1
|
Li X, Zheng Q, Yu H, Du T, Hu T, Gao L, Jia L, Sun Q. BMAL1 rescued the hippocampus-dependent recognition memory induced by sleep deprivation. Neuroscience 2025; 569:1-11. [PMID: 39904474 DOI: 10.1016/j.neuroscience.2025.01.067] [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: 10/03/2024] [Revised: 12/15/2024] [Accepted: 01/31/2025] [Indexed: 02/06/2025]
Abstract
Sleep plays an important role in the process of memory. This study investigated the role of the circadian clock gene, BMAL1 of the master circadian clock in mediating the impairment of hippocampus-dependent recognition memory caused by sleep deprivation. After 4 weeks of sleep deprivation, the novel object recognitiontask was used to evaluate the recognition memory of mice, the expression levels of circadian clock genes, and Nrf2 and PKA/CREB/BDNF signal pathways were detected by Western blot, Realtime-qPCR, and immunofluorescence. The mice in the SD group exhibited a significant decrease in the duration of exploration of novel objects. The protein expression levels of PER1, PER2, CLOCK, and BMAL1, and PKA/CREB/BDNF pathway in the hippocampus of the SD group were significantly reduced, and the Nrf2-mediated anti-oxidative capacity was also compromised in the SD group. Moreover, these aberrations could be mitigated through compensation with BMAL1 in the SCN of the hypothalamus. Sleep deprivation resulted in a reduction in the expression of the core clock gene BMAL1 in the hippocampus, leading to an imbalance in the antioxidant system and damaging down-regulating the PKA/CREB/BDNF signal pathway that related to the proteins associated with recognition memory in the hippocampal synapse plasticity and oxidative stress, which could be reversed by overexpression compensation of BMAL1 in the SCN that might rely on the multi-synaptic neural projections to the hippocampus.
Collapse
Affiliation(s)
- Xiao Li
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, 110004, People's Republic of China.
| | - Qian Zheng
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, 110013, People's Republic of China
| | - Honghong Yu
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, 110013, People's Republic of China
| | - Tingting Du
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, 110013, People's Republic of China
| | - Tian Hu
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, 110013, People's Republic of China
| | - Lanyue Gao
- Experimental Center, China Medical University, Shenyang, Liaoning Province, 110013, People's Republic of China.
| | - Lihong Jia
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, 110013, People's Republic of China.
| | - Qi Sun
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, 110013, People's Republic of China.
| |
Collapse
|
2
|
Agarwal S, Kim ED, Lee S, Simon A, Accardi A, Nimigean CM. Ball-and-chain inactivation of a human large conductance calcium-activated potassium channel. Nat Commun 2025; 16:1769. [PMID: 39971906 PMCID: PMC11840039 DOI: 10.1038/s41467-025-56844-4] [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: 09/23/2024] [Accepted: 02/03/2025] [Indexed: 02/21/2025] Open
Abstract
BK channels are large-conductance calcium (Ca2+)-activated potassium channels crucial for neuronal excitability, muscle contraction, and neurotransmitter release. The pore-forming (α) subunits co-assemble with auxiliary (β and γ) subunits that modulate their function. Previous studies demonstrated that the N-termini of β2-subunits can inactivate BK channels, but with no structural correlate. Here, we investigate BK β2-subunit inactivation using cryo-electron microscopy, electrophysiology and molecular dynamics simulations. We find that the β2 N-terminus occludes the pore only in the Ca2+-bound open state, via a ball-and-chain mechanism. The first three hydrophobic residues of β2 are crucial for occlusion, while the remainder of the N-terminus remains flexible. Neither the closed channel conformation obtained in the absence of Ca2+ nor an intermediate conformation found in the presence of Ca2+ show density for the N-terminus of the β2 subunit in their pore, likely due to narrower side access portals preventing their entry into the channel pore.
Collapse
Affiliation(s)
- Shubhangi Agarwal
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Elizabeth D Kim
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Sangyun Lee
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Alexander Simon
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Alessio Accardi
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Crina M Nimigean
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA.
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA.
| |
Collapse
|
3
|
Du T, Liu S, Yu H, Hu T, Huang L, Gao L, Jia L, Hu J, Yu Y, Sun Q. Chronic sleep deprivation disturbs energy balance modulated by suprachiasmatic nucleus efferents in mice. BMC Biol 2024; 22:296. [PMID: 39710657 DOI: 10.1186/s12915-024-02097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/16/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Epidemiologic researches show that short sleep duration may affect feeding behaviors resulting in higher energy intake and increased risk of obesity, but the further mechanisms that can interpret the causality remain unclear. The circadian rhythm is fine-tuned by the suprachiasmatic nucleus (SCN) as the master clock, which is essential for driving rhythms in food intake and energy metabolism through neuronal projections to the arcuate nucleus (ARC) and paraventricular nucleus (PVN). RESULTS We showed that chronic SD-induced aberrant expressions of AgRP/NPY and POMC attributed to compromised JAK/STAT3 signals and reduced energy expenditure in the mice, which can be rescued with AAV-genetic overexpression of BMAL1 into SCN. The potential mechanism may be related to the disruptions of SCN efferent mediated by BMAL1. CONCLUSIONS Chronic SD impairs energy balance through directly dampening BMAL1 expression, probably in the transcription level, in the SCN, which in turn affects the neuron projections to ARC and PVN. Remarkably, we provide evidence that may explain the causal mechanisms associated with sleep curtailment and obesity in adolescents.
