, University of Ondokuz Mayıs, Samsun, Türkiye
| |
Collapse
|
4
|
Gong Y, Liu R, Zha H, Dong D, Lu N, Yan H, Wan L, Nian Y. Analgesic Buxus alkaloids with Enhanced Selectivity for the Low-Voltage-Gated Calcium Channel Ca v 3.2 over Ca v 3.1 through a New Binding Mode. Angew Chem Int Ed Engl 2024; 63:e202313461. [PMID: 37997012 DOI: 10.1002/anie.202313461] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 11/25/2023]
Abstract
Low-voltage-gated calcium channels (LVGCCs; Cav 3.1-3.3) represent promising drug targets for epilepsy, pain, and essential tremor. At present, modulators with heightened selectivity for a subtype of LVGCCs are still highly desired. In this study we explored three classes of Buxus alkaloids and identified 9(10/19)abeo-artanes Buxusemine H and Buxusemine L (BXSL) as an unprecedented type of Cav 3.2 inhibitors. Particularly, BXSL exhibited Cav 3.2 inhibition comparable to Z944, a non-subtype-selective LVGCCs inhibitor under clinical trial. While lacking specificity for Cav 3.3, BXSL showed a 30-fold selectivity of Cav 3.2 over Cav 3.1. As compared to several well-known inhibitors, the experimental and computational studies suggested BXSL exhibits a distinct binding mode to Cav 3.2, notably through the essential interaction with serine-1543 in domain III. Furthermore, BXSL showed minimal impact on various recombinant and native nociceptive ion channels, while significantly reducing the excitability of isolated mouse dorsal root ganglion neurons. Animal studies in wild-type and Cav 3.2 knock-out mice revealed that BXSL (5 mg/kg), by inhibiting Cav 3.2, exhibits an analgesic effect equivalent to Z944 (10 mg/kg) or mibefradil (10 mg/kg). Moreover, we proposed a structural rationale for the high selectivity of 9(10/19)abeo-artane-type alkaloids towards Cav 3.2 over Cav 3.1. This study introduces a novel analgesic agent and valuable molecular insight for structure-based innovative Cav 3.2 drug development.
Collapse
Affiliation(s)
- Ye Gong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, P. R. China
| | - Rui Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, P. R. China
| | - Hongjing Zha
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, P. R. China
| | - Ding Dong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, P. R. China
| | - Nihong Lu
- Department of Respiratory Medicine, The Third People's Hospital of Kunming, Kunming, 650041, Yunnan, P. R. China
| | - Hui Yan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, P. R. China
| | - Luosheng Wan
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, P. R. China
| | - Yin Nian
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, P. R. China
| |
Collapse
|
5
|
Jiang S, Li WY, Gao BB, Ou YF, Yuan ZF, Zhao QS. Casuattimines A-N, fourteen new Lycopodium alkaloids from Lycopodiastrum casuarinoides with Ca v3.1 channel inhibitory activity. Bioorg Chem 2024; 142:106962. [PMID: 37992623 DOI: 10.1016/j.bioorg.2023.106962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/28/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023]
Abstract
Two new dimeric Lycopodium alkaloids, casuattimines A and B (1 and 2), along with twelve previously undescribed Lycopodium alkaloids, casuattimines C-N (3-14), and eight known Lycopodium alkaloids, were isolated from Lycopodiastrum casuarinoides. Casuattimines A and B (1 and 2) are the first two ether-linked Lycopodium alkaloid dimers. Casuattimines C and D (3 and 4) are unique Lycopodium alkaloids characterized by a long fatty acid chain. Structural elucidation was achieved through HRESIMS, NMR, and electronic circular dichroism (ECD) calculations. In addition, the absolute configurations of compounds 7, 13, and 14 were determined by single crystal X-ray diffraction. Compounds 1, 2, and 4 demonstrated notable Cav3.1 channel inhibitory activities presenting IC50 values of 10.75 ± 1.02 μM, 9.33 ± 0.79 μM, and 7.14 ± 0.86 μM, respectively. The dynamics of compound 4 against the Cav3.1 channel and preliminary structure-activity relationships of these active Lycopodium alkaloids were also discussed.
Collapse
Affiliation(s)
- Shuai Jiang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen-Yan Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bei-Bei Gao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu-Fei Ou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zai-Feng Yuan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qin-Shi Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
6
|
Ishida IG, Sethi S, Mohren TL, Abbott L, Maimon G. Neuronal calcium spikes enable vector inversion in the Drosophila brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.24.568537. [PMID: 38077032 PMCID: PMC10705278 DOI: 10.1101/2023.11.24.568537] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
A typical neuron signals to downstream cells when it is depolarized and firing sodium spikes. Some neurons, however, also fire calcium spikes when hyperpolarized. The function of such bidirectional signaling remains unclear in most circuits. Here we show how a neuron class that participates in vector computation in the fly central complex employs hyperpolarization-elicited calcium spikes to invert two-dimensional mathematical vectors. When cells switch from firing sodium to calcium spikes, this leads to a ~180° realignment between the vector encoded in the neuronal population and the fly's internal heading signal, thus inverting the vector. We show that the calcium spikes rely on the T-type calcium channel Ca-α1T, and argue, via analytical and experimental approaches, that these spikes enable vector computations in portions of angular space that would otherwise be inaccessible. These results reveal a seamless interaction between molecular, cellular and circuit properties for implementing vector math in the brain.
Collapse
Affiliation(s)
- Itzel G. Ishida
- Laboratory of Integrative Brain Function and Howard Hughes Medical Institute, The Rockefeller University, New York NY, USA
| | - Sachin Sethi
- Laboratory of Integrative Brain Function and Howard Hughes Medical Institute, The Rockefeller University, New York NY, USA
| | - Thomas L. Mohren
- Laboratory of Integrative Brain Function and Howard Hughes Medical Institute, The Rockefeller University, New York NY, USA
| | - L.F. Abbott
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York NY, USA
| | - Gaby Maimon
- Laboratory of Integrative Brain Function and Howard Hughes Medical Institute, The Rockefeller University, New York NY, USA
| |
Collapse
|
7
|
Zhang Y, Wei Y, Zheng T, Tao Y, Sun Y, Jiang D, Tao J. Adiponectin receptor 1-mediated stimulation of Cav3.2 channels in trigeminal ganglion neurons induces nociceptive behaviors in mice. J Headache Pain 2023; 24:117. [PMID: 37620777 PMCID: PMC10463856 DOI: 10.1186/s10194-023-01658-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Adipokines, including adiponectin, are implicated in nociceptive pain; however, the underlying cellular and molecular mechanisms remain unknown. METHODS Using electrophysiological recording, immunostaining, molecular biological approaches and animal behaviour tests, we elucidated a pivotal role of adiponectin in regulating membrane excitability and pain sensitivity by manipulating Cav3.2 channels in trigeminal ganglion (TG) neurons. RESULTS Adiponectin enhanced T-type Ca2+ channel currents (IT) in TG neurons through the activation of adiponectin receptor 1 (adipoR1) but independently of heterotrimeric G protein-mediated signaling. Coimmunoprecipitation revealed a physical association between AdipoR1 and casein kinase II alpha-subunits (CK2α) in the TG, and inhibiting CK2 activity by chemical inhibitor or siRNA targeting CK2α prevented the adiponectin-induced IT response. Adiponectin significantly activated protein kinase C (PKC), and this effect was abrogated by CK2α knockdown. Adiponectin increased the membrane abundance of PKC beta1 (PKCβ1). Blocking PKCβ1 pharmacologically or genetically abrogated the adiponectin-induced IT increase. In heterologous expression systems, activation of adipoR1 induced a selective enhancement of Cav3.2 channel currents, dependent on PKCβ1 signaling. Functionally, adiponectin increased TG neuronal excitability and induced mechanical pain hypersensitivity, both attenuated by T-type channel blockade. In a trigeminal neuralgia model induced by chronic constriction injury of infraorbital nerve, blockade of adipoR1 signaling suppressed mechanical allodynia, which was prevented by silencing Cav3.2. CONCLUSION Our study elucidates a novel signaling cascade wherein adiponectin stimulates TG Cav3.2 channels via adipoR1 coupled to a novel CK2α-dependent PKCβ1. This process induces neuronal hyperexcitability and pain hypersensitivity. Insight into adipoR-Cav3.2 signaling in sensory neurons provides attractive targets for pain treatment.
Collapse
Affiliation(s)
- Yuan Zhang
- Clinical Research Center of Neurological Disease & Department of Geriatrics, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004 People’s Republic of China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123 People’s Republic of China
| | - Yuan Wei
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123 People’s Republic of China
| | - Tingting Zheng
- Clinical Research Center of Neurological Disease & Department of Geriatrics, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004 People’s Republic of China
| | - Yu Tao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123 People’s Republic of China
| | - Yufang Sun
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123 People’s Republic of China
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123 People’s Republic of China
| | - Dongsheng Jiang
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, 81377 Munich, Germany
| | - Jin Tao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123 People’s Republic of China
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123 People’s Republic of China
| |
Collapse
|
8
|
Beckinghausen J, Ortiz-Guzman J, Lin T, Bachman B, Salazar Leon LE, Liu Y, Heck DH, Arenkiel BR, Sillitoe RV. The cerebellum contributes to generalized seizures by altering activity in the ventral posteromedial nucleus. Commun Biol 2023; 6:731. [PMID: 37454228 PMCID: PMC10349834 DOI: 10.1038/s42003-023-05100-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: 06/19/2022] [Accepted: 07/05/2023] [Indexed: 07/18/2023] Open
Abstract
Thalamo-cortical networks are central to seizures, yet it is unclear how these circuits initiate seizures. We test whether a facial region of the thalamus, the ventral posteromedial nucleus (VPM), is a source of generalized, convulsive motor seizures and if convergent VPM input drives the behavior. To address this question, we devise an in vivo optogenetic mouse model to elicit convulsive motor seizures by driving these inputs and perform single-unit recordings during awake, convulsive seizures to define the local activity of thalamic neurons before, during, and after seizure onset. We find dynamic activity with biphasic properties, raising the possibility that heterogenous activity promotes seizures. Virus tracing identifies cerebellar and cerebral cortical afferents as robust contributors to the seizures. Of these inputs, only microinfusion of lidocaine into the cerebellar nuclei blocks seizure initiation. Our data reveal the VPM as a source of generalized convulsive seizures, with cerebellar input providing critical signals.
Collapse
Affiliation(s)
- Jaclyn Beckinghausen
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA
| | - Joshua Ortiz-Guzman
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | - Tao Lin
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA
| | - Benjamin Bachman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Luis E Salazar Leon
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA
| | - Yu Liu
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, 103515 University Dr., Duluth, MN, USA
| | - Detlef H Heck
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, 103515 University Dr., Duluth, MN, USA
| | - Benjamin R Arenkiel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Roy V Sillitoe
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA.
