1
|
Zhao S, Zhang JJ, Zhang MY, Yang QQ, Li ZX, Ren XH, Su SX, Si TE, Li JM, Wu HR, Chen SY, Zang WD, Cao J. TET1 participates in oxaliplatin-induced neuropathic pain by regulating microRNA-30b/Nav1.6. J Biol Chem 2025; 301:108228. [PMID: 39864621 PMCID: PMC11894311 DOI: 10.1016/j.jbc.2025.108228] [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: 05/14/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 01/28/2025] Open
Abstract
Chemotherapy-induced neuropathic pain poses significant clinical challenges and severely impacts patient quality of life. Sodium ion channels are crucial in regulating neuronal excitability and pain. Our research indicates that the microRNA-30b (miR-30b) in rat dorsal root ganglia (DRG) contributes to chemotherapy-induced neuropathic pain by regulating the Nav1.6 protein. Additionally, ten-eleven translocation methylcytosine dioxygenase 1 (TET1) plays a crucial role in pain generation by altering gene expression. We established a chemotherapy-induced neuropathy model using intraperitoneal oxaliplatin (OXA) injections and measured TET1 and Nav1.6 protein in the DRG. Using lentivirus and Tet1flox/flox mice, we modulated TET1 expression and assessed pain behaviors, DRG neuronal excitability, Nav1.6 currents, miR-30b-5p, and demethylation of the Mir30b promoter region. We employed chromatin immunoprecipitation to pinpoint TET1-binding sites on the Mir30b promoter. The impacts of miR-30b agomir or antagomir on Nav1.6 expression and pain responses were assessed postintrathecal injections. The results showed that OXA reduced TET1, increasing neuronal excitability, Nav1.6 currents, and miR-30b-5p in the DRG. TET1 knockdown exacerbated these effects and induced pain behaviors. Conversely, TET1 overexpression reversed these effects. TET1 also targeted and enhanced demethylation at the Mir30b promoter (-1103 bp to -1079 bp). miR-30b agomir reduces Nav1.6, whereas miR-30b antagomir reverses TET1's effects on Nav1.6 and pain. In OXA-induced neuropathy, decreased TET1 reduces miR-30b, elevating Nav1.6 expression and currents and contributing to pain. We hypothesize that TET1 mediates this process by regulating the demethylation of the Mir30b promoter.
Collapse
Affiliation(s)
- Sen Zhao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Institute of Neuroscience, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jing-Jing Zhang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; School of Nursing and Health, Zhengzhou University, Zhengzhou, China; Centre for Sport Nutrition and Health, Centre for Nutritional Ecology, School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou, China
| | - Meng-Ya Zhang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qing-Qing Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Institute of Neuroscience, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhi-Xiao Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiu-Hua Ren
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Song-Xue Su
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Tian-En Si
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jian-Min Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Institute of Neuroscience, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hui-Rui Wu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shi-Yue Chen
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wei-Dong Zang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Institute of Neuroscience, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
| | - Jing Cao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Institute of Neuroscience, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; School of Nursing and Health, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
2
|
Lin TL, So EC, Wu SN. Exploring the Effects of Tomatidine ((3β, 5α, 22β, and 25β)-Spirosolan-3-ol) on Voltage-gated Na+ currents: Insights Into Its Ionic Mechanisms of Action on Current Magnitude, Gating, and Frequency Dependence. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2024; 67:298-311. [PMID: 39641137 DOI: 10.4103/ejpi.ejpi-d-24-00076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024]
Abstract
ABSTRACT Tomatidine, a major tomato glycoalkaloid, is effective for the prevention of skeletal muscle wasting and enhancing mitophagy. However, its effects on transmembrane ionic currents are not well explored. In this study, we explored the interactions between tomatidine and Na+ current. GH3 or Neuro-2a cells were used for recording the ion currents employing modified patch-clamp technique under whole-cell configuration. Tomatidine increased both the peak, (transient Na+ current [INa (T)]) and sustained (late Na+ current [INa (L)]) components of voltage-gated Na+ current (INa) in a concentration-dependent manner, with the concentration required for 50% stimulation values of 43.3 μM and 3.1 μM, respectively. The steady-state current-voltage relationship of INa (T) remained unchanged; however, the steady-state inactivation curve of INa (T) in the presence of 3 μM tomatidine was shifted to less depolarized potential by around 6 mV. Tomatidine enhanced the window INa (window Na+ current [INa (W)]), which were attenuated by the ranolazine (Ran) and carbamazepine (CBZ). During a train of depolarizing pulses, tomatidine slowed the exponential decay of INa (T), and this effect was reversed by Ran or dapagliflozin. Tomatidine increased both fast and slow recovery time constants from INa (T) block, affecting the recovery time course. Tomatidine increased the amplitude of persistent Na+ current in response to a sinusoidal waveform. In neuro-2a cells, tomatidine increased INa (T) amplitude and slowed its inactivation, with this effect being attenuated by Ran or CBZ. In conclusion, tomatidine enhanced magnitude and modified its gating behaviors.
Collapse
Affiliation(s)
- Tso-Lin Lin
- Department of Paediatrics, An Nan Hospital, China Medical University, Tainan, Taiwan
| | - Edmund Cheung So
- Department of Anaesthesia, An Nan Hospital, China Medical University, Tainan, Taiwan
- Department of Cell Biology and Anatomy, National Cheng Kung University Medical College, Tainan, Taiwan
- Department of Anaesthesiology, University of Hong Kong, Hong Kong
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan, Taiwan
- Department of Medical Education and Research, An Nan Hospital, China Medical University, Tainan, Taiwan
| |
Collapse
|
3
|
Wu P, Tu Y, Cho H, Yu M, Wu Y, Wu S. An unidentified yet notable modification on I Na and I K (DR) caused by ramelteon. FASEB Bioadv 2024; 6:442-453. [PMID: 39372128 PMCID: PMC11452446 DOI: 10.1096/fba.2024-00008] [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: 01/12/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 10/08/2024] Open
Abstract
Despite advancement in anti-seizure medications, 30% of patients continue to experience recurrent seizures. Previous data indicated the antiepileptic properties of melatonin and its agonists in several animal models. However, the underlying mechanisms of melatonin and its agonists on cellular excitability remain poorly understood. In this study, we demonstrated the electrophysiological changes of two main kinds of ion channels that are responsible for hyperexcitability of neurons after introduction of melatonin agonists- ramelteon (RAM). In Neuro-2a cells, the amplitude of voltage-gated Na+ (I Na) and delayed-rectifier K+ currents (I K (DR)) could be suppressed under RAM. The IC50 values of 8.7 and 2.9 μM, respectively. RAM also diminished the magnitude of window Na+ current (I Na (W)) elicited by short ascending ramp voltage, with unchanged the overall steady-state current-voltage relationship. The decaying time course of I Na during a train of depolarizing pulses arose upon the exposure to RAM. The conditioning train protocol which blocked I Na fitted the recovery time course into two exponential processes and increased the fast and slow time constant of recovery the presence of RAM. In pituitary tumor (GH3) cells, I Na amplitude was also effectively suppressed by the RAM. In addition, GH3-cells exposure to RAM decreased the firing frequency of spontaneous action potentials observed under current-clamp conditions. As a result, the RAM-mediated effect on INa was closely associated with its ability to decrease spontaneous action potentials. Collectively, we found the direct attenuation of I Na and I K (DR) caused by RAM besides the agonistic action on melatonin receptors, which could partially explain its anti-seizure activity.
Collapse
Affiliation(s)
- Po‐Ming Wu
- Institute of Clinical Medicine, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- Department of Pediatrics, National Cheng Kung University Hospital, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Yi‐Fang Tu
- Institute of Clinical Medicine, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- Department of Pediatrics, National Cheng Kung University Hospital, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Hsin‐Yen Cho
- Department of PhysiologyNational Cheng Kung University Medical CollegeTainanTaiwan
| | - Meng‐Cheng Yu
- Department of PhysiologyNational Cheng Kung University Medical CollegeTainanTaiwan
| | - Yen‐Hsien Wu
- Department of PediatricsKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Sheng‐Nan Wu
- Department of PhysiologyNational Cheng Kung University Medical CollegeTainanTaiwan
- Institute of Basic Medical SciencesNational Cheng Kung University Medical CollegeTainanTaiwan
- Department of Research and Education, An Nan HospitalChina Medical UniversityTainanTaiwan
| |
Collapse
|
4
|
Goldfarb M. Fibroblast growth factor homologous factors: canonical and non-canonical mechanisms of action. J Physiol 2024; 602:4097-4110. [PMID: 39083261 DOI: 10.1113/jp286313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 09/01/2024] Open
Abstract
Since their discovery nearly 30 years ago, fibroblast growth factor homologous factors (FHFs) are now known to control the functionality of excitable tissues through a range of mechanisms. Nervous and cardiac system dysfunctions are caused by loss- or gain-of-function mutations in FHF genes. The best understood 'canonical' targets for FHF action are voltage-gated sodium channels, and recent studies have expanded the repertoire of ways that FHFs modulate sodium channel gating. Additional 'non-canonical' functions of FHFs in excitable and non-excitable cells, including cancer cells, have been reported over the past dozen years. This review summarizes and evaluates reported canonical and non-canonical FHF functions.