Collapse
Affiliation(s)
- Tingting Du
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Shuailing Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Honghong Yu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Tian Hu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Lina Huang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Lanyue Gao
- Experimental Center, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Lihong Jia
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Jiajin Hu
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yang Yu
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Qi Sun
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
4
|
Steponenaite A, Lalic T, Atkinson L, Tanday N, Brown L, Mathie A, Cader ZM, Lall GS. TASK-3, two-pore potassium channels, contribute to circadian rhythms in the electrical properties of the suprachiasmatic nucleus and play a role in driving stable behavioural photic entrainment. Chronobiol Int 2024; 41:802-816. [PMID: 38757583 DOI: 10.1080/07420528.2024.2351515] [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: 08/24/2023] [Revised: 03/20/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024]
Abstract
Stable and entrainable physiological circadian rhythms are crucial for overall health and well-being. The suprachiasmatic nucleus (SCN), the primary circadian pacemaker in mammals, consists of diverse neuron types that collectively generate a circadian profile of electrical activity. However, the mechanisms underlying the regulation of endogenous neuronal excitability in the SCN remain unclear. Two-pore domain potassium channels (K2P), including TASK-3, are known to play a significant role in maintaining SCN diurnal homeostasis by inhibiting neuronal activity at night. In this study, we investigated the role of TASK-3 in SCN circadian neuronal regulation and behavioural photoentrainment using a TASK-3 global knockout mouse model. Our findings demonstrate the importance of TASK-3 in maintaining SCN hyperpolarization during the night and establishing SCN sensitivity to glutamate. Specifically, we observed that TASK-3 knockout mice lacked diurnal variation in resting membrane potential and exhibited altered glutamate sensitivity both in vivo and in vitro. Interestingly, despite these changes, the mice lacking TASK-3 were still able to maintain relatively normal circadian behaviour.
Collapse
Affiliation(s)
| | - Tatjana Lalic
- Translational Molecular Neuroscience Group, University of Oxford, Oxford, UK
| | | | - Neil Tanday
- Medway School of Pharmacy, University of Kent, Kent, UK
| | - Lorna Brown
- Medway School of Pharmacy, University of Kent, Kent, UK
| | | | - Zameel M Cader
- Translational Molecular Neuroscience Group, University of Oxford, Oxford, UK
| | | |
Collapse
|
5
|
Echeverría F, Gonzalez-Sanabria N, Alvarado-Sanchez R, Fernández M, Castillo K, Latorre R. Large conductance voltage-and calcium-activated K + (BK) channel in health and disease. Front Pharmacol 2024; 15:1373507. [PMID: 38584598 PMCID: PMC10995336 DOI: 10.3389/fphar.2024.1373507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/12/2024] [Indexed: 04/09/2024] Open
Abstract
Large Conductance Voltage- and Calcium-activated K+ (BK) channels are transmembrane pore-forming proteins that regulate cell excitability and are also expressed in non-excitable cells. They play a role in regulating vascular tone, neuronal excitability, neurotransmitter release, and muscle contraction. Dysfunction of the BK channel can lead to arterial hypertension, hearing disorders, epilepsy, and ataxia. Here, we provide an overview of BK channel functioning and the implications of its abnormal functioning in various diseases. Understanding the function of BK channels is crucial for comprehending the mechanisms involved in regulating vital physiological processes, both in normal and pathological conditions, controlled by BK. This understanding may lead to the development of therapeutic interventions to address BK channelopathies.
Collapse
Affiliation(s)
- Felipe Echeverría
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Naileth Gonzalez-Sanabria
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Rosangelina Alvarado-Sanchez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Miguel Fernández
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Investigación de Estudios Avanzados del Maule, Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
6
|
Abstract
The timing of life on Earth is remarkable: between individuals of the same species, a highly similar temporal pattern is observed, with shared periods of activity and inactivity each day. At the individual level, this means that over the course of a single day, a person alternates between two states. They are either upright, active, and communicative or they lie down in a state of (un)consciousness called sleep where even the characteristic of neuronal signals in the brain shows distinctive properties. The circadian clock governs both of these time stamps-activity and (apparent) inactivity-making them come and go consistently at the same approximate time each day. This behavior thus represents the meeting of two pervasive systems: the circadian clock and metabolism. In this article, we will describe what is known about how the circadian clock anticipates daily changes in oxygen usage, how circadian clock regulation may relate to normal physiology, and to hypoxia and ischemia that can result from pathologies such as myocardial infarction and stroke.
Collapse
Affiliation(s)
- Francesca Sartor
- Institute of Medical Psychology, Medical Faculty, LMU Munich, Germany (F.S., B.F.-B., M.M.)
| | - Borja Ferrero-Bordera
- Institute of Medical Psychology, Medical Faculty, LMU Munich, Germany (F.S., B.F.-B., M.M.)
| | - Jeffrey Haspel
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO (J.H.)
| | - Markus Sperandio
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine, and the Biomedical Center (BMC), Medical Faculty, LMU Munich, Germany (M.S.)
| | - Paul M Holloway
- Radcliffe Department of Medicine, University of Oxford, United Kingdom (P.M.H.)
| | - Martha Merrow
- Institute of Medical Psychology, Medical Faculty, LMU Munich, Germany (F.S., B.F.-B., M.M.)
| |
Collapse
|
7
|
Abstract
Novel KCNMA1 variants, encoding the BK K+ channel, are associated with a debilitating dyskinesia and epilepsy syndrome. Neurodevelopmental delay, cognitive disability, and brain and structural malformations are also diagnosed at lower incidence. More than half of affected individuals present with a rare negative episodic motor disorder, paroxysmal nonkinesigenic dyskinesia (PNKD3). The mechanistic relationship of PNKD3 to epilepsy and the broader spectrum of KCNMA1-associated symptomology is unknown. This review summarizes patient-associated KCNMA1 variants within the BK channel structure, functional classifications, genotype-phenotype associations, disease models, and treatment. Patient and transgenic animal data suggest delineation of gain-of-function (GOF) and loss-of-function KCNMA1 neurogenetic disease, validating two heterozygous alleles encoding GOF BK channels (D434G and N999S) as causing seizure and PNKD3. This discovery led to a variant-defined therapeutic approach for PNKD3, providing initial insight into the neurological basis. A comprehensive clinical definition of monogenic KCNMA1-linked disease and the neuronal mechanisms currently remain priorities for continued investigation.