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
9
|
Vitamin C Modes of Action in Calcium-Involved Signaling in the Brain. Antioxidants (Basel) 2023; 12:antiox12020231. [PMID: 36829790 PMCID: PMC9952025 DOI: 10.3390/antiox12020231] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Vitamin C (ascorbic acid) is well known for its potent antioxidant properties, as it can neutralize ROS and free radicals, thereby protecting cellular elements from oxidative stress. It predominantly exists as an ascorbate anion and after oxidation to dehydroascorbic acid and further breakdown, is removed from the cells. In nervous tissue, a progressive decrease in vitamin C level or its prolonged deficiency have been associated with an increased risk of disturbances in neurotransmission, leading to dysregulation in brain function. Therefore, understanding the regulatory function of vitamin C in antioxidant defence and identification of its molecular targets deserves more attention. One of the key signalling ions is calcium and a transient rise in its concentration is crucial for all neuronal processes. Extracellular Ca2+ influx (through specific ion channels) or Ca2+ release from intracellular stores (endoplasmic reticulum, mitochondria) are precisely controlled. Ca2+ regulates the functioning of the CNS, including growth, development, myelin formation, synthesis of catecholamines, modulation of neurotransmission and antioxidant protection. A growing body of evidence indicates a unique role for vitamin C in these processes. In this short review, we focus on vitamin C in the regulation of calcium-involved pathways under physiological and stress conditions in the brain.
Collapse
|
10
|
Abad-Rodríguez J, Brocca ME, Higuero AM. Glycans and Carbohydrate-Binding/Transforming Proteins in Axon Physiology. ADVANCES IN NEUROBIOLOGY 2023; 29:185-217. [PMID: 36255676 DOI: 10.1007/978-3-031-12390-0_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The mature nervous system relies on the polarized morphology of neurons for a directed flow of information. These highly polarized cells use their somatodendritic domain to receive and integrate input signals while the axon is responsible for the propagation and transmission of the output signal. However, the axon must perform different functions throughout development before being fully functional for the transmission of information in the form of electrical signals. During the development of the nervous system, axons perform environmental sensing functions, which allow them to navigate through other regions until a final target is reached. Some axons must also establish a regulated contact with other cells before reaching maturity, such as with myelinating glial cells in the case of myelinated axons. Mature axons must then acquire the structural and functional characteristics that allow them to perform their role as part of the information processing and transmitting unit that is the neuron. Finally, in the event of an injury to the nervous system, damaged axons must try to reacquire some of their immature characteristics in a regeneration attempt, which is mostly successful in the PNS but fails in the CNS. Throughout all these steps, glycans perform functions of the outermost importance. Glycans expressed by the axon, as well as by their surrounding environment and contacting cells, encode key information, which is fine-tuned by glycan modifying enzymes and decoded by glycan binding proteins so that the development, guidance, myelination, and electrical transmission functions can be reliably performed. In this chapter, we will provide illustrative examples of how glycans and their binding/transforming proteins code and decode instructive information necessary for fundamental processes in axon physiology.
Collapse
Affiliation(s)
- José Abad-Rodríguez
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain.
| | - María Elvira Brocca
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| | - Alonso Miguel Higuero
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| |
Collapse
|
11
|
Su D, Gong Y, Li S, Yang J, Nian Y. Cyclovirobuxine D, a cardiovascular drug from traditional Chinese medicine, alleviates inflammatory and neuropathic pain mainly via inhibition of voltage-gated Ca v3.2 channels. Front Pharmacol 2022; 13:1081697. [PMID: 36618940 PMCID: PMC9811679 DOI: 10.3389/fphar.2022.1081697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Cyclovirobuxine D (CVB-D), the main active constituent of traditional Chinese medicine Buxus microphylla, was developed as a safe and effective cardiovascular drug in China. B. microphylla has also been used to relieve various pain symptoms for centuries. In this study, we examined and uncovered strong and persistent analgesic effects of cyclovirobuxine D against several mouse models of pain, including carrageenan- and CFA-induced inflammatory pain and paclitaxel-mediated neuropathic hypersensitivity. Cyclovirobuxine D shows comparable analgesic effects by intraplantar or intraperitoneal administration. Cyclovirobuxine D potently inhibits voltage-gated Cav2.2 and Cav3.2 channels but has negligible effects on a diverse group of nociceptive ion channels distributed in primary afferent neurons, including Nav1.7, Nav1.8, TRPV1, TPRA1, TRPM8, ASIC3, P2X2 and P2X4. Moreover, inhibition of Cav3.2, rather than Cav2.2, plays a dominant role in attenuating the excitability of isolated dorsal root ganglion neurons and pain relieving effects of cyclovirobuxine D. Our work reveals that a currently in-use cardiovascular drug has strong analgesic effects mainly via blockade of Cav3.2 and provides a compelling rationale and foundation for conducting clinical studies to repurpose cyclovirobuxine D in pain management.
Collapse
Affiliation(s)
- Deyuan Su
- Key Laboratory of Animal Models and Human Disease Mechanisms/Key Laboratory of Bioactive Peptides of Yunnan Province, Ion Channel Research and Drug Development Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ye Gong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Songyu Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jian Yang
- Department of Biological Sciences, Columbia University, New York, NY, United States
| | - Yin Nian
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, China
| |
Collapse
|
12
|
Jin Y, Mao Y, Chen D, Tai Y, Hu R, Yang CL, Zhou J, Chen L, Liu X, Gu E, Jia C, Zhang Z, Tao W. Thalamocortical circuits drive remifentanil-induced postoperative hyperalgesia. J Clin Invest 2022; 132:158742. [PMID: 36519547 PMCID: PMC9754001 DOI: 10.1172/jci158742] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 10/18/2022] [Indexed: 12/15/2022] Open
Abstract
Remifentanil-induced hyperalgesia (RIH) is a severe but common postoperative clinical problem with elusive underlying neural mechanisms. Here, we discovered that glutamatergic neurons in the thalamic ventral posterolateral nucleus (VPLGlu) exhibited significantly elevated burst firing accompanied by upregulation of Cav3.1 T-type calcium channel expression and function in RIH model mice. In addition, we identified a glutamatergic neuronal thalamocortical circuit in the VPL projecting to hindlimb primary somatosensory cortex glutamatergic neurons (S1HLGlu) that mediated RIH. In vivo calcium imaging and multi-tetrode recordings revealed heightened S1HLGlu neuronal activity during RIH. Moreover, preoperative suppression of Cav3.1-dependent burst firing in VPLGlu neurons or chemogenetic inhibition of VPLGlu neuronal terminals in the S1HL abolished the increased S1HLGlu neuronal excitability while alleviating RIH. Our findings suggest that remifentanil induces postoperative hyperalgesia by upregulating T-type calcium channel-dependent burst firing in VPLGlu neurons to activate S1HLGlu neurons, thus revealing an ion channel-mediated neural circuit basis for RIH that can guide analgesic development.
Collapse
Affiliation(s)
- Yan Jin
- Stroke Center and Department of Neurology and,Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Yu Mao
- Stroke Center and Department of Neurology and,Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Danyang Chen
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Yingju Tai
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Rui Hu
- Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chen-Ling Yang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jing Zhou
- Department of head, neck, and breast Surgery, Western district of the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Lijian Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuesheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Erwei Gu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chunhui Jia
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Zhi Zhang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Wenjuan Tao
- Stroke Center and Department of Neurology and,Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
A Combination of Rosa Multiflora and Zizyphus Jujuba Enhance Sleep Quality in Anesthesia-Induced Mice. Int J Mol Sci 2022; 23:ijms232214177. [PMID: 36430653 PMCID: PMC9696267 DOI: 10.3390/ijms232214177] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
Sleep is an essential component of quality of life. The majority of people experience sleep problems that impact their quality of life. Melatonin is currently a representative sleep aid. However, it is classified as a prescription drug in most countries, and consumers cannot purchase it to improve their sleep. This sleep induction experiment in mice aimed to identify a natural combination product (NCP) that can create synergistic sleep-promoting effects. Based on the mechanism of action of sleep, we investigated whether phenomenological indicators of sleep quality change according to the intake of NCP. The sleep onset and sleep time of the mice that consumed the NCP found by this study were improved compared to the existing sleep aids. The mean melatonin level in the blood increased by 197% compared to the control. To our knowledge, this is the first study to demonstrate that Rosa multiflora Thunb. (Yeongsil) can promote sleep similarly to Zizyphus jujuba Miller (Sanjoin). The results indicate a preclinical study of NCPs containing Rosa multiflora Thunb and Zizyphus jujuba Miller developed by us showed significant differences in sleep incubation and duration depending on melatonin concentrations. Our results also suggest that increased melatonin concentrations in the blood are likely to improve sleep quality, especially regarding incubation periods.
Collapse
|
14
|
Loss of Rai1 enhances hippocampal excitability and epileptogenesis in mouse models of Smith-Magenis syndrome. Proc Natl Acad Sci U S A 2022; 119:e2210122119. [PMID: 36256819 PMCID: PMC9618093 DOI: 10.1073/pnas.2210122119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Smith–Magenis syndrome (SMS) is a neurodevelopmental disorder associated with autism and epileptic seizures. SMS is caused by losing one copy of the gene encoding retinoic acid induced 1 (RAI1), a ubiquitously expressed transcriptional regulator. To pinpoint brain regions and cell types contributing to neuronal hyperexcitability in SMS, we combined electrophysiology and three-dimensional imaging of Fos expression in the intact mouse brain. We found that Rai1-deficient hippocampal dentate gyrus granule cells (dGCs) show increased intrinsic excitability and enhanced glutamatergic synaptic transmission. Our findings indicate that Rai1 safeguards the hippocampal network from hyperexcitability and could help explain abnormal brain activity in SMS. Hyperexcitability of brain circuits is a common feature of autism spectrum disorders (ASDs). Genetic deletion of a chromatin-binding protein, retinoic acid induced 1 (RAI1), causes Smith–Magenis syndrome (SMS). SMS is a syndromic ASD associated with intellectual disability, autistic features, maladaptive behaviors, overt seizures, and abnormal electroencephalogram (EEG) patterns. The molecular and neural mechanisms underlying abnormal brain activity in SMS remain unclear. Here we show that panneural Rai1 deletions in mice result in increased seizure susceptibility and prolonged hippocampal seizure duration in vivo and increased dentate gyrus population spikes ex vivo. Brain-wide mapping of neuronal activity pinpointed selective cell types within the limbic system, including the hippocampal dentate gyrus granule cells (dGCs) that are hyperactivated by chemoconvulsant administration or sensory experience in Rai1-deficient brains. Deletion of Rai1 from glutamatergic neurons, but not from gamma-aminobutyric acidergic (GABAergic) neurons, was responsible for increased seizure susceptibility. Deleting Rai1 from the Emx1Cre-lineage glutamatergic neurons resulted in abnormal dGC properties, including increased excitatory synaptic transmission and increased intrinsic excitability. Our work uncovers the mechanism of neuronal hyperexcitability in SMS by identifying Rai1 as a negative regulator of dGC intrinsic and synaptic excitability.