Collapse
Affiliation(s)
- Mitchell Goldfarb
- Department of Biological Sciences, Hunter College of City University, New York, New York, USA
- Biology Program, The Graduate Center City University, New York, New York, USA
| |
Collapse
|
5
|
Seeholzer LF, Julius D. Neuroendocrine cells initiate protective upper airway reflexes. Science 2024; 384:295-301. [PMID: 38669574 PMCID: PMC11407116 DOI: 10.1126/science.adh5483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 02/21/2024] [Indexed: 04/28/2024]
Abstract
Airway neuroendocrine (NE) cells have been proposed to serve as specialized sensory epithelial cells that modulate respiratory behavior by communicating with nearby nerve endings. However, their functional properties and physiological roles in the healthy lung, trachea, and larynx remain largely unknown. In this work, we show that murine NE cells in these compartments have distinct biophysical properties but share sensitivity to two commonly aspirated noxious stimuli, water and acid. Moreover, we found that tracheal and laryngeal NE cells protect the airways by releasing adenosine 5'-triphosphate (ATP) to activate purinoreceptive sensory neurons that initiate swallowing and expiratory reflexes. Our work uncovers the broad molecular and biophysical diversity of NE cells across the airways and reveals mechanisms by which these specialized excitable cells serve as sentinels for activating protective responses.
Collapse
Affiliation(s)
- Laura F. Seeholzer
- Department of Physiology, University of California, San Francisco; San Francisco, 94143, USA
| | - David Julius
- Department of Physiology, University of California, San Francisco; San Francisco, 94143, USA
| |
Collapse
|
6
|
Hung TY, Wu SN, Huang CW. Safinamide, an inhibitor of monoamine oxidase, modulates the magnitude, gating, and hysteresis of sodium ion current. BMC Pharmacol Toxicol 2024; 25:17. [PMID: 38331833 PMCID: PMC10851555 DOI: 10.1186/s40360-024-00739-5] [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: 09/09/2023] [Accepted: 01/30/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Safinamide (SAF), an α-aminoamide derivative and a selective, reversible monoamine oxidase (MAO)-B inhibitor, has both dopaminergic and nondopaminergic (glutamatergic) properties. Several studies have explored the potential of SAF against various neurological disorders; however, to what extent SAF modulates the magnitude, gating, and voltage-dependent hysteresis [Hys(V)] of ionic currents remains unknown. METHODS With the aid of patch-clamp technology, we investigated the effects of SAF on voltage-gated sodium ion (NaV) channels in pituitary GH3 cells. RESULTS SAF concentration-dependently stimulated the transient (peak) and late (sustained) components of voltage-gated sodium ion current (INa) in pituitary GH3 cells. The conductance-voltage relationship of transient INa [INa(T)] was shifted to more negative potentials with the SAF presence; however, the steady-state inactivation curve of INa(T) was shifted in a rightward direction in its existence. SAF increased the decaying time constant of INa(T) induced by a train of depolarizing stimuli. Notably, subsequent addition of ranolazine or mirogabalin reversed the SAF-induced increase in the decaying time constant. SAF also increased the magnitude of window INa induced by an ascending ramp voltage Vramp. Furthermore, SAF enhanced the Hys(V) behavior of persistent INa induced by an upright isosceles-triangular Vramp. Single-channel cell-attached recordings indicated SAF effectively increased the open-state probability of NaV channels. Molecular docking revealed SAF interacts with both MAO and NaV channels. CONCLUSION SAF may interact directly with NaV channels in pituitary neuroendocrine cells, modulating membrane excitability.
Collapse
Affiliation(s)
- Te-Yu Hung
- Department of Pediatrics, Chi-Mei Medical Center, Tainan, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan, Taiwan.
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan, Taiwan.
- School of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
7
|
Shiau AL, Lee KH, Cho HY, Chuang TH, Yu MC, Wu CL, Wu SN. Molnupiravir, a ribonucleoside antiviral prodrug against SARS-CoV-2, alters the voltage-gated sodium current and causes adverse events. Virology 2023; 587:109865. [PMID: 37572519 DOI: 10.1016/j.virol.2023.109865] [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: 03/09/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/14/2023]
Abstract
Molnupiravir (MOL) is a ribonucleoside prodrug for oral treatment of COVID-19. Common adverse effects of MOL are headache, diarrhea, and nausea, which may be associated with altered sodium channel function. Here, we investigated the effect of MOL on voltage-gated Na+ current (INa) in pituitary GH3 cells. We show that MOL had distinct effects on transient and late INa, in combination with decreased time constant in the slow component of INa inactivation. The 50% inhibitory concentration (IC50) values of MOL for suppressing transient and late INa were 26.1 and 6.3 μM, respectively. The overall steady-state current-voltage relationship of INa remained unchanged upon MOL exposure. MOL-induced alteration of INa may lead to changes in physiological function through sodium channels. Apart from its effect on inhibiting RNA virus replication, MOL exerts inhibitory effects on plasmalemma INa, which might constitute an additional yet crucial underlying mechanism of its pharmacological activity or adverse events.
Collapse
Affiliation(s)
- Ai-Li Shiau
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, 60002, Taiwan; Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Kuan-Hsien Lee
- Department of Emergency Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, 60002, Taiwan
| | - Hsin-Yen Cho
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Tzu-Hsien Chuang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Meng-Cheng Yu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Chao-Liang Wu
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, 60002, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| | - Sheng-Nan Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan; School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan.
| |
Collapse
|
8
|
Hung TY, Wu SN, Huang CW. Concerted suppressive effects of carisbamate, an anti-epileptic alkyl-carbamate drug, on voltage-gated Na + and hyperpolarization-activated cation currents. Front Cell Neurosci 2023; 17:1159067. [PMID: 37293624 PMCID: PMC10244622 DOI: 10.3389/fncel.2023.1159067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/02/2023] [Indexed: 06/10/2023] Open
Abstract
Carisbamate (CRS, RWJ-333369) is a new anti-seizure medication. It remains unclear whether and how CRS can perturb the magnitude and/or gating kinetics of membrane ionic currents, despite a few reports demonstrating its ability to suppress voltage-gated Na+ currents. In this study, we observed a set of whole-cell current recordings and found that CRS effectively suppressed the voltage-gated Na+ (INa) and hyperpolarization-activated cation currents (Ih) intrinsically in electrically excitable cells (GH3 cells). The effective IC50 values of CRS for the differential suppression of transient (INa(T)) and late INa (INa(L)) were 56.4 and 11.4 μM, respectively. However, CRS strongly decreased the strength (i.e., Δarea) of the nonlinear window component of INa (INa(W)), which was activated by a short ascending ramp voltage (Vramp); the subsequent addition of deltamethrin (DLT, 10 μM) counteracted the ability of CRS (100 μM, continuous exposure) to suppress INa(W). CRS strikingly decreased the decay time constant of INa(T) evoked during pulse train stimulation; however, the addition of telmisartan (10 μM) effectively attenuated the CRS (30 μM, continuous exposure)-mediated decrease in the decay time constant of the current. During continued exposure to deltamethrin (10 μM), known to be a pyrethroid insecticide, the addition of CRS resulted in differential suppression of the amplitudes of INa(T) and INa(L). The amplitude of Ih activated by a 2-s membrane hyperpolarization was diminished by CRS in a concentration-dependent manner, with an IC50 value of 38 μM. For Ih, CRS altered the steady-state I-V relationship and attenuated the strength of voltage-dependent hysteresis (Hys(V)) activated by an inverted isosceles-triangular Vramp. Moreover, the addition of oxaliplatin effectively reversed the CRS-mediated suppression of Hys(V). The predicted docking interaction between CRS and with a model of the hyperpolarization-activated cyclic nucleotide-gated (HCN) channel or between CRS and the hNaV1.7 channel reflects the ability of CRS to bind to amino acid residues in HCN or hNaV1.7 channel via hydrogen bonds and hydrophobic interactions. These findings reveal the propensity of CRS to modify INa(T) and INa(L) differentially and to effectively suppress the magnitude of Ih. INa and Ih are thus potential targets of the actions of CRS in terms of modulating cellular excitability.