Collapse
Affiliation(s)
- Andrea L Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA;
| |
Collapse
|
8
|
Dinsdale RL, Roache CE, Meredith AL. Disease-associated KCNMA1 variants decrease circadian clock robustness in channelopathy mouse models. J Gen Physiol 2023; 155:e202313357. [PMID: 37728576 PMCID: PMC10510740 DOI: 10.1085/jgp.202313357] [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: 01/30/2023] [Revised: 07/07/2023] [Accepted: 08/24/2023] [Indexed: 09/21/2023] Open
Abstract
KCNMA1 encodes the voltage- and calcium-activated K+ (BK) channel, which regulates suprachiasmatic nucleus (SCN) neuronal firing and circadian behavioral rhythms. Gain-of-function (GOF) and loss-of-function (LOF) alterations in BK channel activity disrupt circadian behavior, but the effect of human disease-associated KCNMA1 channelopathy variants has not been studied on clock function. Here, we assess circadian behavior in two GOF and one LOF mouse lines. Heterozygous Kcnma1N999S/WT and homozygous Kcnma1D434G/D434G mice are validated as GOF models of paroxysmal dyskinesia (PNKD3), but whether circadian rhythm is affected in this hypokinetic locomotor disorder is unknown. Conversely, homozygous LOF Kcnma1H444Q/H444Q mice do not demonstrate PNKD3. We assessed circadian behavior by locomotor wheel running activity. All three mouse models were rhythmic, but Kcnma1N999S/WT and Kcnma1D434G/D434G showed reduced circadian amplitude and decreased wheel activity, corroborating prior studies focused on acute motor coordination. In addition, Kcnma1D434G/D434G mice had a small decrease in period. However, the phase-shifting sensitivity for both GOF mouse lines was abnormal. Both Kcnma1N999S/WT and Kcnma1D434G/D434G mice displayed increased responses to light pulses and took fewer days to re-entrain to a new light:dark cycle. In contrast, the LOF Kcnma1H444Q/H444Q mice showed no difference in any of the circadian parameters tested. The enhanced sensitivity to phase-shifting stimuli in Kcnma1N999S/WT and Kcnma1D434G/D434G mice was similar to other Kcnma1 GOF mice. Together with previous studies, these results suggest that increasing BK channel activity decreases circadian clock robustness, without rhythm ablation.
Collapse
Affiliation(s)
- Ria L. Dinsdale
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cooper E. Roache
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrea L. Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Hermanstyne TO, Yang ND, Granados-Fuentes D, Li X, Mellor RL, Jegla T, Herzog ED, Nerbonne JM. Kv12-encoded K+ channels drive the day-night switch in the repetitive firing rates of SCN neurons. J Gen Physiol 2023; 155:e202213310. [PMID: 37516908 PMCID: PMC10373311 DOI: 10.1085/jgp.202213310] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/11/2023] [Accepted: 07/06/2023] [Indexed: 07/31/2023] Open
Abstract
Considerable evidence suggests that day-night rhythms in the functional expression of subthreshold potassium (K+) channels regulate daily oscillations in the spontaneous firing rates of neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals. The K+ conductance(s) driving these daily rhythms in the repetitive firing rates of SCN neurons, however, have not been identified. To test the hypothesis that subthreshold Kv12.1/Kv12.2-encoded K+ channels play a role, we obtained current-clamp recordings from SCN neurons in slices prepared from adult mice harboring targeted disruptions in the Kcnh8 (Kv12.1-/-) or Kcnh3 (Kv12.2-/-) locus. We found that mean nighttime repetitive firing rates were higher in Kv12.1-/- and Kv12.2-/- than in wild type (WT), SCN neurons. In marked contrast, mean daytime repetitive firing rates were similar in Kv12.1-/-, Kv12.2-/-, and WT SCN neurons, and the day-night difference in mean repetitive firing rates, a hallmark feature of WT SCN neurons, was eliminated in Kv12.1-/- and Kv12.2-/- SCN neurons. Similar results were obtained with in vivo shRNA-mediated acute knockdown of Kv12.1 or Kv12.2 in adult SCN neurons. Voltage-clamp experiments revealed that Kv12-encoded current densities in WT SCN neurons are higher at night than during the day. In addition, the pharmacological block of Kv12-encoded currents increased the mean repetitive firing rate of nighttime, but not daytime, in WT SCN neurons. Dynamic clamp-mediated subtraction of modeled Kv12-encoded currents also selectively increased the mean repetitive firing rates of nighttime WT SCN neurons. Despite the elimination of the nighttime decrease in the mean repetitive firing rates of SCN neurons, however, locomotor (wheel-running) activity remained rhythmic in Kv12.1-/-, Kv12.2-/-, and Kv12.1-targeted shRNA-expressing, and Kv12.2-targeted shRNA-expressing animals.