Collapse
|
15
|
Fei F, Wang X, Xu C, Shi J, Gong Y, Cheng H, Lai N, Ruan Y, Ding Y, Wang S, Chen Z, Wang Y. Discrete subicular circuits control generalization of hippocampal seizures. Nat Commun 2022; 13:5010. [PMID: 36008421 PMCID: PMC9411516 DOI: 10.1038/s41467-022-32742-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 08/15/2022] [Indexed: 11/09/2022] Open
Abstract
Epilepsy is considered a circuit-level dysfunction associated with imbalanced excitation-inhibition, it is therapeutically necessary to identify key brain regions and related circuits in epilepsy. The subiculum is an essential participant in epileptic seizures, but the circuit mechanism underlying its role remains largely elusive. Here we deconstruct the diversity of subicular circuits in a mouse model of epilepsy. We find that excitatory subicular pyramidal neurons heterogeneously control the generalization of hippocampal seizures by projecting to different downstream regions. Notably, anterior thalamus-projecting subicular neurons bidirectionally mediate seizures, while entorhinal cortex-projecting subicular neurons act oppositely in seizure modulation. These two subpopulations are structurally and functionally dissociable. An intrinsically enhanced hyperpolarization-activated current and robust bursting intensity in anterior thalamus-projecting neurons facilitate synaptic transmission, thus contributing to the generalization of hippocampal seizures. These results demonstrate that subicular circuits have diverse roles in epilepsy, suggesting the necessity to precisely target specific subicular circuits for effective treatment of epilepsy.
Collapse
Affiliation(s)
- Fan Fei
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xia Wang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Jiaying Shi
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yiwei Gong
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Heming Cheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Nanxi Lai
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yeping Ruan
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yao Ding
- Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shuang Wang
- Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhong Chen
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China. .,Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China. .,Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yi Wang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China. .,Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China. .,Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
16
|
Abietane derived diterpenoids as Cav3.1 antagonists from Salvia digitaloides. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
17
|
The intersection of astrocytes and the endocannabinoid system in the lateral habenula: on the fast-track to novel rapid-acting antidepressants. Mol Psychiatry 2022; 27:3138-3149. [PMID: 35585261 DOI: 10.1038/s41380-022-01598-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 02/04/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022]
Abstract
Despite attaining significant advances toward better management of depressive disorders, we are still facing several setbacks. Developing rapid-acting antidepressants with sustained effects is an aspiration that requires thinking anew to explore possible novel targets. Recently, the lateral habenula (LHb), the brain's "anti-reward system", has been shown to go awry in depression in terms of various molecular and electrophysiological signatures. Some of the presumed contributors to such observed aberrations are astrocytes. These star-shaped cells of the brain can alter the firing pattern of the LHb, which keeps the activity of the midbrain's aminergic centers under tight control. Astrocytes are also integral parts of the tripartite synapses, and can therefore modulate synaptic plasticity and leave long-lasting changes in the brain. On the other hand, it was discovered that astrocytes express cannabinoid type 1 receptors (CB1R), which can also take part in long-term plasticity. Herein, we recount how the LHb of a depressed brain deviates from the "normal" one from a molecular perspective. We then try to touch upon the alterations of the endocannabinoid system in the LHb, and cast the idea that modulation of astroglial CB1R may help regulate habenular neuronal activity and synaptogenesis, thereby acting as a new pharmacological tool for regulation of mood and amelioration of depressive symptoms.
Collapse
|
18
|
Xia F, Du SZ, Wu MK, Liu R, Ye YS, Yang J, Xu G, Nian Y. Icetexane diterpenoids as Ca v3.2 T-type calcium channel inhibitors from Salvia prattii and analgesic effect of their Semi-synthesized derivatives. Bioorg Chem 2022; 128:106059. [PMID: 35933895 DOI: 10.1016/j.bioorg.2022.106059] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/13/2022] [Accepted: 07/21/2022] [Indexed: 11/02/2022]
Abstract
Ten new icetexane diterpenoids, salpratins E-N (1-10) and a known analogue (11) were characterized from Salvia prattii Hemsl. Structurally, 1 is the first 19(4 → 3)-abeo-icetexane diterpenoid featuring with a 6/7/6 ring system. The structures of isolated compounds were determined by comprehensive analyses of spectroscopic data, ECD calculation, and single-crystal X-ray diffraction. Biological studies initially revealed that 1, 7, 10, and 11 are notable Cav3.2 T-type Ca2+ channel (TTCC) inhibitors with IC50 values of 2.9, 5.1, 2.3, and 3.2 μM, respectively. Five icetexane related derivatives (13-17) were synthesized from an abietane type precursor, (+)-carnosic acid (12), for the purpose of overcoming the poor water solubility of aforementioned active compounds and further investigating diverse diterpenes with valuable activity. Among them, 13 and 14 showed potent inhibitions on Cav3.2, having IC50 values of 6.7 and 2.4 μM, respectively. Significantly, they exhibited dose-dependent (1, 3, and 10 mg/kg) and comparable analgesic effects as that of Z944, a TTCCs inhibitor under clinical trial for pain management, in the mouse acetic acid writhing test. These findings further enrich structural diversity and bioactivity of Salvia diterpenoids, as well as provide promising structural templates for the development of Cav3.2 analgesics.
Collapse
Affiliation(s)
- Fan Xia
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, People's Republic of China
| | - Shu-Zong Du
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, People's Republic of China; Key Laboratory of Animal Models and Human Disease Mechanisms, and Ion Channel Research and Drug Development Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Ming-Kun Wu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Rui Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, People's Republic of China
| | - Yan-Song Ye
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, People's Republic of China
| | - Jian Yang
- Department of Biological Sciences, Columbia University, New York NY 10027, USA
| | - Gang Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, People's Republic of China.
| | - Yin Nian
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, People's Republic of China.
| |
Collapse
|
19
|
Chang B, Byun J, Kim KK, Lee SE, Lee B, Kim KS, Ryu H, Shin HS, Cheong E. Deletion of Phospholipase C β1 in the Thalamic Reticular Nucleus Induces Absence Seizures. Exp Neurobiol 2022; 31:116-130. [PMID: 35674000 PMCID: PMC9194639 DOI: 10.5607/en22007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/22/2022] [Accepted: 04/05/2022] [Indexed: 11/29/2022] Open
Abstract
Absence seizures are caused by abnormal synchronized oscillations in the thalamocortical (TC) circuit, which result in widespread spike-and-wave discharges (SWDs) on electroencephalography (EEG) as well as impairment of consciousness. Thalamic reticular nucleus (TRN) and TC neurons are known to interact dynamically to generate TC circuitry oscillations during SWDs. Clinical studies have suggested the association of Plcβ1 with early-onset epilepsy, including absence seizures. However, the brain regions and circuit mechanisms related to the generation of absence seizures with Plcβ1 deficiency are unknown. In this study, we found that loss of Plcβ1 in mice caused spontaneous complex-type seizures, including convulsive and absence seizures. Importantly, TRN-specific deletion of Plcβ1 led to the development of only spontaneous SWDs, and no other types of seizures were observed. Ex vivo slice patch recording demonstrated that the number of spikes, an intrinsic TRN neuronal property, was significantly reduced in both tonic and burst firing modes in the absence of Plcβ1. We conclude that the loss of Plcβ1 in the TRN leads to decreased excitability and impairs normal inhibitory neuronal function, thereby disrupting feedforward inhibition of the TC circuitry, which is sufficient to cause hypersynchrony of the TC system and eventually leads to spontaneous absence seizures. Our study not only provides a novel mechanism for the induction of SWDs in Plcβ1-deficient patients but also offers guidance for the development of diagnostic and therapeutic tools for absence epilepsy.
Collapse
Affiliation(s)
- Bomi Chang
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea.,Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Junweon Byun
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Ko Keun Kim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Seung Eun Lee
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Boyoung Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Key-Sun Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Hee-Sup Shin
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
20
|
de Hoog E, Spencer GE. Activity-dependent modulation of neuronal K V channels by retinoic acid enhances Ca V channel activity. J Biol Chem 2022; 298:101959. [PMID: 35452677 PMCID: PMC9127218 DOI: 10.1016/j.jbc.2022.101959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 12/18/2022] Open
Abstract
The metabolite of vitamin A, retinoic acid (RA), is known to affect synaptic plasticity in the nervous system and to play an important role in learning and memory. A ubiquitous mechanism by which neuronal plasticity develops in the nervous system is through modulation of voltage-gated Ca2+ (CaV) and voltage-gated K+ channels. However, how retinoids might regulate the activity of these channels has not been determined. Here, we show that RA modulates neuronal firing by inducing spike broadening and complex spiking in a dose-dependent manner in peptidergic and dopaminergic cell types. Using patch-clamp electrophysiology, we show that RA-induced complex spiking is activity dependent and involves enhanced inactivation of delayed rectifier voltage-gated K+ channels. The prolonged depolarizations observed during RA-modulated spiking lead to an increase in Ca2+ influx through CaV channels, though we also show an opposing effect of RA on the same neurons to inhibit Ca2+ influx. At physiological levels of Ca2+, this inhibition is specific to CaV2 (not CaV1) channels. Examining the interaction between the spike-modulating effects of RA and its inhibition of CaV channels, we found that inhibition of CaV2 channels limits the Ca2+ influx resulting from spike modulation. Our data thus provide novel evidence to suggest that retinoid signaling affects both delayed rectifier K+ channels and CaV channels to fine-tune Ca2+ influx through CaV2 channels. As these channels play important roles in synaptic function, we propose that these modulatory effects of retinoids likely contribute to synaptic plasticity in the nervous system.