Collapse
Affiliation(s)
- Te-Yu Hung
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- College of Medicine, Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
9
|
Zang Y, Marder E, Marom S. Sodium channel slow inactivation normalizes firing in axons with uneven conductance distributions. Curr Biol 2023; 33:1818-1824.e3. [PMID: 37023754 PMCID: PMC10175232 DOI: 10.1016/j.cub.2023.03.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/17/2023] [Accepted: 03/15/2023] [Indexed: 04/08/2023]
Abstract
The Na+ channels that are important for action potentials show rapid inactivation, a state in which they do not conduct, although the membrane potential remains depolarized.1,2 Rapid inactivation is a determinant of millisecond-scale phenomena, such as spike shape and refractory period. Na+ channels also inactivate orders of magnitude more slowly, and this slow inactivation has impacts on excitability over much longer timescales than those of a single spike or a single inter-spike interval.3,4,5,6,7,8,9,10 Here, we focus on the contribution of slow inactivation to the resilience of axonal excitability11,12 when ion channels are unevenly distributed along the axon. We study models in which the voltage-gated Na+ and K+ channels are unevenly distributed along axons with different variances, capturing the heterogeneity that biological axons display.13,14 In the absence of slow inactivation, many conductance distributions result in spontaneous tonic activity. Faithful axonal propagation is achieved with the introduction of Na+ channel slow inactivation. This "normalization" effect depends on relations between the kinetics of slow inactivation and the firing frequency. Consequently, neurons with characteristically different firing frequencies will need to implement different sets of channel properties to achieve resilience. The results of this study demonstrate the importance of the intrinsic biophysical properties of ion channels in normalizing axonal function.
Collapse
Affiliation(s)
- Yunliang Zang
- Volen Center and Biology Department, Brandeis University, Waltham, MA 02454, USA.
| | - Eve Marder
- Volen Center and Biology Department, Brandeis University, Waltham, MA 02454, USA
| | - Shimon Marom
- Technion - Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
10
|
Strege PR, Cowan LM, Alcaino C, Mazzone A, Ahern CA, Milescu LS, Farrugia G, Beyder A. Mechanosensitive pore opening of a prokaryotic voltage-gated sodium channel. eLife 2023; 12:e79271. [PMID: 36912788 PMCID: PMC10038658 DOI: 10.7554/elife.79271] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
Voltage-gated ion channels (VGICs) orchestrate electrical activities that drive mechanical functions in contractile tissues such as the heart and gut. In turn, contractions change membrane tension and impact ion channels. VGICs are mechanosensitive, but the mechanisms of mechanosensitivity remain poorly understood. Here, we leverage the relative simplicity of NaChBac, a prokaryotic voltage-gated sodium channel from Bacillus halodurans, to investigate mechanosensitivity. In whole-cell experiments on heterologously transfected HEK293 cells, shear stress reversibly altered the kinetic properties of NaChBac and increased its maximum current, comparably to the mechanosensitive eukaryotic sodium channel NaV1.5. In single-channel experiments, patch suction reversibly increased the open probability of a NaChBac mutant with inactivation removed. A simple kinetic mechanism featuring a mechanosensitive pore opening transition explained the overall response to force, whereas an alternative model with mechanosensitive voltage sensor activation diverged from the data. Structural analysis of NaChBac identified a large displacement of the hinged intracellular gate, and mutagenesis near the hinge diminished NaChBac mechanosensitivity, further supporting the proposed mechanism. Our results suggest that NaChBac is overall mechanosensitive due to the mechanosensitivity of a voltage-insensitive gating step associated with the pore opening. This mechanism may apply to eukaryotic VGICs, including NaV1.5.
Collapse
Affiliation(s)
- Peter R Strege
- Enteric Neuroscience Program (ENSP), Division of Gastroenterology & Hepatology, Department of Medicine, Mayo ClinicRochesterUnited States
| | - Luke M Cowan
- Enteric Neuroscience Program (ENSP), Division of Gastroenterology & Hepatology, Department of Medicine, Mayo ClinicRochesterUnited States
| | - Constanza Alcaino
- Enteric Neuroscience Program (ENSP), Division of Gastroenterology & Hepatology, Department of Medicine, Mayo ClinicRochesterUnited States
| | - Amelia Mazzone
- Enteric Neuroscience Program (ENSP), Division of Gastroenterology & Hepatology, Department of Medicine, Mayo ClinicRochesterUnited States
| | - Christopher A Ahern
- Department of Molecular Physiology and Biophysics, University of IowaIowa CityUnited States
| | - Lorin S Milescu
- Department of Biology, University of Maryland, College ParkCollege ParkUnited States
| | - Gianrico Farrugia
- Enteric Neuroscience Program (ENSP), Division of Gastroenterology & Hepatology, Department of Medicine, Mayo ClinicRochesterUnited States
- Department of Physiology and Biomedical Engineering, Mayo ClinicRochesterUnited States
| | - Arthur Beyder
- Enteric Neuroscience Program (ENSP), Division of Gastroenterology & Hepatology, Department of Medicine, Mayo ClinicRochesterUnited States
- Department of Physiology and Biomedical Engineering, Mayo ClinicRochesterUnited States
| |
Collapse
|
11
|
Pellizzari S, Hu M, Amaral-Silva L, Saunders SE, Santin JM. Neuron populations use variable combinations of short-term feedback mechanisms to stabilize firing rate. PLoS Biol 2023; 21:e3001971. [PMID: 36689462 PMCID: PMC9894548 DOI: 10.1371/journal.pbio.3001971] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/02/2023] [Accepted: 12/19/2022] [Indexed: 01/24/2023] Open
Abstract
Neurons tightly regulate firing rate and a failure to do so leads to multiple neurological disorders. Therefore, a fundamental question in neuroscience is how neurons produce reliable activity patterns for decades to generate behavior. Neurons have built-in feedback mechanisms that allow them to monitor their output and rapidly stabilize firing rate. Most work emphasizes the role of a dominant feedback system within a neuronal population for the control of moment-to-moment firing. In contrast, we find that respiratory motoneurons use 2 activity-dependent controllers in unique combinations across cells, dynamic activation of an Na+ pump subtype, and rapid potentiation of Kv7 channels. Both systems constrain firing rate by reducing excitability for up to a minute after a burst of action potentials but are recruited by different cellular signals associated with activity, increased intracellular Na+ (the Na+ pump), and membrane depolarization (Kv7 channels). Individual neurons do not simply contain equal amounts of each system. Rather, neurons under strong control of the Na+ pump are weakly regulated by Kv7 enhancement and vice versa along a continuum. Thus, each motoneuron maintains its characteristic firing rate through a unique combination of the Na+ pump and Kv7 channels, which are dynamically regulated by distinct feedback signals. These results reveal a new organizing strategy for stable circuit output involving multiple fast activity sensors scaled inversely across a neuronal population.
Collapse
Affiliation(s)
- Sarah Pellizzari
- University of North Carolina at Greensboro, Greensboro, North Carolina, United States of America
| | - Min Hu
- University of North Carolina at Greensboro, Greensboro, North Carolina, United States of America
| | - Lara Amaral-Silva
- University of Missouri, Columbia, Missouri, United States of America
| | - Sandy E. Saunders
- University of Missouri, Columbia, Missouri, United States of America
| | - Joseph M. Santin
- University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
12
|
Lin MH, Lin JF, Yu MC, Wu SN, Wu CL, Cho HY. Characterization in Potent Modulation on Voltage-Gated Na + Current Exerted by Deltamethrin, a Pyrethroid Insecticide. Int J Mol Sci 2022; 23:14733. [PMID: 36499059 PMCID: PMC9737322 DOI: 10.3390/ijms232314733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Deltamethrin (DLT) is a type-II pyrethroid ester insecticide used in agricultural and domestic applications as well as in public health. However, transmembrane ionic channels perturbed by this compound remain largely unclear, although the agent is thought to alter the gating characteristics of voltage-gated Na+ (NaV) channel current. In this study, we reappraised whether and how it and other related compounds can make any further modifications on voltage-gated Na+ current (INa) in pituitary tumor (GH3) cells. Cell exposure to DLT produced a differential and dose-dependent stimulation of peak (transient, INa(T)) or sustained (late, INa(L)) INa; consequently, the EC50 value required for DLT-stimulated INa(T) or INa(L) was determined to be 11.2 or 2.5 μM, respectively. However, neither the fast nor slow component in the inactivation time constant of INa(T) activated by short depolarizing pulse was changed with the DLT presence; conversely, tefluthrin (Tef), a type-I pyrethroid insecticide, can accentuate INa with a slowing in inactivation time course of the current. The INa(L) augmented by DLT was attenuated by further application of either dapagliflozin (Dapa) or amiloride, but not by chlorotoxin. During pulse train (PT) stimulation, with the Tef or DLT presence, the cumulative inhibition of INa(T) became slowed; moreover, following PT stimuli, a large tail current with a slowly recovering process was observed. Alternatively, during rapid depolarizing pulse, the amplitude of INa(L) and tail INa (INa(Tail)) for each depolarizing pulse became progressively increased by adding DLT, not by Tef. The recovery time constant following PT stimulation with continued presence of Tef or DLT was shortened by further addition of Dapa. The voltage-dependent hysteresis (Hys(V)) of persistent INa was differentially augmented by Tef or DLT. Taken together, the magnitude, gating, frequency dependence, as well as Hys(V) behavior of INa exerted by the presence of DLT or Tef might exert a synergistic impact on varying functional activities of excitable cells in culture or in vivo.