Collapse
Affiliation(s)
- Tracey O. Hermanstyne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nien-Du Yang
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | | | - Xiaofan Li
- Department of Biology, The Pennsylvania State University, University Park, State College, PA, USA
| | - Rebecca L. Mellor
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Timothy Jegla
- Department of Biology, The Pennsylvania State University, University Park, State College, PA, USA
| | - Erik D. Herzog
- Department of Biology, Washington University, St. Louis, MO, USA
| | - Jeanne M. Nerbonne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| |
Collapse
|
10
|
Yang ND, Mellor RL, Hermanstyne TO, Nerbonne JM. Effects of NALCN-Encoded Na + Leak Currents on the Repetitive Firing Properties of SCN Neurons Depend on K +-Driven Rhythmic Changes in Input Resistance. J Neurosci 2023; 43:5132-5141. [PMID: 37339878 PMCID: PMC10342223 DOI: 10.1523/jneurosci.0182-23.2023] [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: 01/30/2023] [Revised: 06/02/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
Neurons in the suprachiasmatic nucleus (SCN) generate circadian changes in the rates of spontaneous action potential firing that regulate and synchronize daily rhythms in physiology and behavior. Considerable evidence suggests that daily rhythms in the repetitive firing rates (higher during the day than at night) of SCN neurons are mediated by changes in subthreshold potassium (K+) conductance(s). An alternative "bicycle" model for circadian regulation of membrane excitability in clock neurons, however, suggests that an increase in NALCN-encoded sodium (Na+) leak conductance underlies daytime increases in firing rates. The experiments reported here explored the role of Na+ leak currents in regulating daytime and nighttime repetitive firing rates in identified adult male and female mouse SCN neurons: vasoactive intestinal peptide-expressing (VIP+), neuromedin S-expressing (NMS+) and gastrin-releasing peptide-expressing (GRP+) cells. Whole-cell recordings from VIP+, NMS+, and GRP+ neurons in acute SCN slices revealed that Na+ leak current amplitudes/densities are similar during the day and at night, but have a larger impact on membrane potentials in daytime neurons. Additional experiments, using an in vivo conditional knockout approach, demonstrated that NALCN-encoded Na+ currents selectively regulate daytime repetitive firing rates of adult SCN neurons. Dynamic clamp-mediated manipulation revealed that the effects of NALCN-encoded Na+ currents on the repetitive firing rates of SCN neurons depend on K+ current-driven changes in input resistances. Together, these findings demonstrate that NALCN-encoded Na+ leak channels contribute to regulating daily rhythms in the excitability of SCN neurons by a mechanism that depends on K+ current-mediated rhythmic changes in intrinsic membrane properties.SIGNIFICANCE STATEMENT Elucidating the ionic mechanisms responsible for generating daily rhythms in the rates of spontaneous action potential firing of neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals, is an important step toward understanding how the molecular clock controls circadian rhythms in physiology and behavior. While numerous studies have focused on identifying subthreshold K+ channel(s) that mediate day-night changes in the firing rates of SCN neurons, a role for Na+ leak currents has also been suggested. The results of the experiments presented here demonstrate that NALCN-encoded Na+ leak currents differentially modulate daily rhythms in the daytime/nighttime repetitive firing rates of SCN neurons as a consequence of rhythmic changes in subthreshold K+ currents.
Collapse
Affiliation(s)
- Nien-Du Yang
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri 63110
| | | | - Tracey O Hermanstyne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Jeanne M Nerbonne
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri 63110
- Department of Medicine, Cardiovascular Division
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
11
|
Gonzalez JC, Lee H, Vincent AM, Hill AL, Goode LK, King GD, Gamble KL, Wadiche JI, Overstreet-Wadiche L. Circadian regulation of dentate gyrus excitability mediated by G-protein signaling. Cell Rep 2023; 42:112039. [PMID: 36749664 PMCID: PMC10404305 DOI: 10.1016/j.celrep.2023.112039] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 10/27/2022] [Accepted: 01/12/2023] [Indexed: 02/08/2023] Open
Abstract
The central circadian regulator within the suprachiasmatic nucleus transmits time of day information by a diurnal spiking rhythm driven by molecular clock genes controlling membrane excitability. Most brain regions, including the hippocampus, harbor similar intrinsic circadian transcriptional machinery, but whether these molecular programs generate oscillations of membrane properties is unclear. Here, we show that intrinsic excitability of mouse dentate granule neurons exhibits a 24-h oscillation that controls spiking probability. Diurnal changes in excitability are mediated by antiphase G-protein regulation of potassium and sodium currents that reduce excitability during the Light phase. Disruption of the circadian transcriptional machinery by conditional deletion of Bmal1 enhances excitability selectively during the Light phase by removing G-protein regulation. These results reveal that circadian transcriptional machinery regulates intrinsic excitability by coordinated regulation of ion channels by G-protein signaling, highlighting a potential novel mechanism of cell-autonomous oscillations.
Collapse
Affiliation(s)
- Jose Carlos Gonzalez
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Haeun Lee
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Angela M Vincent
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Angela L Hill
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lacy K Goode
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gwendalyn D King
- Department of Biology, Creighton University, Omaha, NE 68178, USA
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jacques I Wadiche
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Linda Overstreet-Wadiche
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
12
|
Ancatén-González C, Segura I, Alvarado-Sánchez R, Chávez AE, Latorre R. Ca 2+- and Voltage-Activated K + (BK) Channels in the Nervous System: One Gene, a Myriad of Physiological Functions. Int J Mol Sci 2023; 24:3407. [PMID: 36834817 PMCID: PMC9967218 DOI: 10.3390/ijms24043407] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 02/11/2023] Open
Abstract
BK channels are large conductance potassium channels characterized by four pore-forming α subunits, often co-assembled with auxiliary β and γ subunits to regulate Ca2+ sensitivity, voltage dependence and gating properties. BK channels are abundantly expressed throughout the brain and in different compartments within a single neuron, including axons, synaptic terminals, dendritic arbors, and spines. Their activation produces a massive efflux of K+ ions that hyperpolarizes the cellular membrane. Together with their ability to detect changes in intracellular Ca2+ concentration, BK channels control neuronal excitability and synaptic communication through diverse mechanisms. Moreover, increasing evidence indicates that dysfunction of BK channel-mediated effects on neuronal excitability and synaptic function has been implicated in several neurological disorders, including epilepsy, fragile X syndrome, mental retardation, and autism, as well as in motor and cognitive behavior. Here, we discuss current evidence highlighting the physiological importance of this ubiquitous channel in regulating brain function and its role in the pathophysiology of different neurological disorders.