Collapse
Affiliation(s)
- Eric de Hoog
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock's Way, St Catharines, Ontario. Canada. L2S 3A1
| | - Gaynor E Spencer
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock's Way, St Catharines, Ontario. Canada. L2S 3A1.
| |
Collapse
|
21
|
Histone methylation-mediated microRNA-32-5p down-regulation in sensory neurons regulates pain behaviors via targeting Cav3.2 channels. Proc Natl Acad Sci U S A 2022; 119:e2117209119. [PMID: 35353623 PMCID: PMC9168926 DOI: 10.1073/pnas.2117209119] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In this study, we identify microRNA-32-5p (miR-32-5p) as a key functional noncoding RNA in trigeminal-mediated neuropathic pain. We report that injury-induced histone methylation attenuates the binding of glucocorticoid receptor to the promoter region of the miR-32-5p gene and decreases the expression of miR-32-5p, in turn promoting the development of neuropathic pain through regulation of Cav3.2 channels. miRNA-mediated gene regulation has been proposed as a therapeutic approach in neuropathic pain. Our findings identify miR-32-5p replenishment as a therapeutic strategy for treating chronic neuropathic pain. microRNA (miRNA)–mediated gene regulation has been studied as a therapeutic approach, but its functional regulatory mechanism in neuropathic pain is not well understood. Here, we identify that miRNA-32-5p (miR-32-5p) is a functional RNA in regulating trigeminal-mediated neuropathic pain. High-throughput sequencing and qPCR analysis showed that miR-32-5p was the most down-regulated miRNA in the injured trigeminal ganglion (TG) of rats. Intra-TG injection of miR-32-5p agomir or overexpression of miR-32-5p by lentiviral delivery in neurons of the injured TG attenuated established trigeminal neuropathic pain. miR-32-5p overexpression did not affect acute physiological pain, while miR-32-5p down-regulation in intact rats was sufficient to cause pain-related behaviors. Nerve injury increased the methylated histone occupancy of binding sites for the transcription factor glucocorticoid receptor in the miR-32-5p promoter region. Inhibition of the enzymes that catalyze H3K9me2 and H3K27me3 restored the expression of miR-32-5p and markedly attenuated pain behaviors. Further, miR-32-5p–targeted Cav3.2 T-type Ca2+ channels and decreased miR-32-5p associated with neuropathic pain caused an increase in Cav3.2 protein expression and T-type channel currents. Conversely, miR-32-5p overexpression in injured TG suppressed the increased expression of Cav3.2 and reversed mechanical allodynia. Together, we conclude that histone methylation-mediated miR-32-5p down-regulation in TG neurons regulates trigeminal neuropathic pain by targeting Cav3.2 channels.
Collapse
|
22
|
Papazoglou A, Arshaad MI, Henseler C, Daubner J, Broich K, Hescheler J, Ehninger D, Haenisch B, Weiergräber M. Ca v3 T-Type Voltage-Gated Ca 2+ Channels and the Amyloidogenic Environment: Pathophysiology and Implications on Pharmacotherapy and Pharmacovigilance. Int J Mol Sci 2022; 23:3457. [PMID: 35408817 PMCID: PMC8998330 DOI: 10.3390/ijms23073457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/07/2022] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) were reported to play a crucial role in neurotransmitter release, dendritic resonance phenomena and integration, and the regulation of gene expression. In the septohippocampal system, high- and low-voltage-activated (HVA, LVA) Ca2+ channels were shown to be involved in theta genesis, learning, and memory processes. In particular, HVA Cav2.3 R-type and LVA Cav3 T-type Ca2+ channels are expressed in the medial septum-diagonal band of Broca (MS-DBB), hippocampal interneurons, and pyramidal cells, and ablation of both channels was proven to severely modulate theta activity. Importantly, Cav3 Ca2+ channels contribute to rebound burst firing in septal interneurons. Consequently, functional impairment of T-type Ca2+ channels, e.g., in null mutant mouse models, caused tonic disinhibition of the septohippocampal pathway and subsequent enhancement of hippocampal theta activity. In addition, impairment of GABA A/B receptor transcription, trafficking, and membrane translocation was observed within the septohippocampal system. Given the recent findings that amyloid precursor protein (APP) forms complexes with GABA B receptors (GBRs), it is hypothesized that T-type Ca2+ current reduction, decrease in GABA receptors, and APP destabilization generate complex functional interdependence that can constitute a sophisticated proamyloidogenic environment, which could be of potential relevance in the etiopathogenesis of Alzheimer's disease (AD). The age-related downregulation of T-type Ca2+ channels in humans goes together with increased Aβ levels that could further inhibit T-type channels and aggravate the proamyloidogenic environment. The mechanistic model presented here sheds new light on recent reports about the potential risks of T-type Ca2+ channel blockers (CCBs) in dementia, as observed upon antiepileptic drug application in the elderly.
Collapse
Affiliation(s)
- Anna Papazoglou
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (A.P.); (M.I.A.); (C.H.); (J.D.)
| | - Muhammad Imran Arshaad
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (A.P.); (M.I.A.); (C.H.); (J.D.)
| | - Christina Henseler
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (A.P.); (M.I.A.); (C.H.); (J.D.)
| | - Johanna Daubner
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (A.P.); (M.I.A.); (C.H.); (J.D.)
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (K.B.); (B.H.)
| | - Jürgen Hescheler
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany;
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Dan Ehninger
- Translational Biogerontology, German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany;
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Britta Haenisch
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (K.B.); (B.H.)
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
- Center for Translational Medicine, Medical Faculty, University of Bonn, 53113 Bonn, Germany
| | - Marco Weiergräber
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (A.P.); (M.I.A.); (C.H.); (J.D.)
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (K.B.); (B.H.)
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany;
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| |
Collapse
|
23
|
Thalamic T-Type Calcium Channels as Targets for Hypnotics and General Anesthetics. Int J Mol Sci 2022; 23:ijms23042349. [PMID: 35216466 PMCID: PMC8876360 DOI: 10.3390/ijms23042349] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/19/2022] Open
Abstract
General anesthetics mainly act by modulating synaptic inhibition on the one hand (the potentiation of GABA transmission) or synaptic excitation on the other (the inhibition of NMDA receptors), but they can also have effects on numerous other proteins, receptors, and channels. The effects of general anesthetics on ion channels have been the subject of research since the publication of reports of direct actions of these drugs on ion channel proteins. In particular, there is considerable interest in T-type voltage-gated calcium channels that are abundantly expressed in the thalamus, where they control patterns of cellular excitability and thalamocortical oscillations during awake and sleep states. Here, we summarized and discussed our recent studies focused on the CaV3.1 isoform of T-channels in the nonspecific thalamus (intralaminar and midline nuclei), which acts as a key hub through which natural sleep and general anesthesia are initiated. We used mouse genetics and in vivo and ex vivo electrophysiology to study the role of thalamic T-channels in hypnosis induced by a standard general anesthetic, isoflurane, as well as novel neuroactive steroids. From the results of this study, we conclude that CaV3.1 channels contribute to thalamocortical oscillations during anesthetic-induced hypnosis, particularly the slow-frequency range of δ oscillations (0.5–4 Hz), by generating “window current” that contributes to the resting membrane potential. We posit that the role of the thalamic CaV3.1 isoform of T-channels in the effects of various classes of general anesthetics warrants consideration.
Collapse
|
24
|
Bortolin A, Neto E, Lamghari M. Calcium Signalling in Breast Cancer Associated Bone Pain. Int J Mol Sci 2022; 23:ijms23031902. [PMID: 35163823 PMCID: PMC8836937 DOI: 10.3390/ijms23031902] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 02/05/2023] Open
Abstract
Calcium (Ca2+) is involved as a signalling mediator in a broad variety of physiological processes. Some of the fastest responses in human body like neuronal action potential firing, to the slowest gene transcriptional regulation processes are controlled by pathways involving calcium signalling. Under pathological conditions these mechanisms are also involved in tumoral cells reprogramming, resulting in the altered expression of genes associated with cell proliferation, metastatisation and homing to the secondary metastatic site. On the other hand, calcium exerts a central function in nociception, from cues sensing in distal neurons, to signal modulation and interpretation in the central nervous system leading, in pathological conditions, to hyperalgesia, allodynia and pain chronicization. It is well known the relationship between cancer and pain when tumoral metastatic cells settle in the bones, especially in late breast cancer stage, where they alter the bone micro-environment leading to bone lesions and resulting in pain refractory to the conventional analgesic therapies. The purpose of this review is to address the Ca2+ signalling mechanisms involved in cancer cell metastatisation as well as the function of the same signalling tools in pain regulation and transmission. Finally, the possible interactions between these two cells types cohabiting the same Ca2+ rich environment will be further explored attempting to highlight new possible therapeutical targets.
Collapse
Affiliation(s)
- Andrea Bortolin
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal; (A.B.); (E.N.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal
- FEUP—Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Estrela Neto
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal; (A.B.); (E.N.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal
| | - Meriem Lamghari
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal; (A.B.); (E.N.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal
- Correspondence:
| |
Collapse
|
25
|
Studtmann C, Ladislav M, Topolski MA, Safari M, Swanger SA. NaV1.1 haploinsufficiency impairs glutamatergic and GABAergic neuron function in the thalamus. Neurobiol Dis 2022; 167:105672. [DOI: 10.1016/j.nbd.2022.105672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 02/08/2022] [Accepted: 02/22/2022] [Indexed: 11/16/2022] Open
|
26
|
Klemz A, Wildner F, Tütüncü E, Gerevich Z. Regulation of Hippocampal Gamma Oscillations by Modulation of Intrinsic Neuronal Excitability. Front Neural Circuits 2022; 15:778022. [PMID: 35177966 PMCID: PMC8845518 DOI: 10.3389/fncir.2021.778022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Ion channels activated around the subthreshold membrane potential determine the likelihood of neuronal firing in response to synaptic inputs, a process described as intrinsic neuronal excitability. Long-term plasticity of chemical synaptic transmission is traditionally considered the main cellular mechanism of information storage in the brain; however, voltage- and calcium-activated channels modulating the inputs or outputs of neurons are also subjects of plastic changes and play a major role in learning and memory formation. Gamma oscillations are associated with numerous higher cognitive functions such as learning and memory, but our knowledge of their dependence on intrinsic plasticity is by far limited. Here we investigated the roles of potassium and calcium channels activated at near subthreshold membrane potentials in cholinergically induced persistent gamma oscillations measured in the CA3 area of rat hippocampal slices. Among potassium channels, which are responsible for the afterhyperpolarization in CA3 pyramidal cells, we found that blockers of SK (KCa2) and KV7.2/7.3 (KCNQ2/3), but not the BK (KCa1.1) and IK (KCa3.1) channels, increased the power of gamma oscillations. On the contrary, activators of these channels had an attenuating effect without affecting the frequency. Pharmacological blockade of the low voltage-activated T-type calcium channels (CaV3.1–3.3) reduced gamma power and increased the oscillation peak frequency. Enhancement of these channels also inhibited the peak power without altering the frequency of the oscillations. The presented data suggest that voltage- and calcium-activated ion channels involved in intrinsic excitability strongly regulate the power of hippocampal gamma oscillations. Targeting these channels could represent a valuable pharmacological strategy against cognitive impairment.