Collapse
Affiliation(s)
- Mao-Hsun Lin
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 600, Taiwan
| | - Jen-Feng Lin
- Department of Emergency Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 600, Taiwan
| | - Meng-Cheng Yu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 701, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 701, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 701, Taiwan
- Department of Post-Baccalaureate Medicine, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Chao-Liang Wu
- Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 600, Taiwan
| | - Hsin-Yen Cho
- Department of Physiology, National Cheng Kung University Medical College, Tainan 701, Taiwan
| |
Collapse
|
13
|
Wu PM, Lin YC, Chiang CW, Cho HY, Chuang TH, Yu MC, Wu SN, Tu YF. Effective Modulation by Lacosamide on Cumulative Inhibition of INa during High-Frequency Stimulation and Recovery of INa Block during Conditioning Pulse Train. Int J Mol Sci 2022; 23:11966. [PMID: 36233266 PMCID: PMC9570249 DOI: 10.3390/ijms231911966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 12/03/2022] Open
Abstract
The effects of lacosamide (LCS, Vimpat®), an anti-convulsant and analgesic, on voltage-gated Na+ current (INa) were investigated. LCS suppressed both the peak (transient, INa(T)) and sustained (late, INa(L)) components of INa with the IC50 values of 78 and 34 μM found in GH3 cells and of 112 and 26 μM in Neuro-2a cells, respectively. In GH3 cells, the voltage-dependent hysteresis of persistent INa (INa(P)) during the triangular ramp pulse was strikingly attenuated, and the decaying time constant (τ) of INa(T) or INa(L) during a train of depolarizing pulses was further shortened by LCS. The recovery time course from the INa block elicited by the preceding conditioning train can be fitted by two exponential processes, while the single exponential increase in current recovery without a conditioning train was adequately fitted. The fast and slow τ's of recovery from the INa block by the same conditioning protocol arose in the presence of LCS. In Neuro-2a cells, the strength of the instantaneous window INa (INa(W)) during the rapid ramp pulse was reduced by LCS. This reduction could be reversed by tefluthrin. Moreover, LCS accelerated the inactivation time course of INa activated by pulse train stimulation, and veratridine reversed its decrease in the decaying τ value in current inactivation. The docking results predicted the capability of LCS binding to some amino-acid residues in sodium channels owing to the occurrence of hydrophobic contact. Overall, our findings unveiled that LCS can interact with the sodium channels to alter the magnitude, gating, voltage-dependent hysteresis behavior, and use dependence of INa in excitable cells.
Collapse
Affiliation(s)
- Po-Ming Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yu-Ching Lin
- Department of Physical Medicine and Rehabilitation, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Chi-Wu Chiang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Hsin-Yen Cho
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Tzu-Hsien Chuang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Meng-Cheng Yu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Fang Tu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
14
|
Shiau AL, Liao CS, Tu CW, Wu SN, Cho HY, Yu MC. Characterization in Effective Stimulation on the Magnitude, Gating, Frequency Dependence, and Hysteresis of INa Exerted by Picaridin (or Icaridin), a Known Insect Repellent. Int J Mol Sci 2022; 23:9696. [PMID: 36077093 PMCID: PMC9456182 DOI: 10.3390/ijms23179696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Picaridin (icaridin), a member of the piperidine chemical family, is a broad-spectrum arthropod repellent. Its actions have been largely thought to be due to its interaction with odorant receptor proteins. However, to our knowledge, to what extent the presence of picaridin can modify the magnitude, gating, and/or the strength of voltage-dependent hysteresis (Hys(V)) of plasmalemmal ionic currents, such as, voltage-gated Na+ current [INa], has not been entirely explored. In GH3 pituitary tumor cells, we demonstrated that with exposure to picaridin the transient (INa(T)) and late (INa(L)) components of voltage-gated Na+ current (INa) were differentially stimulated with effective EC50's of 32.7 and 2.8 μM, respectively. Upon cell exposure to it, the steady-state current versus voltage relationship INa(T) was shifted to more hyperpolarized potentials. Moreover, its presence caused a rightward shift in the midpoint for the steady-state inactivate curve of the current. The cumulative inhibition of INa(T) induced during repetitive stimuli became retarded during its exposure. The recovery time course from the INa block elicited, following the conditioning pulse stimulation, was satisfactorily fitted by two exponential processes. Moreover, the fast and slow time constants of recovery from the INa block by the same conditioning protocol were noticeably increased in the presence of picaridin. However, the fraction in fast or slow component of recovery time course was, respectively, increased or decreased with an increase in picaridin concentrations. The Hys(V)'s strength of persistent INa (INa(P)), responding to triangular ramp voltage, was also enhanced during cell exposure to picaridin. The magnitude of resurgent INa (INa(R)) was raised in its presence. Picaritin-induced increases of INa(P) or INa(R) intrinsically in GH3 cells could be attenuated by further addition of ranolazine. The predictions of molecular docking also disclosed that there are possible interactions of the picaridin molecule with the hNaV1.7 channel. Taken literally, the stimulation of INa exerted by the exposure to picaridin is expected to exert impacts on the functional activities residing in electrically excitable cells.
Collapse
Affiliation(s)
- Ai-Li Shiau
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Chih-Szu Liao
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Chi-Wen Tu
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| | - Hsin-Yen Cho
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| | - Meng-Cheng Yu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| |
Collapse
|
15
|
Wu SN, Wu CL, Cho HY, Chiang CW. Effective Perturbations by Small-Molecule Modulators on Voltage-Dependent Hysteresis of Transmembrane Ionic Currents. Int J Mol Sci 2022; 23:9453. [PMID: 36012718 PMCID: PMC9408818 DOI: 10.3390/ijms23169453] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
The non-linear voltage-dependent hysteresis (Hys(V)) of voltage-gated ionic currents can be robustly activated by the isosceles-triangular ramp voltage (Vramp) through digital-to-analog conversion. Perturbations on this Hys(V) behavior play a role in regulating membrane excitability in different excitable cells. A variety of small molecules may influence the strength of Hys(V) in different types of ionic currents elicited by long-lasting triangular Vramp. Pirfenidone, an anti-fibrotic drug, decreased the magnitude of Ih's Hys(V) activated by triangular Vramp, while dexmedetomidine, an agonist of α2-adrenoceptors, effectively suppressed Ih as well as diminished the Hys(V) strength of Ih. Oxaliplatin, a platinum-based anti-neoplastic drug, was noted to enhance the Ih's Hys(V) strength, which is thought to be linked to the occurrence of neuropathic pain, while honokiol, a hydroxylated biphenyl compound, decreased Ih's Hys(V). Cell exposure to lutein, a xanthophyll carotenoid, resulted in a reduction of Ih's Hys(V) magnitude. Moreover, with cell exposure to UCL-2077, SM-102, isoplumbagin, or plumbagin, the Hys(V) strength of erg-mediated K+ current activated by triangular Vramp was effectively diminished, whereas the presence of either remdesivir or QO-58 respectively decreased or increased Hys(V) magnitude of M-type K+ current. Zingerone, a methoxyphenol, was found to attenuate Hys(V) (with low- and high-threshold loops) of L-type Ca2+ current induced by long-lasting triangular Vramp. The Hys(V) properties of persistent Na+ current (INa(P)) evoked by triangular Vramp were characterized by a figure-of-eight (i.e., ∞) configuration with two distinct loops (i.e., low- and high-threshold loops). The presence of either tefluthrin, a pyrethroid insecticide, or t-butyl hydroperoxide, an oxidant, enhanced the Hys(V) strength of INa(P). However, further addition of dapagliflozin can reverse their augmenting effects in the Hys(V) magnitude of the current. Furthermore, the addition of esaxerenone, mirogabalin, or dapagliflozin was effective in inhibiting the strength of INa(P). Taken together, the observed perturbations by these small-molecule modulators on Hys(V) strength in different types of ionic currents evoked during triangular Vramp are expected to influence the functional activities (e.g., electrical behaviors) of different excitable cells in vitro or in vivo.