Collapse
Affiliation(s)
- Carlos Ancatén-González
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Ignacio Segura
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Rosangelina Alvarado-Sánchez
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Doctorado en Ciencias Mención Biofísica y Biología Computacional, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Andrés E. Chávez
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| |
Collapse
|
13
|
Hermanstyne TO, Yang ND, Granados-Fuentes D, Li X, Mellor RL, Jegla T, Herzog ED, Nerbonne JM. Kv12-Encoded K + Channels Drive the Day-Night Switch in the Repetitive Firing Rates of SCN Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526323. [PMID: 36778242 PMCID: PMC9915524 DOI: 10.1101/2023.01.30.526323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Considerable evidence suggests that day-night rhythms in the functional expression of subthreshold potassium (K + ) channels regulate daily oscillations in the rates of spontaneous action potential firing of neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals. The K + conductance(s) driving these daily rhythms in repetitive firing rates, however, have not been identified. To test the hypothesis that subthreshold Kv12.1/Kv12.2-encoded K + channels play a role, we obtained current-clamp recordings from SCN neurons in slices prepared from adult mice harboring targeted disruptions in the Kcnh8 (Kv12.1 -/- ) or Kcnh3 (Kv12.2 -/- ) locus. We found that mean nighttime repetitive firing rates were higher in Kv12.1 -/- and Kv12.2 -/- , than in wild type (WT), SCN neurons. In marked contrast, mean daytime repetitive firing rates were similar in Kv12.1 -/- , Kv12.2 -/- and WT SCN neurons, and the day-night difference in mean repetitive firing rates, a hallmark feature of WT SCN neurons, was eliminated in Kv12.1 -/- and Kv12.2 -/- SCN neurons. Similar results were obtained with in vivo shRNA-mediated acute knockdown of Kv12.1 or Kv12.2 in adult SCN neurons. Voltage-clamp experiments revealed that Kv12-encoded current densities in WT SCN neurons are higher at night than during the day. In addition, pharmacological block of Kv12-encoded currents increased the mean repetitive firing rate of nighttime, but not daytime, in WT SCN neurons. Dynamic clamp-mediated subtraction of modeled Kv12-encoded currents also selectively increased the mean repetitive firing rates of nighttime WT SCN neurons. Despite the elimination of nighttime decrease in the mean repetitive firing rates of SCN neurons, however, locomotor (wheel-running) activity remained rhythmic in Kv12.1 -/- , Kv12.2 -/- , Kv12.1-targeted shRNA-expressing, and Kv12.2-targeted shRNA-expressing animals.
Collapse
Affiliation(s)
- Tracey O. Hermanstyne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
| | - Nien-Du Yang
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO
| | | | - Xiaofan Li
- Department of Biology, The Pennsylvania State University, University Park, PA
| | - Rebecca L. Mellor
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO
| | - Timothy Jegla
- Department of Biology, The Pennsylvania State University, University Park, PA
| | - Erik D. Herzog
- Department of Biology, Washington University, St. Louis, MO
| | - Jeanne M. Nerbonne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
14
|
Zheng Y, Pan L, Wang F, Yan J, Wang T, Xia Y, Yao L, Deng K, Zheng Y, Xia X, Su Z, Chen H, Lin J, Ding Z, Zhang K, Zhang M, Chen Y. Neural function of Bmal1: an overview. Cell Biosci 2023; 13:1. [PMID: 36593479 PMCID: PMC9806909 DOI: 10.1186/s13578-022-00947-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
Bmal1 (Brain and muscle arnt-like, or Arntl) is a bHLH/PAS domain transcription factor central to the transcription/translation feedback loop of the biologic clock. Although Bmal1 is well-established as a major regulator of circadian rhythm, a growing number of studies in recent years have shown that dysfunction of Bmal1 underlies a variety of psychiatric, neurodegenerative-like, and endocrine metabolism-related disorders, as well as potential oncogenic roles. In this review, we systematically summarized Bmal1 expression in different brain regions, its neurological functions related or not to circadian rhythm and biological clock, and pathological phenotypes arising from Bmal1 knockout. This review also discusses oscillation and rhythmicity, especially in the suprachiasmatic nucleus, and provides perspective on future progress in Bmal1 research.
Collapse
Affiliation(s)
- Yuanjia Zheng
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China ,grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingyun Pan
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Feixue Wang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinglan Yan
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Taiyi Wang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yucen Xia
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Yao
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kelin Deng
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqi Zheng
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoye Xia
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhikai Su
- grid.411866.c0000 0000 8848 7685The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong China
| | - Hongjie Chen
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jie Lin
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenwei Ding
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kaitong Zhang
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meng Zhang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongjun Chen
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China ,grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China ,Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China
| |
Collapse
|
15
|
Chadwick SR, Güler AD. Local Drd1-neurons input to subgroups of arcuate AgRP/NPY-neurons. iScience 2022; 25:104605. [PMID: 35789850 PMCID: PMC9250019 DOI: 10.1016/j.isci.2022.104605] [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: 11/23/2021] [Revised: 05/02/2022] [Accepted: 06/08/2022] [Indexed: 11/21/2022] Open
Abstract
Obesity is a pandemic afflicting more than 300 million people worldwide, driven by consumption of calorically dense and highly rewarding foods. Dopamine (DA) signaling has been implicated in neural responses to highly palatable nutrients, but the exact mechanisms through which DA modulates homeostatic feeding circuits remains unknown. A subpopulation of arcuate (ARC) agouti-related peptide (AgRP)/neuropeptide Y (NPY) (ARCAgRP/NPY+) neurons express the D(1A) dopamine receptor (Drd1) and are stimulated by DA, suggesting one potential avenue for dopaminergic regulation of food intake. Using patch clamp electrophysiology, we evaluated the responses of ARC Drd1-expressing (ARCDrd1+) neurons to overnight fasting and leptin. Collectively, ARCDrd1+ neurons were less responsive to caloric deficit than ARCAgRP/NPY+ neurons; however, ARCDrd1+ neurons were inhibited by the satiety hormone leptin. Using Channelrhodopsin-2-Assisted Circuit Mapping, we identified novel subgroups of ARCDrd1+ neurons that inhibit or excite ARCAgRP/NPY+ neurons. These findings suggest dopamine receptive neurons have multimodal actions in food intake circuits.