Collapse
|
27
|
Abdelaal MS, Midorikawa M, Suzuki T, Kobayashi K, Takata N, Miyata M, Mimura M, Tanaka KF. OUP accepted manuscript. Brain Commun 2022; 4:fcac010. [PMID: 35243344 PMCID: PMC8887905 DOI: 10.1093/braincomms/fcac010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/02/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Spike-and-wave discharges and an accompanying loss of consciousness are hallmarks of absence seizure, which is a childhood generalized epilepsy disorder. In absence seizure, dysfunction of the cortico-thalamo-cortico circuitry is thought to engage in abnormal cortical rhythms. Previous studies demonstrated that the thalamic reticular nucleus has a critical role in the formation of normal cortical rhythms; however, whether thalamic reticular nucleus dysfunction leads directly to abnormal rhythms, such as epilepsy, is largely unknown. We found that expressing the inhibitory opsin, archaerhodopsin, including in the thalamic reticular nucleus, caused abnormal cortical rhythms in Pvalb-tetracycline transactivator::tetO-ArchT (PV-ArchT) double transgenic mice. We validated the PV-ArchT line as a new mouse model of absence seizure through physiological and pharmacological analyses, as well as through examining their behavioural features. We then discovered that archaerhodopsin expression exclusively in thalamic reticular nucleus parvalbumin-positive neurons was sufficient to induce cortical spike-and-wave discharges using adeno-associated virus-mediated thalamic reticular nucleus targeting. Furthermore, we found that archaerhodopsin expression impaired rebound burst firing and T-current in thalamic reticular nucleus parvalbumin-positive cells by slice physiology. Although T-current in the thalamic reticular nucleus was impaired, the T-current blocker ethosuximide still had a therapeutic effect in PV-ArchT mice, suggesting a gain of function of T-type calcium channels in this absence seizure model. However, we did not find any over- or misexpression of T-type calcium channel genes in the thalamus or the cortex. Thus, we demonstrated that thalamic reticular nucleus dysfunction led to an absence seizure-like phenotype in mice. In a final set of experiments, we showed that the archaerhodopsin-mediated absence seizure-like phenotype disappeared after the removal of archaerhodopsin by using a time-controllable transgenic system. These data may provide a hint as to why many absence seizures naturally regress.
Collapse
Affiliation(s)
- Manal S. Abdelaal
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Mitsuharu Midorikawa
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women’s Medical University, Tokyo 162-8666, Japan
| | - Toru Suzuki
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Norio Takata
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Mariko Miyata
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women’s Medical University, Tokyo 162-8666, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kenji F. Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan
- Correspondence to: Kenji F. Tanaka, MD, PhD Division of Brain Sciences, Institute for Advanced Medical Research Keio University School of Medicine 35 Shinanomachi Shinjuku-ku Tokyo 160-8582, Japan E-mail:
| |
Collapse
|
28
|
Maksemous N, Blayney CD, Sutherland HG, Smith RA, Lea RA, Tran KN, Ibrahim O, McArthur JR, Haupt LM, Cader MZ, Finol-Urdaneta RK, Adams DJ, Griffiths LR. Investigation of CACNA1I Cav3.3 Dysfunction in Hemiplegic Migraine. Front Mol Neurosci 2022; 15:892820. [PMID: 35928792 PMCID: PMC9345121 DOI: 10.3389/fnmol.2022.892820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/13/2022] [Indexed: 01/12/2023] Open
Abstract
Familial hemiplegic migraine (FHM) is a severe neurogenetic disorder for which three causal genes, CACNA1A, SCN1A, and ATP1A2, have been implicated. However, more than 80% of referred diagnostic cases of hemiplegic migraine (HM) are negative for exonic mutations in these known FHM genes, suggesting the involvement of other genes. Using whole-exome sequencing data from 187 mutation-negative HM cases, we identified rare variants in the CACNA1I gene encoding the T-type calcium channel Cav3.3. Burden testing of CACNA1I variants showed a statistically significant increase in allelic burden in the HM case group compared to gnomAD (OR = 2.30, P = 0.00005) and the UK Biobank (OR = 2.32, P = 0.0004) databases. Dysfunction in T-type calcium channels, including Cav3.3, has been implicated in a range of neurological conditions, suggesting a potential role in HM. Using patch-clamp electrophysiology, we compared the biophysical properties of five Cav3.3 variants (p.R111G, p.M128L, p.D302G, p.R307H, and p.Q1158H) to wild-type (WT) channels expressed in HEK293T cells. We observed numerous functional alterations across the channels with Cav3.3-Q1158H showing the greatest differences compared to WT channels, including reduced current density, right-shifted voltage dependence of activation and inactivation, and slower current kinetics. Interestingly, we also found significant differences in the conductance properties exhibited by the Cav3.3-R307H and -Q1158H variants compared to WT channels under conditions of acidosis and alkalosis. In light of these data, we suggest that rare variants in CACNA1I may contribute to HM etiology.
Collapse
Affiliation(s)
- Neven Maksemous
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Claire D Blayney
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Heidi G Sutherland
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Robert A Smith
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Rod A Lea
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kim Ngan Tran
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Omar Ibrahim
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jeffrey R McArthur
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Larisa M Haupt
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - M Zameel Cader
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - David J Adams
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
29
|
Sadrhaghighi G, Abbaszadeh S, Babataheri S, Garjani A, Soraya H. Effects of pre-treatment with metoprolol and diltiazem on cerebral ischemia/reperfusion-induced injuries. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e21086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
30
|
Zhao JJ, Li SY, Xia F, Hu YL, Nian Y, Xu G. Isoprenylated Flavonoids as Ca v3.1 Low Voltage-Gated Ca 2+ Channel Inhibitors from Salvia digitaloides. NATURAL PRODUCTS AND BIOPROSPECTING 2021; 11:671-678. [PMID: 33893991 PMCID: PMC8599534 DOI: 10.1007/s13659-021-00307-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/15/2021] [Indexed: 06/12/2023]
Abstract
Saldigones A-C (1, 3, 4), three new isoprenylated flavonoids with diverse flavanone, pterocarpan, and isoflavanone architectures, were characterized from the roots of Salvia digitaloides, together with a known isoprenylated flavanone (2). Notably, it's the first report of isoprenylated flavonoids from Salvia species. The structures of these isolates were elucidated by extensive spectroscopic analysis. All of the compounds were evaluated for their activities on Cav3.1 low voltage-gated Ca2+ channel (LVGCC), of which 2 strongly and dose-dependently inhibited Cav3.1 peak current.
Collapse
Affiliation(s)
- Jian-Jun Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Song-Yu Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Fan Xia
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, People's Republic of China
| | - Ya-Li Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Yin Nian
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, People's Republic of China.
| | - Gang Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, People's Republic of China.
| |
Collapse
|
31
|
Bueschke N, Amaral-Silva L, Hu M, Santin JM. Lactate ions induce synaptic plasticity to enhance output from the central respiratory network. J Physiol 2021; 599:5485-5504. [PMID: 34761806 DOI: 10.1113/jp282062] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/29/2021] [Indexed: 12/16/2022] Open
Abstract
Lactate ion sensing has emerged as a process that regulates ventilation during metabolic challenges. Most work has focused on peripheral sensing of lactate for the control of breathing. However, lactate also rises in the central nervous system (CNS) during disturbances to blood gas homeostasis and exercise. Using an amphibian model, we recently showed that lactate ions, independently of pH and pyruvate metabolism, act directly in the brainstem to increase respiratory-related motor outflow. This response had a long washout time and corresponded with potentiated excitatory synaptic strength of respiratory motoneurons. Thus, we tested the hypothesis that lactate ions enhance respiratory output using cellular mechanisms associated with long-term synaptic plasticity within motoneurons. In this study, we confirm that 2 mM sodium lactate, but not sodium pyruvate, increases respiratory motor output in brainstem-spinal cord preparations, persisting for 2 h upon the removal of lactate. Lactate also led to prolonged increases in the amplitude of AMPA-glutamate receptor (AMPAR) currents in individual motoneurons from brainstem slices. Both motor facilitation and AMPAR potentiation by lactate required classic effectors of synaptic plasticity, L-type Ca2+ channels and NMDA receptors, as part of the transduction process but did not correspond with increased expression of immediate-early genes often associated with activity-dependent neuronal plasticity. Altogether these results show that lactate ions enhance respiratory motor output by inducing conserved mechanisms of synaptic plasticity and suggest a new mechanism that may contribute to coupling ventilation to metabolic demands in vertebrates. KEY POINTS: Lactate ions, independently of pH and metabolism, induce long-term increases in respiratory-related motor outflow in American bullfrogs. Lactate triggers a persistent increase in strength of AMPA-glutamatergic synapses onto respiratory motor neurons. Long-term plasticity of motor output and synaptic strength by lactate involves L-type Ca2+ channels and NMDA-receptors as part of the transduction process. Enhanced AMPA receptor function in response to lactate in the intact network is causal for motor plasticity. In sum, well-conserved synaptic plasticity mechanisms couple the brainstem lactate ion concentration to respiratory motor drive in vertebrates.
Collapse
Affiliation(s)
- Nikolaus Bueschke
- Department of Biology, The University of North Carolina, Greensboro, NC, USA
| | - Lara Amaral-Silva
- Department of Biology, The University of North Carolina, Greensboro, NC, USA
| | - Min Hu
- Department of Biology, The University of North Carolina, Greensboro, NC, USA
| | - Joseph M Santin
- Department of Biology, The University of North Carolina, Greensboro, NC, USA
| |
Collapse
|
32
|
Zhang Y, Qian Z, Jiang D, Sun Y, Gao S, Jiang X, Wang H, Tao J. Neuromedin B receptor stimulation of Cav3.2 T-type Ca 2+ channels in primary sensory neurons mediates peripheral pain hypersensitivity. Theranostics 2021; 11:9342-9357. [PMID: 34646374 PMCID: PMC8490515 DOI: 10.7150/thno.62255] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 09/01/2021] [Indexed: 01/21/2023] Open
Abstract
Background: Neuromedin B (Nmb) is implicated in the regulation of nociception of sensory neurons. However, the underlying cellular and molecular mechanisms remain unknown. Methods: Using patch clamp recording, western blot analysis, immunofluorescent labelling, enzyme-linked immunosorbent assays, adenovirus-mediated shRNA knockdown and animal behaviour tests, we studied the effects of Nmb on the sensory neuronal excitability and peripheral pain sensitivity mediated by Cav3.2 T-type channels. Results: Nmb reversibly and concentration-dependently increased T-type channel currents (IT) in small-sized trigeminal ganglion (TG) neurons through the activation of neuromedin B receptor (NmbR). This NmbR-mediated IT response was Gq protein-coupled, but independent of protein kinase C activity. Either intracellular application of the QEHA peptide or shRNA-mediated knockdown of Gβ abolished the NmbR-induced IT response. Inhibition of protein kinase A (PKA) or AMP-activated protein kinase (AMPK) completely abolished the Nmb-induced IT response. Analysis of phospho-AMPK (p-AMPK) revealed that Nmb significantly activated AMPK, while AMPK inhibition prevented the Nmb-induced increase in PKA activity. In a heterologous expression system, activation of NmbR significantly enhanced the Cav3.2 channel currents, while the Cav3.1 and Cav3.3 channel currents remained unaffected. Nmb induced TG neuronal hyperexcitability and concomitantly induced mechanical and thermal hypersensitivity, both of which were attenuated by T-type channel blockade. Moreover, blockade of NmbR signalling prevented mechanical hypersensitivity in a mouse model of complete Freund's adjuvant-induced inflammatory pain, and this effect was attenuated by siRNA knockdown of Cav3.2. Conclusions: Our study reveals a novel mechanism by which NmbR stimulates Cav3.2 channels through a Gβγ-dependent AMPK/PKA pathway. In mouse models, this mechanism appears to drive the hyperexcitability of TG neurons and induce pain hypersensitivity.