Collapse
Affiliation(s)
- Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 70101, Taiwan
- Department of Post-Baccalaureate Medicine, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Chao-Liang Wu
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Hsin-Yen Cho
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| | - Chi-Wu Chiang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
16
|
Wu PM, Lai PC, Cho HY, Chuang TH, Wu SN, Tu YF. Effective Perturbations by Phenobarbital on INa, IK(erg), IK(M) and IK(DR) during Pulse Train Stimulation in Neuroblastoma Neuro-2a Cells. Biomedicines 2022; 10:1968. [PMID: 36009515 PMCID: PMC9405590 DOI: 10.3390/biomedicines10081968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Phenobarbital (PHB, Luminal Sodium®) is a medication of the barbiturate and has long been recognized to be an anticonvulsant and a hypnotic because it can facilitate synaptic inhibition in the central nervous system through acting on the γ-aminobutyric acid (GABA) type A (GABAA) receptors. However, to what extent PHB could directly perturb the magnitude and gating of different plasmalemmal ionic currents is not thoroughly explored. In neuroblastoma Neuro-2a cells, we found that PHB effectively suppressed the magnitude of voltage-gated Na+ current (INa) in a concentration-dependent fashion, with an effective IC50 value of 83 µM. The cumulative inhibition of INa, evoked by pulse train stimulation, was enhanced by PHB. However, tefluthrin, an activator of INa, could attenuate PHB-induced reduction in the decaying time constant of INa inhibition evoked by pulse train stimuli. In addition, the erg (ether-à-go-go-related gene)-mediated K+ current (IK(erg)) was also blocked by PHB. The PHB-mediated inhibition on IK(erg) could not be overcome by flumazenil (GABA antagonist) or chlorotoxin (chloride channel blocker). The PHB reduced the recovery of IK(erg) by a two-step voltage protocol with a geometrics-based progression, but it increased the decaying rate of IK(erg), evoked by the envelope-of-tail method. About the M-type K+ currents (IK(M)), PHB caused a reduction of its amplitude, which could not be counteracted by flumazenil or chlorotoxin, and PHB could enhance its cumulative inhibition during pulse train stimulation. Moreover, the magnitude of delayed-rectifier K+ current (IK(DR)) was inhibited by PHB, while the cumulative inhibition of IK(DR) during 10 s of repetitive stimulation was enhanced. Multiple ionic currents during pulse train stimulation were subject to PHB, and neither GABA antagonist nor chloride channel blocker could counteract these PHB-induced reductions. It suggests that these actions might conceivably participate in different functional activities of excitable cells and be independent of GABAA receptors.
Collapse
Affiliation(s)
- Po-Ming Wu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Pei-Chun Lai
- Education Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Hsin-Yen Cho
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Tzu-Hsien Chuang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Fang Tu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
17
|
Wu PM, Cho HY, Chiang CW, Chuang TH, Wu SN, Tu YF. Characterization in Inhibitory Effectiveness of Carbamazepine in Voltage-Gated Na + and Erg-Mediated K + Currents in a Mouse Neural Crest-Derived (Neuro-2a) Cell Line. Int J Mol Sci 2022; 23:7892. [PMID: 35887240 PMCID: PMC9321339 DOI: 10.3390/ijms23147892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/15/2022] [Accepted: 07/15/2022] [Indexed: 12/18/2022] Open
Abstract
Carbamazepine (CBZ, Tegretol®) is an anticonvulsant used in the treatment of epilepsy and neuropathic pain; however, several unwanted effects of this drug have been noticed. Therefore, the regulatory actions of CBZ on ionic currents in electrically excitable cells need to be reappraised, although its efficacy in suppressing voltage-gated Na+ current (INa) has been disclosed. This study was undertaken to explore the modifications produced by CBZ on ionic currents (e.g., INa and erg-mediated K+ current [IK(erg)]) measured from Neuro-2a (N2a) cells. In these cells, we found that this drug differentially suppressed the peak (transient, INa(T)) and sustained (late, INa(L)) components of INa in a concentration-dependent manner with effective IC50 of 56 and 18 μM, respectively. The overall current-voltage relationship of INa(T) with or without the addition of CBZ remained unchanged; however, the strength (i.e., ∆area) in the window component of INa (INa(W)) evoked by the short ascending ramp pulse (Vramp) was overly lessened in the CBZ presence. Tefluthrin (Tef), a synthetic pyrethroid, known to stimulate INa, augmented the strength of the voltage-dependent hysteresis (Hys(V)) of persistent INa (INa(P)) in response to the isosceles-triangular Vramp; moreover, further application of CBZ attenuated Tef-mediated accentuation of INa(P)'s Hys(V). With a two-step voltage protocol, the recovery of INa(T) inactivation seen in Neuro-2a cells became progressively slowed by adding CBZ; however, the cumulative inhibition of INa(T) evoked by pulse train stimulation was enhanced during exposure to this drug. Neuro-2a-cell exposure to CBZ (100 μM), the magnitude of erg-mediated K+ current measured throughout the entire voltage-clamp steps applied was mildly inhibited. The docking results regarding the interaction of CBZ and voltage-gate Na+ (NaV) channel predicted the ability of CBZ to bind to some amino-acid residues in NaV due to the existence of a hydrogen bond or hydrophobic contact. It is conceivable from the current investigations that the INa (INa(T), INa(L), INa(W), and INa(P)) residing in Neuro-2a cells are susceptible to being suppressed by CBZ, and that its block on INa(L) is larger than that on INa(T). Collectively, the magnitude and gating of NaV channels produced by the CBZ presence might have an impact on its anticonvulsant and analgesic effects occurring in vivo.
Collapse
Affiliation(s)
- Po-Ming Wu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Hsin-Yen Cho
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
| | - Chi-Wu Chiang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Tzu-Hsien Chuang
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| | - Yi-Fang Tu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
18
|
Upchurch CM, Combe CL, Knowlton CJ, Rousseau VG, Gasparini S, Canavier CC. Long-Term Inactivation of Sodium Channels as a Mechanism of Adaptation in CA1 Pyramidal Neurons. J Neurosci 2022; 42:3768-3782. [PMID: 35332085 PMCID: PMC9087813 DOI: 10.1523/jneurosci.1914-21.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/28/2022] [Accepted: 03/09/2022] [Indexed: 11/21/2022] Open
Abstract
Many hippocampal CA1 pyramidal cells function as place cells, increasing their firing rate when a specific place field is traversed. The dependence of CA1 place cell firing on position within the place field is asymmetric. We investigated the source of this asymmetry by injecting triangular depolarizing current ramps to approximate the spatially tuned, temporally diffuse depolarizing synaptic input received by these neurons while traversing a place field. Ramps were applied to CA1 pyramidal neurons from male rats in vitro (slice electrophysiology) and in silico (multicompartmental NEURON model). Under control conditions, CA1 neurons fired more action potentials at higher frequencies on the up-ramp versus the down-ramp. This effect was more pronounced for dendritic compared with somatic ramps. We incorporated a four-state Markov scheme for NaV1.6 channels into our model and calibrated the spatial dependence of long-term inactivation according to the literature; this spatial dependence was sufficient to explain the difference in dendritic versus somatic ramps. Long-term inactivation reduced the firing frequency by decreasing open-state occupancy, and reduced spike amplitude during trains by decreasing occupancy in the closed state, which comprises the available pool. PKC activator phorbol-dibutyrate, known to reduce NaV long-term inactivation, removed spike amplitude attenuation in vitro more visibly in dendrites and greatly reduced adaptation, consistent with our hypothesized mechanism. Intracellular application of a peptide inducing long-term NaV inactivation elicited spike amplitude attenuation during spike trains in the soma and greatly enhanced adaptation. Our synergistic experimental/computational approach shows that long-term inactivation of NaV1.6 is a key mechanism of adaptation in CA1 pyramidal cells.SIGNIFICANCE STATEMENT The hippocampus plays an important role in certain types of memory, in part through context-specific firing of "place cells"; these cells were first identified in rodents as being particularly active when an animal is in a specific location in an environment, called the place field of that neuron. In this in vitro/in silico study, we found that long-term inactivation of sodium channels causes adaptation in the firing rate that could potentially skew the firing of CA1 hippocampal pyramidal neurons earlier within a place field. A computational model of the sodium channel revealed differential regulation of spike frequency and amplitude by long-term inactivation, which may be a general mechanism for spike frequency adaptation in the CNS.