Collapse
Affiliation(s)
- Sean R. Chadwick
- Program in Fundamental Neuroscience and the Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Ali D. Güler
- Program in Fundamental Neuroscience and the Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
16
|
Park SM, Roache CE, Iffland PH, Moldenhauer HJ, Matychak KK, Plante AE, Lieberman AG, Crino PB, Meredith A. BK channel properties correlate with neurobehavioral severity in three KCNMA1-linked channelopathy mouse models. eLife 2022; 11:e77953. [PMID: 35819138 PMCID: PMC9275823 DOI: 10.7554/elife.77953] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/01/2022] [Indexed: 12/14/2022] Open
Abstract
KCNMA1 forms the pore of BK K+ channels, which regulate neuronal and muscle excitability. Recently, genetic screening identified heterozygous KCNMA1 variants in a subset of patients with debilitating paroxysmal non-kinesigenic dyskinesia, presenting with or without epilepsy (PNKD3). However, the relevance of KCNMA1 mutations and the basis for clinical heterogeneity in PNKD3 has not been established. Here, we evaluate the relative severity of three KCNMA1 patient variants in BK channels, neurons, and mice. In heterologous cells, BKN999S and BKD434G channels displayed gain-of-function (GOF) properties, whereas BKH444Q channels showed loss-of-function (LOF) properties. The relative degree of channel activity was BKN999S > BKD434G>WT > BKH444Q. BK currents and action potential firing were increased, and seizure thresholds decreased, in Kcnma1N999S/WT and Kcnma1D434G/WT transgenic mice but not Kcnma1H444Q/WT mice. In a novel behavioral test for paroxysmal dyskinesia, the more severely affected Kcnma1N999S/WT mice became immobile after stress. This was abrogated by acute dextroamphetamine treatment, consistent with PNKD3-affected individuals. Homozygous Kcnma1D434G/D434G mice showed similar immobility, but in contrast, homozygous Kcnma1H444Q/H444Q mice displayed hyperkinetic behavior. These data establish the relative pathogenic potential of patient alleles as N999S>D434G>H444Q and validate Kcnma1N999S/WT mice as a model for PNKD3 with increased seizure propensity.
Collapse
Affiliation(s)
- Su Mi Park
- Department of Physiology, University of Maryland School of MedicineBaltimoreUnited States
| | - Cooper E Roache
- Department of Physiology, University of Maryland School of MedicineBaltimoreUnited States
| | - Philip H Iffland
- Department of Neurology, University of Maryland School of MedicineBaltimoreUnited States
| | - Hans J Moldenhauer
- Department of Physiology, University of Maryland School of MedicineBaltimoreUnited States
| | - Katia K Matychak
- Department of Physiology, University of Maryland School of MedicineBaltimoreUnited States
| | - Amber E Plante
- Department of Physiology, University of Maryland School of MedicineBaltimoreUnited States
| | - Abby G Lieberman
- Department of Pharmacology, University of Maryland School of MedicineBaltimoreUnited States
| | - Peter B Crino
- Department of Neurology, University of Maryland School of MedicineBaltimoreUnited States
| | - Andrea Meredith
- Department of Physiology, University of Maryland School of MedicineBaltimoreUnited States
| |
Collapse
|
17
|
Morioka E, Kasuga Y, Kanda Y, Moritama S, Koizumi H, Yoshikawa T, Miura N, Ikeda M, Higashida H, Holmes TC, Ikeda M. Mitochondrial LETM1 drives ionic and molecular clock rhythms in circadian pacemaker neurons. Cell Rep 2022; 39:110787. [PMID: 35545046 DOI: 10.1016/j.celrep.2022.110787] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/14/2022] [Accepted: 04/14/2022] [Indexed: 11/18/2022] Open
Abstract
The mechanisms that generate robust ionic oscillation in circadian pacemaker neurons are under investigation. Here, we demonstrate critical functions of the mitochondrial cation antiporter leucine zipper-EF-hand-containing transmembrane protein 1 (LETM1), which exchanges K+/H+ in Drosophila and Ca2+/H+ in mammals, in circadian pacemaker neurons. Letm1 knockdown in Drosophila pacemaker neurons reduced circadian cytosolic H+ rhythms and prolonged nuclear PERIOD/TIMELESS expression rhythms and locomotor activity rhythms. In rat pacemaker neurons in the hypothalamic suprachiasmatic nucleus (SCN), circadian rhythms in cytosolic Ca2+ and Bmal1 transcription were dampened by Letm1 knockdown. Mitochondrial Ca2+ uptake peaks late during the day were also observed in rat SCN neurons following photolytic elevation of cytosolic Ca2+. Since cation transport by LETM1 is coupled to mitochondrial energy synthesis, we propose that LETM1 integrates metabolic, ionic, and molecular clock rhythms in the central clock system in both invertebrates and vertebrates.