Collapse
|
33
|
Coulter I, Timic Stamenic T, Eggan P, Fine BR, Corrigan T, Covey DF, Yang L, Pan JQ, Todorovic SM. Different roles of T-type calcium channel isoforms in hypnosis induced by an endogenous neurosteroid epipregnanolone. Neuropharmacology 2021; 197:108739. [PMID: 34339750 PMCID: PMC8478885 DOI: 10.1016/j.neuropharm.2021.108739] [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: 03/19/2021] [Revised: 07/09/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Many neuroactive steroids induce sedation/hypnosis by potentiating γ-aminobutyric acid (GABAA) currents. However, we previously demonstrated that an endogenous neuroactive steroid epipregnanolone [(3β,5β)-3-hydroxypregnan-20-one] (EpiP) exerts potent peripheral analgesia and blocks T-type calcium currents while sparing GABAA currents in rat sensory neurons. This study seeks to investigate the behavioral effects elicited by systemic administration of EpiP and to characterize its use as an adjuvant agent to commonly used general anesthetics (GAs). METHODS Here, we utilized electroencephalographic (EEG) recordings to characterize thalamocortical oscillations, as well as behavioral assessment and mouse genetics with wild-type (WT) and different knockout (KO) models of T-channel isoforms to investigate potential sedative/hypnotic and immobilizing properties of EpiP. RESULTS Consistent with increased oscillations in slower EEG frequencies, EpiP induced an hypnotic state in WT mice when injected alone intra-peritoneally (i.p.) and effectively facilitated anesthetic effects of isoflurane (ISO) and sevoflurane (SEVO). The CaV3.1 (Cacna1g) KO mice demonstrated decreased sensitivity to EpiP-induced hypnosis when compared to WT mice, whereas no significant difference was noted between CaV3.2 (Cacna1h), CaV3.3 (Cacna1i) and WT mice. Finally, when compared to WT mice, onset of EpiP-induced hypnosis was delayed in CaV3.2 KO mice but not in CaV3.1 and CaV3.3 KO mice. CONCLUSION We posit that EpiP may have an important role as novel hypnotic and/or adjuvant to volatile anesthetic agents. We speculate that distinct hypnotic effects of EpiP across all three T-channel isoforms is due to their differential expression in thalamocortical circuitry.
Collapse
Affiliation(s)
- Ian Coulter
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Tamara Timic Stamenic
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Pierce Eggan
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Brier R. Fine
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Timothy Corrigan
- Department of Pediatrics, Division of Neurology,
Translational Epilepsy Research Program, University of Colorado, Anschutz Medical
Campus, Aurora, CO 80045, USA
| | - Douglas F. Covey
- Department of Developmental Biology, Washington University
School of Medicine, St. Louis, MO 63110, USA;,Taylor Family Institute for Innovative Psychiatric
Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lingling Yang
- Stanley Center for Psychiatric Research, Broad Institute of
Harvard and MIT
| | - Jen Q. Pan
- Stanley Center for Psychiatric Research, Broad Institute of
Harvard and MIT
| | - Slobodan M. Todorovic
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045;,Neuroscience, University of Colorado, Anschutz Medical
Campus, Aurora 80045;,Pharmacology Graduate Programs, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| |
Collapse
|
34
|
Harding EK, Dedek A, Bonin RP, Salter MW, Snutch TP, Hildebrand ME. The T-type calcium channel antagonist, Z944, reduces spinal excitability and pain hypersensitivity. Br J Pharmacol 2021; 178:3517-3532. [PMID: 33871884 PMCID: PMC8453510 DOI: 10.1111/bph.15498] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/02/2021] [Accepted: 04/05/2021] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose T‐type voltage‐gated calcium channels are an emerging therapeutic target for neurological disorders including epilepsy and pain. Inhibition of T‐type channels reduces the excitability of peripheral nociceptive sensory neurons and reverses pain hypersensitivity in male rodent pain models. However, administration of peripherally restricted T‐type antagonists failed to show efficacy in multiple clinical and preclinical pain trials, suggesting that inhibition of peripheral T‐type channels alone may be insufficient for pain relief. Experimental Approach We utilized the selective and CNS‐penetrant T‐type channel antagonist, Z944, in electrophysiological, calcium imaging and behavioural paradigms to determine its effect on lamina I neuron excitability and inflammatory pain behaviours. Key Results Voltage‐clamp recordings from lamina I spinal neurons of adult rats revealed that approximately 80% of neurons possess a low threshold T‐type current, which was blocked by Z944. Due to this highly prevalent T‐type current, Z944 potently blocked action‐potential evoked somatic and dendritic calcium transients in lamina I neurons. Moreover, application of Z944 to spinal cord slices attenuated action potential firing rates in over half of laminae I/II neurons. Finally, we found that intraperitoneal injection of Z944 (1–10 mg·kg−1) dose‐dependently reversed mechanical allodynia in the complete Freund's adjuvant model of persistent inflammatory pain, with a similar magnitude and time course of analgesic effects between male and female rats. Conclusion and Implications T‐type calcium channels critically shape the excitability of lamina I pain processing neurons and inhibition of these channels by the clinical stage antagonist Z944 potently reverses pain hypersensitivity across sexes.
Collapse
Affiliation(s)
- Erika K Harding
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Annemarie Dedek
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Robert P Bonin
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,University of Toronto Centre for the Study of Pain, University of Toronto, Toronto, Ontario, Canada
| | - Michael W Salter
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Terrance P Snutch
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael E Hildebrand
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
35
|
Dhanalakshmi C, Janakiraman U, Moutal A, Fukunaga K, Khanna R, Nelson MA. Evaluation of the effects of the T-type calcium channel enhancer SAK3 in a rat model of TAF1 deficiency. Neurobiol Dis 2021; 149:105224. [PMID: 33359140 PMCID: PMC8230513 DOI: 10.1016/j.nbd.2020.105224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/03/2020] [Accepted: 12/16/2020] [Indexed: 11/18/2022] Open
Abstract
The TATA-box binding protein associated factor 1 (TAF1) is part of the TFIID complex that plays a key role during the initiation of transcription. Variants of TAF1 are associated with neurodevelopmental disorders. Previously, we found that CRISPR/Cas9 based editing of the TAF1 gene disrupts the morphology of the cerebral cortex and blunts the expression as well as the function of the CaV3.1 (T-type) voltage gated calcium channel. Here, we tested the efficacy of SAK3 (ethyl 8'-methyl-2', 4-dioxo-2-(piperidin-1-yl)-2'H-spiro [cyclopentane-1, 3'-imidazo [1, 2-a] pyridine]-2-ene-3-carboxylate), a T-type calcium channel enhancer, in an animal model of TAF1 intellectual disability (ID) syndrome. At post-natal day 3, rat pups were subjected to intracerebroventricular (ICV) injection of either gRNA-control or gRNA-TAF1 CRISPR/Cas9 viruses. At post-natal day 21, the rat pups were given SAK3 (0.25 mg/kg, p.o.) or vehicle for 14 days (i.e. till post-natal day 35) and then subjected to behavioral, morphological, and molecular studies. Oral administration of SAK3 (0.25 mg/kg, p.o.) significantly rescued locomotion abnormalities associated with TAF1 gene editing. SAK3 treatment prevented the loss of cortical neurons and GFAP-positive astrocytes observed after TAF1 gene editing. In addition, SAK3 protected cells from apoptosis. SAK3 also restored the Brain-derived neurotrophic factor/protein kinase B/Glycogen Synthase Kinase 3 Beta (BDNF/AKT/GSK3β) signaling axis in TAF1 edited animals. Finally, SAK3 normalized the levels of three GSK3β substrates - CaV3.1, FOXP2, and CRMP2. We conclude that the T-type calcium channel enhancer SAK3 is beneficial against the deleterious effects of TAF1 gene-editing, in part, by stimulating the BDNF/AKT/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Chinnasamy Dhanalakshmi
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Udaiyappan Janakiraman
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, United States; The BIO5 Institute, University of Arizona, United States
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, United States; The BIO5 Institute, University of Arizona, United States
| | - Mark A Nelson
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA.
| |
Collapse
|
36
|
Arshaad MI, Siwek ME, Henseler C, Daubner J, Ehninger D, Hescheler J, Sachinidis A, Broich K, Papazoglou A, Weiergräber M. Enhanced hippocampal type II theta activity AND altered theta architecture in mice lacking the Ca v3.2 T-type voltage-gated calcium channel. Sci Rep 2021; 11:1099. [PMID: 33441788 PMCID: PMC7806756 DOI: 10.1038/s41598-020-79763-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/08/2020] [Indexed: 12/20/2022] Open
Abstract
T-type Ca2+ channels are assumed to contribute to hippocampal theta oscillations. We used implantable video-EEG radiotelemetry and qPCR to unravel the role of Cav3.2 Ca2+ channels in hippocampal theta genesis. Frequency analysis of spontaneous long-term recordings in controls and Cav3.2-/- mice revealed robust increase in relative power in the theta (4-8 Hz) and theta-alpha (4-12 Hz) ranges, which was most prominent during the inactive stages of the dark cycles. Urethane injection experiments also showed enhanced type II theta activity and altered theta architecture following Cav3.2 ablation. Next, gene candidates from hippocampal transcriptome analysis of control and Cav3.2-/- mice were evaluated using qPCR. Dynein light chain Tctex-Type 1 (Dynlt1b) was significantly reduced in Cav3.2-/- mice. Furthermore, a significant reduction of GABA A receptor δ subunits and GABA B1 receptor subunits was observed in the septohippocampal GABAergic system. Our results demonstrate that ablation of Cav3.2 significantly alters type II theta activity and theta architecture. Transcriptional changes in synaptic transporter proteins and GABA receptors might be functionally linked to the electrophysiological phenotype.