Collapse
Affiliation(s)
- Carol M Upchurch
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Crescent L Combe
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Christopher J Knowlton
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Valery G Rousseau
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Sonia Gasparini
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Carmen C Canavier
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| |
Collapse
|
19
|
Wu CL, Chuang CW, Cho HY, Chuang TH, Wu SN. The Evidence for Effective Inhibition of INa Produced by Mirogabalin ((1R,5S,6S)-6-(aminomethyl)-3-ethyl-bicyclo [3.2.0] hept-3-ene-6-acetic acid), a Known Blocker of Ca V Channels. Int J Mol Sci 2022; 23:3845. [PMID: 35409204 PMCID: PMC8998350 DOI: 10.3390/ijms23073845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 01/27/2023] Open
Abstract
Mirogabalin (MGB, Tarlige®), an inhibitor of the α2δ-1 subunit of voltage-gated Ca2+ (CaV) channels, is used as a way to alleviate peripheral neuropathic pain and diabetic neuropathy. However, to what extent MGB modifies the magnitude, gating, and/or hysteresis of various types of plasmalemmal ionic currents remains largely unexplored. In pituitary tumor (GH3) cells, we found that MGB was effective at suppressing the peak (transient, INa(T)) and sustained (late, INa(L)) components of the voltage-gated Na+ current (INa) in a concentration-dependent manner, with an effective IC50 of 19.5 and 7.3 μM, respectively, while the KD value calculated on the basis of minimum reaction scheme was 8.2 μM. The recovery of INa(T) inactivation slowed in the presence of MGB, although the overall current-voltage relation of INa(T) was unaltered; however, there was a leftward shift in the inactivation curve of the current. The magnitude of the window (INa(W)) or resurgent INa (INa(R)) evoked by the respective ascending or descending ramp pulse (Vramp) was reduced during cell exposure to MGB. MGB-induced attenuation in INa(W) or INa(R) was reversed by the further addition of tefluthrin, a pyrethroid insecticide known to stimulate INa. MGB also effectively lessened the strength of voltage-dependent hysteresis of persistent INa in response to the isosceles triangular Vramp. The cumulative inhibition of INa(T), evoked by pulse train stimulation, was enhanced in its presence. Taken together, in addition to the inhibition of CaV channels, the NaV channel attenuation produced by MGB might have an impact in its analgesic effects occurring in vivo.
Collapse
Affiliation(s)
- Chao-Liang Wu
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan;
| | - Chao-Wei Chuang
- Department of Ophthalmology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan;
| | - Hsin-Yen Cho
- Department of Physiology, National Cheng Kung University Medical College, Tainan City 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
| | - Tzu-Hsien Chuang
- Department of Physiology, National Cheng Kung University Medical College, Tainan City 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan City 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan City 70101, Taiwan
| |
Collapse
|
20
|
Membrane electrical properties of mouse hippocampal CA1 pyramidal neurons during strong inputs. Biophys J 2022; 121:644-657. [PMID: 34999132 PMCID: PMC8873947 DOI: 10.1016/j.bpj.2022.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/21/2021] [Accepted: 01/05/2022] [Indexed: 11/24/2022] Open
Abstract
In this work, we highlight an electrophysiological feature often observed in recordings from mouse CA1 pyramidal cells that has so far been ignored by experimentalists and modelers. It consists of a large and dynamic increase in the depolarization baseline (i.e., the minimum value of the membrane potential between successive action potentials during a sustained input) in response to strong somatic current injections. Such an increase can directly affect neurotransmitter release properties and, more generally, the efficacy of synaptic transmission. However, it cannot be explained by any currently available conductance-based computational model. Here we present a model addressing this issue, demonstrating that experimental recordings can be reproduced by assuming that an input current modifies, in a time-dependent manner, the electrical and permeability properties of the neuron membrane by shifting the ionic reversal potentials and channel kinetics. For this reason, we propose that any detailed model of ion channel kinetics for neurons exhibiting this characteristic should be adapted to correctly represent the response and the synaptic integration process during strong and sustained inputs.
Collapse
|
21
|
Ransdell JL, Moreno JD, Bhagavan D, Silva JR, Nerbonne JM. Intrinsic mechanisms in the gating of resurgent Na + currents. eLife 2022; 11:e70173. [PMID: 35076394 PMCID: PMC8824471 DOI: 10.7554/elife.70173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
The resurgent component of the voltage-gated sodium current (INaR) is a depolarizing conductance, revealed on membrane hyperpolarizations following brief depolarizing voltage steps, which has been shown to contribute to regulating the firing properties of numerous neuronal cell types throughout the central and peripheral nervous systems. Although mediated by the same voltage-gated sodium (Nav) channels that underlie the transient and persistent Nav current components, the gating mechanisms that contribute to the generation of INaR remain unclear. Here, we characterized Nav currents in mouse cerebellar Purkinje neurons, and used tailored voltage-clamp protocols to define how the voltage and the duration of the initial membrane depolarization affect the amplitudes and kinetics of INaR. Using the acquired voltage-clamp data, we developed a novel Markov kinetic state model with parallel (fast and slow) inactivation pathways and, we show that this model reproduces the properties of the resurgent, as well as the transient and persistent, Nav currents recorded in (mouse) cerebellar Purkinje neurons. Based on the acquired experimental data and the simulations, we propose that resurgent Na+ influx occurs as a result of fast inactivating Nav channels transitioning into an open/conducting state on membrane hyperpolarization, and that the decay of INaR reflects the slow accumulation of recovered/opened Nav channels into a second, alternative and more slowly populated, inactivated state. Additional simulations reveal that extrinsic factors that affect the kinetics of fast or slow Nav channel inactivation and/or impact the relative distribution of Nav channels in the fast- and slow-inactivated states, such as the accessory Navβ4 channel subunit, can modulate the amplitude of INaR.
Collapse
Affiliation(s)
- Joseph L Ransdell
- Departments of Medicine, Cardiovascular Division, Washington UniversitySt. LouisUnited States
| | - Jonathan D Moreno
- Developmental Biomedical Engineering, Washington UniversitySt. LouisUnited States
| | - Druv Bhagavan
- Developmental Biomedical Engineering, Washington UniversitySt. LouisUnited States
| | - Jonathan R Silva
- Developmental Biomedical Engineering, Washington UniversitySt. LouisUnited States
| | - Jeanne M Nerbonne
- Departments of Medicine, Cardiovascular Division, Washington UniversitySt. LouisUnited States
- Developmental Biology, Washington UniversitySt. LouisUnited States
| |
Collapse
|
22
|
Valle-Bautista R, Márquez-Valadez B, Herrera-López G, Griego E, Galván EJ, Díaz NF, Arias-Montaño JA, Molina-Hernández A. Long-Term Functional and Cytoarchitectonic Effects of the Systemic Administration of the Histamine H1 Receptor Antagonist/Inverse Agonist Chlorpheniramine During Gestation in the Rat Offspring Primary Motor Cortex. Front Neurosci 2022; 15:740282. [PMID: 35140581 PMCID: PMC8820484 DOI: 10.3389/fnins.2021.740282] [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: 07/12/2021] [Accepted: 12/20/2021] [Indexed: 11/26/2022] Open
Abstract
The transient histaminergic system is among the first neurotransmitter systems to appear during brain development in the rat mesencephalon/rhombencephalon. Histamine increases FOXP2-positive deep-layer neuron differentiation of cortical neural stem cells through H1 receptor activation in vitro. The in utero or systemic administration of chlorpheniramine (H1 receptor antagonist/inverse agonist) during deep-layer cortical neurogenesis decreases FOXP2 neurons in the developing cortex, and H1R- or histidine decarboxylase-knockout mice show impairment in learning and memory, wakefulness and nociception, functions modulated by the cerebral cortex. Due to the role of H1R in cortical neural stem cell neurogenesis, the purpose of this study was to evaluate the postnatal impact of the systemic administration of chlorpheniramine during deep-layer cortical neuron differentiation (E12–14) in the primary motor cortex (M1) of neonates (P0) and 21-day-old pups (P21). Chlorpheniramine or vehicle were systemically administered (5 mg/kg, i.p.) to pregnant Wistar rats at gestational days 12–14, and the expression and distribution of deep- (FOXP2 and TBR1) and superficial-layer (SATB2) neuronal cortical markers were analyzed in neonates from both groups. The qRT-PCR analysis revealed a reduction in the expression of Satb2 and FoxP2. However, Western blot and immunofluorescence showed increased protein levels in the chlorpheniramine-treated group. In P21 pups, the three markers showed impaired distribution and increased immunofluorescence in the experimental group. The Sholl analysis evidenced altered dendritic arborization of deep-layer neurons, with lower excitability in response to histamine, as evaluated by whole-cell patch-clamp recording, as well as diminished depolarization-evoked [3H]-glutamate release from striatal slices. Overall, these results suggest long-lasting effects of blocking H1Rs during early neurogenesis that may impact the pathways involved in voluntary motor activity and cognition.