Collapse
Affiliation(s)
- Eri Morioka
- Graduate School of Science and Engineering, University of Toyama, Gofuku, Toyama 930-8555, Japan
| | - Yusuke Kasuga
- Graduate School of Science and Engineering, University of Toyama, Gofuku, Toyama 930-8555, Japan
| | - Yuzuki Kanda
- Graduate School of Science and Engineering, University of Toyama, Gofuku, Toyama 930-8555, Japan
| | - Saki Moritama
- Graduate School of Science and Engineering, University of Toyama, Gofuku, Toyama 930-8555, Japan
| | - Hayato Koizumi
- Graduate School of Innovative Life Science, University of Toyama, Gofuku, Toyama 930-8555, Japan
| | - Tomoko Yoshikawa
- Organization for International Education and Exchange, University of Toyama, Toyama 930-8555, Japan
| | - Nobuhiko Miura
- Department of Health Medicine, Yokohama University of Pharmacy, Yokohama, Kanagawa 245-0061, Japan
| | - Masaaki Ikeda
- Department of Physiology, Faculty of Medicine, Saitama Medical University, Saitama 350-0495, Japan
| | - Haruhiro Higashida
- Research Center for Child Mental Development, Kanazawa University, Ishikawa 920-8640, Japan
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Masayuki Ikeda
- Graduate School of Innovative Life Science, University of Toyama, Gofuku, Toyama 930-8555, Japan; Organization for International Education and Exchange, University of Toyama, Toyama 930-8555, Japan.
| |
Collapse
|
18
|
North KC, Zhang M, Singh AK, Zaytseva D, Slayden AV, Bukiya AN, Dopico AM. Cholesterol Inhibition of Slo1 Channels Is Calcium-Dependent and Can Be Mediated by Either High-Affinity Calcium-Sensing Site in the Slo1 Cytosolic Tail. Mol Pharmacol 2022; 101:132-143. [PMID: 34969832 PMCID: PMC8969144 DOI: 10.1124/molpharm.121.000392] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/27/2021] [Indexed: 11/22/2022] Open
Abstract
Calcium- and voltage-gated K+ channels of large conductance (BKs) are expressed in the cell membranes of all excitable tissues. Currents mediated by BK channel-forming slo1 homotetramers are consistently inhibited by increases in membrane cholesterol (CLR). The molecular mechanisms leading to this CLR action, however, remain unknown. Slo1 channels are activated by increases in calcium (Ca2+) nearby Ca2+-recognition sites in the slo1 cytosolic tail: one high-affinity and one low-affinity site locate to the regulator of conductance for K+ (RCK) 1 domain, whereas another high-affinity site locates within the RCK2 domain. Here, we first evaluated the crosstalking between Ca2+ and CLR on the function of slo1 (cbv1 isoform) channels reconstituted into planar lipid bilayers. CLR robustly reduced channel open probability while barely decreasing unitary current amplitude, with CLR maximal effects being observed at 10-30 µM internal Ca2+ CLR actions were not only modulated by internal Ca2+ levels but also disappeared in absence of this divalent. Moreover, in absence of Ca2+, BK channel-activating concentrations of magnesium (10 mM) did not support CLR action. Next, we evaluated CLR actions on channels where the different Ca2+-sensing sites present in the slo1 cytosolic domain became nonfunctional via mutagenesis. CLR still reduced the activity of low-affinity Ca2+ (RCK1:E379A, E404A) mutants. In contrast, CLR became inefficacious when both high-affinity Ca2+ sites were mutated (RCK1:D367A,D372A and RCK2:D899N,D900N,D901N,D902N,D903N), yet still was able to decrease the activity of each high-affinity site mutant. Therefore, BK channel inhibition by CLR selectively requires optimal levels of Ca2+ being recognized by either of the slo1 high-affinity Ca2+-sensing sites. SIGNIFICANCE STATEMENT: Results reveal that inhibition of calcium/voltage-gated K+ channel of large conductance (BK) (slo1) channels by membrane cholesterol requires a physiologically range of internal calcium (Ca2+) and is selectively linked to the two high-affinity Ca2+-sensing sites located in the cytosolic tail domain, which underscores that Ca2+ and cholesterol actions are allosterically coupled to the channel gate. Cholesterol modification of BK channel activity likely contributes to disruption of normal physiology by common health conditions that are triggered by disruption of cholesterol homeostasis.
Collapse
Affiliation(s)
- Kelsey C North
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Man Zhang
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Aditya K Singh
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Dasha Zaytseva
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Alexandria V Slayden
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
19
|
Moldenhauer HJ, Dinsdale RL, Alvarez S, Fernández-Jaén A, Meredith AL. Effect of an autism-associated KCNMB2 variant, G124R, on BK channel properties. Curr Res Physiol 2022; 5:404-413. [PMID: 36203817 PMCID: PMC9531041 DOI: 10.1016/j.crphys.2022.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/26/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
BK K+ channels are critical regulators of neuron and muscle excitability, comprised of a tetramer of pore-forming αsubunits from the KCNMA1 gene and cell- and tissue-selective β subunits (KCNMB1-4). Mutations in KCNMA1 are associated with neurological disorders, including autism. However, little is known about the role of neuronal BK channel β subunits in human neuropathology. The β2 subunit is expressed in central neurons and imparts inactivation to BK channels, as well as altering activation and deactivation gating. In this study, we report the functional effect of G124R, a novel KCNMB2 mutation obtained from whole-exome sequencing of a patient diagnosed with autism spectrum disorder. Residue G124, located in the extracellular loop between TM1 and TM2, is conserved across species, and the G124R missense mutation is predicted deleterious with computational tools. To investigate the pathogenicity potential, BK channels were co-expressed with β2WT and β2G124R subunits in HEK293T cells. BK/β2 currents were assessed from inside-out patches under physiological K+ conditions (140/6 mM K+ and 10 μM Ca2+) during activation and inactivation (voltage-dependence and kinetics). Using β2 subunits lacking inactivation (β2IR) revealed that currents from BK/β2IRG124R channels activated 2-fold faster and deactivated 2-fold slower compared with currents from BK/β2IRWT channels, with no change in the voltage-dependence of activation (V1/2). Despite the changes in the BK channel opening and closing, BK/β2G124R inactivation rates (τinact and τrecovery), and the V1/2 of inactivation, were unaltered compared with BK/β2WT channels under standard steady-state voltage protocols. Action potential-evoked current was also unchanged. Thus, the mutant phenotype suggests the β2G124R TM1-TM2 extracellular loop could regulate BK channel activation and deactivation kinetics. However, additional evidence is needed to validate pathogenicity for this patient-associated variant in KCNMB2. KCNMA1 channelopathy is a neurobehavioral disorder associated with seizures, dyskinesia, and intellectual disability. KCNMB2 encodes an accessory β subunit that confers inactivation to the KCNMA1 pore-forming α subunit BK channel. The KCNMB2-G124R variant, identified in an autistic individual, affects BK/β2 channel activation but not inactivation.