Collapse
Affiliation(s)
- Muhammad Imran Arshaad
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Magdalena Elisabeth Siwek
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Christina Henseler
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Johanna Daubner
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Dan Ehninger
- Molecular and Cellular Cognition, German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Sigmund-Freud-Str. 27, 53127, Bonn, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, Faculty of Medicine, Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Agapios Sachinidis
- Institute of Neurophysiology, University of Cologne, Faculty of Medicine, Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Anna Papazoglou
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Marco Weiergräber
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany.
| |
Collapse
|
37
|
Timic Stamenic T, Feseha S, Valdez R, Zhao W, Klawitter J, Todorovic SM. Alterations in Oscillatory Behavior of Central Medial Thalamic Neurons Demonstrate a Key Role of CaV3.1 Isoform of T-Channels During Isoflurane-Induced Anesthesia. Cereb Cortex 2020; 29:4679-4696. [PMID: 30715245 DOI: 10.1093/cercor/bhz002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 11/14/2022] Open
Abstract
Although the central medial nucleus (CeM) of the thalamus is an essential part of the arousal system for sleep and anesthesia initiation, the precise mechanisms that regulate its activity are not well studied. We examined the role of CaV3.1 isoform of T-type calcium channels (T-channels) in the excitability and rhythmic activity of CeM neurons during isoflurane (ISO)-induced anesthesia by using mouse genetics and selective pharmacology. Patch-clamp recordings taken from acute brain slices revealed that CaV3.1 channels in CeM are inhibited by prototypical volatile anesthetic ISO (250 and 500 μM) and selective T-channels blocker 3,5-dichloro-N-[1-(2,2-dimethyl-tetrahydro-pyran-4-ylmethyl)-4-fluoro-piperidin-4-ylmethyl]-benzamide (TTA-P2). Both TTA-P2 and ISO attenuated tonic and burst firing modes, and hyperpolarized CeM neurons from wild type (WT) mice. These effects were greatly diminished or abolished in CaV3.1 null mice. Our ensuing in vivo local field potential (LFP) recordings from CeM indicated that the ability of TTA-P2 and anesthetic concentrations of ISO to promote δ oscillation was substantially weakened in CaV3.1 null mice. Furthermore, escalating ISO concentrations induced stronger burst-suppression LFP pattern in mutant than in WT mice. Our results demonstrate for the first time the importance of CaV3.1 channels in thalamocortical oscillations from the non-specific thalamic nuclei that underlie clinically important effects of ISO.
Collapse
Affiliation(s)
- Tamara Timic Stamenic
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Simon Feseha
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Robert Valdez
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Wanzhu Zhao
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Jost Klawitter
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Slobodan M Todorovic
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,Neuroscience Graduate Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
38
|
Marks WN, Zabder NK, Snutch TP, Howland JG. T-type calcium channels regulate the acquisition and recall of conditioned fear in male, Wistar rats. Behav Brain Res 2020; 393:112747. [PMID: 32504730 DOI: 10.1016/j.bbr.2020.112747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/15/2020] [Accepted: 05/29/2020] [Indexed: 10/24/2022]
Abstract
The T-type calcium channel blocker, Z944, has been used as a pharmacological tool to assess T-type calcium channel function and examined for use as an anti-epileptic. As Z944 affects fear learning and memory in a rodent model of absence epilepsy, it is important to determine the effect of Z944 on learning and memory in a non-disease outbred rodent strain. This study examined the dose-dependent effects (5 mg/kg, 10 mg/kg, i.p.) of acute systemic treatment with Z944 on the learning and memory of fear conditioning and extinction in male Wistar rats. Z944 administered prior to the acquisition of fear conditioning significantly increased freezing prior to acquisition and extinction, during acquisition, and impaired recall of fear memory 24 h later. These findings suggest that T-type calcium channel activity may be required during associative learning for intact long-term memory. Enhanced fear behaviour observed prior to acquisition and extinction, and during acquisition could reflect an increase in anxiety, however, further testing is needed to determine the effect of Z944 on anxiety during fear conditioning and extinction. The use of Z944 for therapeutic purposes should consider the potential effects of Z944 on learning and memory in clinical populations.
Collapse
Affiliation(s)
- Wendie N Marks
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| | - Nadine K Zabder
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Terrance P Snutch
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - John G Howland
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| |
Collapse
|
39
|
T-Type Calcium Channels Contribute to Burst Firing in a Subpopulation of Medial Habenula Neurons. eNeuro 2020; 7:ENEURO.0201-20.2020. [PMID: 32719103 PMCID: PMC7433892 DOI: 10.1523/eneuro.0201-20.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 11/24/2022] Open
Abstract
Action potential (AP) burst firing caused by the activation of low-voltage-activated T-type Ca2+ channels is a unique mode of neuronal firing. T-type channels have been implicated in diverse physiological and pathophysiological processes, including epilepsy, autism, and mood regulation, but the brain structures involved remain incompletely understood. The medial habenula (MHb) is an epithalamic structure implicated in anxiety-like and withdrawal behavior. Previous studies have shown that MHb neurons fire tonic APs at a frequency of ∼2–10 Hz or display depolarized low-amplitude membrane oscillations. Here, we report in C57BL/6J mice that a subpopulation of MHb neurons are capable of firing transient, high-frequency AP bursts mediated by T-type channels. Burst firing was observed following rebounding from hyperpolarizing current injections or during depolarization from hyperpolarized membrane potentials in ∼20% of MHb neurons. It was rarely observed at baseline but could be evoked in MHb neurons displaying different initial activity states. Further, we show that T-type channel mRNA, in particular Cav3.1, is expressed in the MHb in both cholinergic and substance P-ergic neurons. Pharmacological Cav3 antagonism blocked both burst firing and evoked Ca2+ currents in MHb neurons. Additionally, we observed high-frequency AP doublet firing at sustained depolarized membrane potentials that was independent of T-type channels. Thus, there is a greater diversity of AP firing patterns in MHb neurons than previously identified, including T-type channel-mediated burst firing, which may uniquely contribute to behaviors with relevance to neuropsychiatric disease.
Collapse
|
40
|
Hashimoto K. Mechanisms for the resonant property in rodent neurons. Neurosci Res 2020; 156:5-13. [DOI: 10.1016/j.neures.2019.12.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 11/20/2019] [Accepted: 12/09/2019] [Indexed: 01/18/2023]
|
41
|
Hu H, Cui Y, Yang Y. Circuits and functions of the lateral habenula in health and in disease. Nat Rev Neurosci 2020; 21:277-295. [PMID: 32269316 DOI: 10.1038/s41583-020-0292-4] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2020] [Indexed: 12/14/2022]
Abstract
The past decade has witnessed exponentially growing interest in the lateral habenula (LHb) owing to new discoveries relating to its critical role in regulating negatively motivated behaviour and its implication in major depression. The LHb, sometimes referred to as the brain's 'antireward centre', receives inputs from diverse limbic forebrain and basal ganglia structures, and targets essentially all midbrain neuromodulatory systems, including the noradrenergic, serotonergic and dopaminergic systems. Its unique anatomical position enables the LHb to act as a hub that integrates value-based, sensory and experience-dependent information to regulate various motivational, cognitive and motor processes. Dysfunction of the LHb may contribute to the pathophysiology of several psychiatric disorders, especially major depression. Recently, exciting progress has been made in identifying the molecular and cellular mechanisms in the LHb that underlie negative emotional state in animal models of drug withdrawal and major depression. A future challenge is to translate these advances into effective clinical treatments.
Collapse
Affiliation(s)
- Hailan Hu
- Department of Psychiatry of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China. .,NHC and CAMS Key Laboratory of Medical Neurobiology, Mental Health Center, Zhejiang University, Hangzhou, China. .,Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, China. .,Fountain-Valley Institute for Life Sciences, Guangzhou, China.
| | - Yihui Cui
- The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| | - Yan Yang
- The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| |
Collapse
|
42
|
Ye YS, Li WY, Du SZ, Yang J, Nian Y, Xu G. Congenetic Hybrids Derived from Dearomatized Isoprenylated Acylphloroglucinol with Opposite Effects on Ca v3.1 Low Voltage-Gated Ca 2+ Channel. J Med Chem 2020; 63:1709-1716. [PMID: 31999455 DOI: 10.1021/acs.jmedchem.9b02056] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A hybrid of dearomatized isoprenylated acylphloroglucinol (DIAP) and monoterpenoid, hypatone A (1), together with its biosynthetic analogues 2-4 is characterized from Hypericum patulum. Structurally, 1 possesses an unprecedented spiro[bicyclo[3.2.1]octane-6,1'-cyclohexan]-2',4',6'-trione core as elucidated by extensive spectroscopic and X-ray crystallographic analyses. Biological studies reveal that compounds 1 and 2-4 produce opposite effects on Cav3.1 low voltage-gated Ca2+ channel, with 1 and 4, respectively, being the most potent Cav3.1 agonist and antagonist from natural products. Further studies suggest that compound 1 and its biogenetical precursor, 2, have the same binding site on Cav3.1 and that the rigid cagelike moiety at C-5 and C-6 is a key structural feature responsible for 1 being an agonist. Furthermore, 1 can normalize the pathological gating of a mutant Cav3.1 channel found in spinocerebellar ataxia 42 (SCA42), a hereditary neurodegenerative disorder with no available therapy. Collectively, our findings provide valuable tools for future studies on Cav3.1 physiology and pathophysiology, as well as afford possible leads for developing new drugs against SCA42, epilepsy, and pain.
Collapse
Affiliation(s)
- Yan-Song Ye
- State Key Laboratory of Phytochemistry and Plant Resources in West China and Yunnan Key Laboratory of Natural Medicinal Chemistry , Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650201 , People's Republic of China.,University of Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Wen-Yan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Ion Channel Research and Drug Development Center , Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming 650223 , People's Republic of China.,University of Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Shu-Zong Du
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Ion Channel Research and Drug Development Center , Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming 650223 , People's Republic of China.,University of Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Jian Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Ion Channel Research and Drug Development Center , Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming 650223 , People's Republic of China
| | - Yin Nian
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Ion Channel Research and Drug Development Center , Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming 650223 , People's Republic of China
| | - Gang Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China and Yunnan Key Laboratory of Natural Medicinal Chemistry , Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650201 , People's Republic of China
| |
Collapse
|
43
|
Scholarinine A, a N3 type caged-monoterpene indole alkaloid as Cav3.1 T-type calcium channel inhibitor from Alstonia scholaris. Tetrahedron Lett 2020. [DOI: 10.1016/j.tetlet.2019.151354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
44
|
Hyphenone A, the first 3,3-diisoprenylated bicyclic polyprenylated acylphloroglucinols as Cav3.1 T-type calcium channel inhibitor from Hypericum henryi. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.151220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
45
|
Wang H, Wei Y, Pu Y, Jiang D, Jiang X, Zhang Y, Tao J. Brain-derived neurotrophic factor stimulation of T-type Ca2+ channels in sensory neurons contributes to increased peripheral pain sensitivity. Sci Signal 2019; 12:12/600/eaaw2300. [DOI: 10.1126/scisignal.aaw2300] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although brain-derived neurotrophic factor (BDNF) is implicated in the nociceptive signaling of peripheral sensory neurons, the underlying mechanisms remain largely unknown. Here, we elucidated the effects of BDNF on the neuronal excitability of trigeminal ganglion (TG) neurons and the pain sensitivity of rats mediated by T-type Ca2+ channels. BDNF reversibly and dose-dependently enhanced T-type channel currents through the activation of tropomyosin receptor kinase B (TrkB). Antagonism of phosphatidylinositol 3-kinase (PI3K) but not of its downstream target, the kinase AKT, abolished the BDNF-induced T-type channel response. BDNF application activated p38 mitogen-activated protein kinase (MAPK), and this effect was prevented by inhibition of PI3K but not of protein kinase A (PKA). Antagonism of either PI3K or p38 MAPK prevented the BDNF-induced stimulation of PKA activity, whereas PKA inhibition blocked the BDNF-mediated increase in T-type currents. BDNF increased the rate of action potential firing in TG neurons and enhanced the pain sensitivity of rats to mechanical stimuli. Moreover, inhibition of TrkB signaling abolished the increased mechanical sensitivity in a rat model of chronic inflammatory pain, and this effect was attenuated by either T-type channel blockade or knockdown of the channel Cav3.2. Together, our findings indicate that BDNF enhances T-type currents through the stimulation of TrkB coupled to PI3K-p38-PKA signaling, thereby inducing neuronal hyperexcitability of TG neurons and pain hypersensitivity in rats.