Collapse
Affiliation(s)
- Rocío Valle-Bautista
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
- Laboratorio de Investigación en Células Troncales y Biología del Desarrollo, Departamento de Fisiología y Desarrollo Celular, Subdirección de Investigación Biomédica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | - Berenice Márquez-Valadez
- Laboratorio de Investigación en Células Troncales y Biología del Desarrollo, Departamento de Fisiología y Desarrollo Celular, Subdirección de Investigación Biomédica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | - Gabriel Herrera-López
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Ernesto Griego
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Emilio J. Galván
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Néstor-Fabián Díaz
- Laboratorio de Investigación en Células Troncales y Biología del Desarrollo, Departamento de Fisiología y Desarrollo Celular, Subdirección de Investigación Biomédica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | - José-Antonio Arias-Montaño
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Anayansi Molina-Hernández
- Laboratorio de Investigación en Células Troncales y Biología del Desarrollo, Departamento de Fisiología y Desarrollo Celular, Subdirección de Investigación Biomédica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
- *Correspondence: Anayansi Molina-Hernández, ; orcid.org/0000-0002-4787-312X
| |
Collapse
|
23
|
Page SC, Sripathy SR, Farinelli F, Ye Z, Wang Y, Hiler DJ, Pattie EA, Nguyen CV, Tippani M, Moses RL, Chen HY, Tran MN, Eagles NJ, Stolz JM, Catallini JL, Soudry OR, Dickinson D, Berman KF, Apud JA, Weinberger DR, Martinowich K, Jaffe AE, Straub RE, Maher BJ. Electrophysiological measures from human iPSC-derived neurons are associated with schizophrenia clinical status and predict individual cognitive performance. Proc Natl Acad Sci U S A 2022; 119:e2109395119. [PMID: 35017298 PMCID: PMC8784142 DOI: 10.1073/pnas.2109395119] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 11/10/2021] [Indexed: 12/11/2022] Open
Abstract
Neurons derived from human induced pluripotent stem cells (hiPSCs) have been used to model basic cellular aspects of neuropsychiatric disorders, but the relationship between the emergent phenotypes and the clinical characteristics of donor individuals has been unclear. We analyzed RNA expression and indices of cellular function in hiPSC-derived neural progenitors and cortical neurons generated from 13 individuals with high polygenic risk scores (PRSs) for schizophrenia (SCZ) and a clinical diagnosis of SCZ, along with 15 neurotypical individuals with low PRS. We identified electrophysiological measures in the patient-derived neurons that implicated altered Na+ channel function, action potential interspike interval, and gamma-aminobutyric acid-ergic neurotransmission. Importantly, electrophysiological measures predicted cardinal clinical and cognitive features found in these SCZ patients. The identification of basic neuronal physiological properties related to core clinical characteristics of illness is a potentially critical step in generating leads for novel therapeutics.
Collapse
Affiliation(s)
| | | | | | - Zengyou Ye
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | - Yanhong Wang
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | - Daniel J Hiler
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | | | | | | | | | - Huei-Ying Chen
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | - Matthew Nguyen Tran
- Lieber Institute for Brain Development, Baltimore, MD 21205
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | | | - Joshua M Stolz
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | - Joseph L Catallini
- Lieber Institute for Brain Development, Baltimore, MD 21205
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | | | - Dwight Dickinson
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health Intramural Research Program, NIH, Bethesda, MD 20892
| | - Karen F Berman
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health Intramural Research Program, NIH, Bethesda, MD 20892
| | - Jose A Apud
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health Intramural Research Program, NIH, Bethesda, MD 20892
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Baltimore, MD 21205
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Keri Martinowich
- Lieber Institute for Brain Development, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Baltimore, MD 21205
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | | | - Brady J Maher
- Lieber Institute for Brain Development, Baltimore, MD 21205;
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205
| |
Collapse
|
24
|
Knowlton CJ, Ziouziou TI, Hammer N, Roeper J, Canavier CC. Inactivation mode of sodium channels defines the different maximal firing rates of conventional versus atypical midbrain dopamine neurons. PLoS Comput Biol 2021; 17:e1009371. [PMID: 34534209 PMCID: PMC8480832 DOI: 10.1371/journal.pcbi.1009371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/29/2021] [Accepted: 08/23/2021] [Indexed: 12/21/2022] Open
Abstract
Two subpopulations of midbrain dopamine (DA) neurons are known to have different dynamic firing ranges in vitro that correspond to distinct projection targets: the originally identified conventional DA neurons project to the dorsal striatum and the lateral shell of the nucleus accumbens, whereas an atypical DA population with higher maximum firing frequencies projects to prefrontal regions and other limbic regions including the medial shell of nucleus accumbens. Using a computational model, we show that previously identified differences in biophysical properties do not fully account for the larger dynamic range of the atypical population and predict that the major difference is that originally identified conventional cells have larger occupancy of voltage-gated sodium channels in a long-term inactivated state that recovers slowly; stronger sodium and potassium conductances during action potential firing are also predicted for the conventional compared to the atypical DA population. These differences in sodium channel gating imply that longer intervals between spikes are required in the conventional population for full recovery from long-term inactivation induced by the preceding spike, hence the lower maximum frequency. These same differences can also change the bifurcation structure to account for distinct modes of entry into depolarization block: abrupt versus gradual. The model predicted that in cells that have entered depolarization block, it is much more likely that an additional depolarization can evoke an action potential in conventional DA population. New experiments comparing lateral to medial shell projecting neurons confirmed this model prediction, with implications for differential synaptic integration in the two populations. We developed a theoretical and mathematical framework that could explain the major electrophysiological differences between the conventional midbrain dopamine (DA) neurons with a low maximum firing rate, and the more recently identified atypical DA neurons. Testable predictions from this framework were then verified with in vitro patch-clamp recordings from DA neurons with identified phenotypes and projection targets. Since different subpopulations of DA neurons participate in different circuits, and these circuits are likely differentially dysregulated in diseases such as addiction, Parkinson disease, and schizophrenia, it is important to identify the differences of their intrinsic electrophysiological properties as a prelude to developing more precisely targeted therapies.
Collapse
Affiliation(s)
- Christopher J. Knowlton
- Department of Cell Biology and Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | | | - Niklas Hammer
- Institut für Neurophysiologie, Goethe University, Frankfurt, Germany
| | - Jochen Roeper
- Institut für Neurophysiologie, Goethe University, Frankfurt, Germany
| | - Carmen C. Canavier
- Department of Cell Biology and Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
25
|
Carbone E. Fast inactivation of Nav1.3 channels by FGF14 proteins: An unconventional way to regulate the slow firing of adrenal chromaffin cells. J Gen Physiol 2021; 153:211934. [PMID: 33792614 PMCID: PMC8020463 DOI: 10.1085/jgp.202112879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Using Nav1.3 and FGF14 KO mice, Martinez-Espinosa et al. provide new findings on how intracellular FGF14 proteins interfere with the endogenous fast inactivation gating and regulate the “long-term inactivation” of Nav1.3 channels that sets Nav channel availability and spike adaptation during sustained stimulation in adrenal chromaffin cells.
Collapse
Affiliation(s)
- Emilio Carbone
- Department of Drug Science, Lab of Cell Physiology and Molecular Neuroscience, University of Torino, Torino, Italy
| |
Collapse
|
26
|
Martin E, Ernoult M, Laydevant J, Li S, Querlioz D, Petrisor T, Grollier J. EqSpike: spike-driven equilibrium propagation for neuromorphic implementations. iScience 2021; 24:102222. [PMID: 33748709 PMCID: PMC7970361 DOI: 10.1016/j.isci.2021.102222] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/15/2021] [Accepted: 02/18/2021] [Indexed: 11/06/2022] Open
Abstract
Finding spike-based learning algorithms that can be implemented within the local constraints of neuromorphic systems, while achieving high accuracy, remains a formidable challenge. Equilibrium propagation is a promising alternative to backpropagation as it only involves local computations, but hardware-oriented studies have so far focused on rate-based networks. In this work, we develop a spiking neural network algorithm called EqSpike, compatible with neuromorphic systems, which learns by equilibrium propagation. Through simulations, we obtain a test recognition accuracy of 97.6% on the MNIST handwritten digits dataset (Mixed National Institute of Standards and Technology), similar to rate-based equilibrium propagation, and comparing favorably to alternative learning techniques for spiking neural networks. We show that EqSpike implemented in silicon neuromorphic technology could reduce the energy consumption of inference and training, respectively, by three orders and two orders of magnitude compared to graphics processing units. Finally, we also show that during learning, EqSpike weight updates exhibit a form of spike-timing-dependent plasticity, highlighting a possible connection with biology.
Collapse
Affiliation(s)
- Erwann Martin
- Thales Research and Technology, 91767 Palaiseau, France
| | - Maxence Ernoult
- Unité Mixte de Physique, CNRS, Thales, Université Paris-Saclay, 91767 Palaiseau, France
- Université Paris-Saclay, CNRS, Centre de Nanosciences et de Nanotechnologies, 91120 Palaiseau, France
| | - Jérémie Laydevant
- Unité Mixte de Physique, CNRS, Thales, Université Paris-Saclay, 91767 Palaiseau, France
| | - Shuai Li
- Unité Mixte de Physique, CNRS, Thales, Université Paris-Saclay, 91767 Palaiseau, France
| | - Damien Querlioz
- Université Paris-Saclay, CNRS, Centre de Nanosciences et de Nanotechnologies, 91120 Palaiseau, France
| | | | - Julie Grollier
- Unité Mixte de Physique, CNRS, Thales, Université Paris-Saclay, 91767 Palaiseau, France
| |
Collapse
|
27
|
Martinez-Espinosa PL, Yang C, Xia XM, Lingle CJ. Nav1.3 and FGF14 are primary determinants of the TTX-sensitive sodium current in mouse adrenal chromaffin cells. J Gen Physiol 2021; 153:211839. [PMID: 33651884 PMCID: PMC8020717 DOI: 10.1085/jgp.202012785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/07/2021] [Accepted: 01/19/2021] [Indexed: 12/29/2022] Open
Abstract
Adrenal chromaffin cells (CCs) in rodents express rapidly inactivating, tetrodotoxin (TTX)-sensitive sodium channels. The resulting current has generally been attributed to Nav1.7, although a possible role for Nav1.3 has also been suggested. Nav channels in rat CCs rapidly inactivate via two independent pathways which differ in their time course of recovery. One subpopulation recovers with time constants similar to traditional fast inactivation and the other ∼10-fold slower, but both pathways can act within a single homogenous population of channels. Here, we use Nav1.3 KO mice to probe the properties and molecular components of Nav current in CCs. We find that the absence of Nav1.3 abolishes all Nav current in about half of CCs examined, while a small, fast inactivating Nav current is still observed in the rest. To probe possible molecular components underlying slow recovery from inactivation, we used mice null for fibroblast growth factor homology factor 14 (FGF14). In these cells, the slow component of recovery from fast inactivation is completely absent in most CCs, with no change in the time constant of fast recovery. The use dependence of Nav current reduction during trains of stimuli in WT cells is completely abolished in FGF14 KO mice, directly demonstrating a role for slow recovery from inactivation in determining Nav current availability. Our results indicate that FGF14-mediated inactivation is the major determinant defining use-dependent changes in Nav availability in CCs. These results establish that Nav1.3, like other Nav isoforms, can also partner with FGF subunits, strongly regulating Nav channel function.
Collapse
Affiliation(s)
| | - Chengtao Yang
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| | - Xiao-Ming Xia
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
28
|
Martinez-Espinosa PL, Neely A, Ding J, Lingle CJ. Fast inactivation of Nav current in rat adrenal chromaffin cells involves two independent inactivation pathways. J Gen Physiol 2021; 153:211834. [PMID: 33647101 PMCID: PMC7927663 DOI: 10.1085/jgp.202012784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/07/2021] [Accepted: 01/19/2021] [Indexed: 12/13/2022] Open
Abstract
Voltage-dependent sodium (Nav) current in adrenal chromaffin cells (CCs) is rapidly inactivating and tetrodotoxin (TTX)–sensitive. The fractional availability of CC Nav current has been implicated in regulation of action potential (AP) frequency and the occurrence of slow-wave burst firing. Here, through recordings of Nav current in rat CCs, primarily in adrenal medullary slices, we describe unique inactivation properties of CC Nav inactivation that help define AP firing rates in CCs. The key feature of CC Nav current is that recovery from inactivation, even following brief (5 ms) inactivation steps, exhibits two exponential components of similar amplitude. Various paired pulse protocols show that entry into the fast and slower recovery processes result from largely independent competing inactivation pathways, each of which occurs with similar onset times at depolarizing potentials. Over voltages from −120 to −80 mV, faster recovery varies from ∼3 to 30 ms, while slower recovery varies from ∼50 to 400 ms. With strong depolarization (above −10 mV), the relative entry into slow or fast recovery pathways is similar and independent of voltage. Trains of short depolarizations favor recovery from fast recovery pathways and result in cumulative increases in the slow recovery fraction. Dual-pathway fast inactivation, by promoting use-dependent accumulation in slow recovery pathways, dynamically regulates Nav availability. Consistent with this finding, repetitive AP clamp waveforms at 1–10 Hz frequencies reduce Nav availability 80–90%, depending on holding potential. These results indicate that there are two distinct pathways of fast inactivation, one leading to conventional fast recovery and the other to slower recovery, which together are well-suited to mediate use-dependent changes in Nav availability.
Collapse
Affiliation(s)
| | - Alan Neely
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| | - Jiuping Ding
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
29
|
Xiao J, Chen Z, Yu B. A Potential Mechanism of Sodium Channel Mediating the General Anesthesia Induced by Propofol. Front Cell Neurosci 2020; 14:593050. [PMID: 33343303 PMCID: PMC7746837 DOI: 10.3389/fncel.2020.593050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
General anesthesia has revolutionized healthcare over the past 200 years and continues to show advancements. However, many phenomena induced by general anesthetics including paradoxical excitation are still poorly understood. Voltage-gated sodium channels (NaV) were believed to be one of the proteins targeted during general anesthesia. Based on electrophysiological measurements before and after propofol treatments of different concentrations, we mathematically modified the Hodgkin–Huxley sodium channel formulations and constructed a thalamocortical model to investigate the potential roles of NaV. The ion channels of individual neurons were modeled using the Hodgkin–Huxley type equations. The enhancement of propofol-induced GABAa current was simulated by increasing the maximal conductance and the time-constant of decay. Electroencephalogram (EEG) was evaluated as the post-synaptic potential from pyramidal (PY) cells. We found that a left shift in activation of NaV was induced primarily by a low concentration of propofol (0.3–10 μM), while a left shift in inactivation of NaV was induced by an increasing concentration (0.3–30 μM). Mathematical simulation indicated that a left shift of NaV activation produced a Hopf bifurcation, leading to cell oscillations. Left shift of NaV activation around a value of 5.5 mV in the thalamocortical models suppressed normal bursting of thalamocortical (TC) cells by triggering its chaotic oscillations. This led to irregular spiking of PY cells and an increased frequency in EEG readings. This observation suggests a mechanism leading to paradoxical excitation during general anesthesia. While a left shift in inactivation led to light hyperpolarization in individual cells, it inhibited the activity of the thalamocortical model after a certain depth of anesthesia. This finding implies that high doses of propofol inhibit the network partly by accelerating NaV toward inactivation. Additionally, this result explains why the application of sodium channel blockers decreases the requirement for general anesthetics. Our study provides an insight into the roles that NaV plays in the mechanism of general anesthesia. Since the activation and inactivation of NaV are structurally independent, it should be possible to avoid side effects by state-dependent binding to the NaV to achieve precision medicine in the future.
Collapse
Affiliation(s)
- Jinglei Xiao
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengguo Chen
- College of Computer, National University of Defence Technology, Changsha, China
| | - Buwei Yu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|