Collapse
|
46
|
Sekiguchi F, Kawabata A. [Role of Ca v3.2 T-type Ca 2+ channels in prostate cancer cells]. Nihon Yakurigaku Zasshi 2019; 154:97-102. [PMID: 31527367 DOI: 10.1254/fpj.154.97] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Among voltage-gated Ca2+ channels, T-type Ca2+ channels, which are activated by low voltages, regulate neuronal excitability, spontaneous neurotransmitter release, hormone secretion, etc. and also participate in proliferation of distinct cancer cells. Among three isoforms of T-type Ca2+ channels, Cav3.2 is detectable in 100% of biopsy samples from prostate cancer patients. In general, prostate cancer cells are highly sensitive to androgen deprivation therapy, but often acquire hormone-therapy resistance. The androgen deprivation may trigger neuroendocrine (NE)-like differentiation of some prostate cancer cells. We have analyzed the expression and function of Cav3.2 in human prostate cancer LNCaP cells during NE-like differentiation. NE-like LNCaP cells overexpress Cav3.2 through the CREB/Egr-1 pathway and also cystathionine-γ-lyase (CSE), which generates H2S that enhances the channel activity of Cav3.2. H2S generated by upregulated CSE appears to enhance the activity of upregulated Cav3.2 after the differentiation. The enhanced Cav3.2 activity in NE-like cells may contribute to increased secretion of mitogenic factors essential for androgen-independent proliferation of surrounding prostate cancer cells. It is known that increased extracellular glucose levels enhance Cav3.2 activity through asparagine (N)-linked glycosylation of Cav3.2, which might contribute to diabetic neuropathy. We then found that high glucose accelerates the enhanced channel function and overexpression of Cav3.2 in NE-like LNCaP cells, which might be associated with clinical evidence for diabetes-related poor prognosis of prostate cancer and development of hormone therapy resistance. Thus, Cav3.2 is considered to play a role in the pathophysiology of prostate cancer, and may serve as a therapeutic target.
Collapse
Affiliation(s)
- Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University
| |
Collapse
|
47
|
Jin X, Chen Q, Song Y, Zheng J, Xiao K, Shao S, Fu Z, Yi M, Yang Y, Huang Z. Dopamine D2 receptors regulate the action potential threshold by modulating T‐type calcium channels in stellate cells of the medial entorhinal cortex. J Physiol 2019; 597:3363-3387. [PMID: 31049961 DOI: 10.1113/jp277976] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 04/24/2019] [Indexed: 11/08/2022] Open
Affiliation(s)
- Xueqin Jin
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science Centre Beijing 100191 China
| | - Qian Chen
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science Centre Beijing 100191 China
| | - Yan Song
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science Centre Beijing 100191 China
| | - Jie Zheng
- Neuroscience Research InstitutePeking University Health Science Center Beijing 100191 China
| | - Kuo Xiao
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science Centre Beijing 100191 China
| | - Shan Shao
- Neuroscience Research InstitutePeking University Health Science Center Beijing 100191 China
| | - Zibing Fu
- Neuroscience Research InstitutePeking University Health Science Center Beijing 100191 China
| | - Ming Yi
- Neuroscience Research InstitutePeking University Health Science Center Beijing 100191 China
- Key Laboratory for NeuroscienceMinistry of Education/National Health and Family Planning CommissionPeking University Beijing 100191 China
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular PharmacologyCollege of Pharmacy, Purdue University West Lafayette IN 47907 USA
- Purdue Institute for Integrative Neuroscience 575 Stadium Mall Drive West Lafayette IN 47907 USA
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science Centre Beijing 100191 China
- Key Laboratory for NeuroscienceMinistry of Education/National Health and Family Planning CommissionPeking University Beijing 100191 China
- Department of Molecular and Cellular PharmacologyPeking University Health Science Center 38 Xue Yuan Road Beijing 100191 China
| |
Collapse
|
48
|
Inhibition of T-Type calcium channels in mEC layer II stellate neurons reduces neuronal hyperexcitability associated with epilepsy. Epilepsy Res 2019; 154:132-138. [PMID: 31132598 DOI: 10.1016/j.eplepsyres.2019.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/01/2019] [Accepted: 05/07/2019] [Indexed: 01/20/2023]
Abstract
Temporal lobe epilepsy (TLE) is a form of adult epilepsy involving the entorhinal cortex (EC). Layer II neurons of the medial EC (mEC) are spared and become hyperexcitable in TLE. Studies have suggested a role for T-type calcium channels (T-type Ca2+ channels) in facilitating increases in neuronal activity associated with TLE within the hippocampus. We sought to determine if T-type Ca2+ channels play a role in facilitating neuronal hyperexcitability of layer II mEC stellate neurons in TLE. TLE was induced in rats by electrical stimulation of the hippocampus to induce status epilepticus (SE). Brain slices were prepared from rats exhibiting spontaneous seizures and compared with age-matched control rats. Action potentials (APs) were evoked either by current injection steps or via presynaptic stimulation of mEC deep layers. The selective T-type Ca2+ channel antagonist, TTA-P2 (1 μM), was applied to determine the role of T-type Ca2+ channels in maintaining neuronal excitability. Quantitative PCR techniques were used to assess T-type Ca2+ channel isoform mRNA levels within the mEC layer II. TLE mEC layer II stellate neurons were hyperexcitable compared to control neurons, evoking a higher frequency of APs and generating bursts of APs when synaptically stimulated. TTA-P2 (1 μM) reduced firing frequencies in TLE and control neurons and reduced AP burst firing in TLE stellate neurons. TTA-P2 had little effect on synaptically evoked AP's in control neurons. TTA-P2 also inhibited rebound APs evoked in TLE neurons to a greater degree than in control neurons. TLE tissue had almost a 3-fold increase in Cav3.1 mRNA compared to controls. Cav3.2 or Cav3.3 levels were unchanged. These findings support a role for T-type Ca2+ channel in establishing neuronal hyperexcitability of mEC layer II stellate neurons in TLE. Increased expression of Cav3.1 may be important for establishing neuronal hyperexcitability of mEC layer II neurons in TLE.
Collapse
|
49
|
Cui Y, Hu S, Hu H. Lateral Habenular Burst Firing as a Target of the Rapid Antidepressant Effects of Ketamine. Trends Neurosci 2019; 42:179-191. [PMID: 30823984 DOI: 10.1016/j.tins.2018.12.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/23/2018] [Accepted: 12/10/2018] [Indexed: 12/28/2022]
Abstract
The revolutionary discovery of the rapid antidepressant ketamine has been a milestone in psychiatry field in the last half century. Unlike conventional antidepressants that often take weeks to months to show efficacy, ketamine causes rapid antidepressant effects, emerging as early as within 1h after administration. However, how ketamine improves mood symptoms so quickly has remained elusive. Here, we first introduce the historical background of ketamine as a rapid antidepressant. We then discuss current hypotheses underlying ketamine's rapid antidepressant effects, with a focus on our latest discovery that ketamine silences NMDAR-dependent burst firing in the 'anti-reward center', the lateral habenula. While ketamine may act on many brain regions, we argue that its rapid antidepressant effects are critically dependent on ketamine's action in the lateral habenula, with this brain region acting as a primary site of action (or one among a few primary nodes). This molecular-, cellular-, and circuit-based mechanism advances our understanding of the etiology of depression and suggests a new conceptual framework for the rapid antidepressant effects of ketamine.
Collapse
Affiliation(s)
- Yihui Cui
- Center for Neuroscience and Department of Psychiatry of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Mental Health Center, Zhejiang University, Hangzhou 310058, China
| | - Shaohua Hu
- Center for Neuroscience and Department of Psychiatry of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Brain Research Institute of Zhejiang University, Hangzhou 310003, China
| | - Hailan Hu
- Center for Neuroscience and Department of Psychiatry of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; Interdisciplinary Institute of Neuroscience and Technology, Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Mental Health Center, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
50
|
Rebellato P, Kaczynska D, Kanatani S, Rayyes IA, Zhang S, Villaescusa C, Falk A, Arenas E, Hermanson O, Louhivuori L, Uhlén P. The T-type Ca 2+ Channel Ca v3.2 Regulates Differentiation of Neural Progenitor Cells during Cortical Development via Caspase-3. Neuroscience 2019; 402:78-89. [PMID: 30677486 DOI: 10.1016/j.neuroscience.2019.01.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/11/2018] [Accepted: 01/12/2019] [Indexed: 01/02/2023]
Abstract
Here we report that the low-voltage-dependent T-type calcium (Ca2+) channel Cav3.2, encoded by the CACNA1H gene, regulates neuronal differentiation during early embryonic brain development through activating caspase-3. At the onset of neuronal differentiation, neural progenitor cells exhibited spontaneous Ca2+ activity. This activity strongly correlated with the upregulation of CACNA1H mRNA. Cells exhibiting robust spontaneous Ca2+ signaling had increased caspase-3 activity unrelated to apoptosis. Inhibition of Cav3.2 by drugs or viral CACNA1H knock down resulted in decreased caspase-3 activity followed by suppressed neurogenesis. In contrast, when CACNA1H was overexpressed, increased neurogenesis was detected. Cortical slices from Cacna1h knockout mice showed decreased spontaneous Ca2+ activity, a significantly lower protein level of cleaved caspase-3, and microanatomical abnormalities in the subventricular/ventricular and cortical plate zones when compared to their respective embryonic controls. In summary, we demonstrate a novel relationship between Cav3.2 and caspase-3 signaling that affects neurogenesis in the developing brain.
Collapse
Affiliation(s)
- Paola Rebellato
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Dagmara Kaczynska
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Shigeaki Kanatani
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Ibrahim Al Rayyes
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Songbai Zhang
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Carlos Villaescusa
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Ernest Arenas
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Lauri Louhivuori
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| | - Per Uhlén
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|