Collapse
Affiliation(s)
- Hans J. Moldenhauer
- Dept. of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ria L. Dinsdale
- Dept. of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Alberto Fernández-Jaén
- Dept. of Pediatric Neurology, Hospital Universitario Quirónsalud, School of Medicine, Universidad Europea de, Madrid, Spain
| | - Andrea L. Meredith
- Dept. of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Corresponding author. Dept. of Physiology University of Maryland School of Medicine, 655 W. Baltimore St. Baltimore, MD, 21201, USA.
| |
Collapse
|
20
|
Bano-Otalora B, Moye MJ, Brown T, Lucas RJ, Diekman CO, Belle MD. Daily electrical activity in the master circadian clock of a diurnal mammal. eLife 2021; 10:68179. [PMID: 34845984 PMCID: PMC8631794 DOI: 10.7554/elife.68179] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 10/09/2021] [Indexed: 11/13/2022] Open
Abstract
Circadian rhythms in mammals are orchestrated by a central clock within the suprachiasmatic nuclei (SCN). Our understanding of the electrophysiological basis of SCN activity comes overwhelmingly from a small number of nocturnal rodent species, and the extent to which these are retained in day-active animals remains unclear. Here, we recorded the spontaneous and evoked electrical activity of single SCN neurons in the diurnal rodent Rhabdomys pumilio, and developed cutting-edge data assimilation and mathematical modeling approaches to uncover the underlying ionic mechanisms. As in nocturnal rodents, R. pumilio SCN neurons were more excited during daytime hours. By contrast, the evoked activity of R. pumilio neurons included a prominent suppressive response that is not present in the SCN of nocturnal rodents. Our modeling revealed and subsequent experiments confirmed transient subthreshold A-type potassium channels as the primary determinant of this response, and suggest a key role for this ionic mechanism in optimizing SCN function to accommodate R. pumilio's diurnal niche.
Collapse
Affiliation(s)
- Beatriz Bano-Otalora
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom.,Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Matthew J Moye
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, United States.,Department of Quantitative Pharmacology and Pharmacometrics (QP2), Kenilworth, United States
| | - Timothy Brown
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Robert J Lucas
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom.,Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Casey O Diekman
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, United States.,EPSRC Centre for Predictive Modelling in Healthcare, Living Systems Institute, University of Exeter, Exeter, United Kingdom
| | - Mino Dc Belle
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
21
|
McNally BA, Plante AE, Meredith AL. Contributions of Ca V1.3 Channels to Ca 2+ Current and Ca 2+-Activated BK Current in the Suprachiasmatic Nucleus. Front Physiol 2021; 12:737291. [PMID: 34650447 PMCID: PMC8505962 DOI: 10.3389/fphys.2021.737291] [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: 07/06/2021] [Accepted: 08/30/2021] [Indexed: 11/29/2022] Open
Abstract
Daily regulation of Ca2+– and voltage-activated BK K+ channel activity is required for action potential rhythmicity in the suprachiasmatic nucleus (SCN) of the hypothalamus, the brain's circadian clock. In SCN neurons, BK activation is dependent upon multiple types of Ca2+ channels in a circadian manner. Daytime BK current predominantly requires Ca2+ influx through L-type Ca2+ channels (LTCCs), a time when BK channels are closely coupled with their Ca2+ source. Here we show that daytime BK current is resistant to the Ca2+ chelator BAPTA. However, at night when LTCCs contribute little to BK activation, BK current decreases by a third in BAPTA compared to control EGTA conditions. In phase with this time-of-day specific effect on BK current activation, LTCC current is larger during the day. The specific Ca2+ channel subtypes underlying the LTCC current in SCN, as well as the subtypes contributing the Ca2+ influx relevant for BK current activation, have not been identified. SCN neurons express two LTCC subtypes, CaV1.2 and CaV1.3. While a role for CaV1.2 channels has been identified during the night, CaV1.3 channel modulation has also been suggested to contribute to daytime SCN action potential activity, as well as subthreshold Ca2+ oscillations. Here we characterize the role of CaV1.3 channels in LTCC and BK current activation in SCN neurons using a global deletion of CACNA1D in mouse (CaV1.3 KO). CaV1.3 KO SCN neurons had a 50% reduction in the daytime LTCC current, but not total Ca2+ current, with no difference in Ca2+ current levels at night. During the day, CaV1.3 KO neurons exhibited oscillations in membrane potential, and most neurons, although not all, also had BK currents. Changes in BK current activation were only detectable at the highest voltage tested. These data show that while CaV1.3 channels contribute to the daytime Ca2+ current, this does not translate into a major effect on the daytime BK current. These data suggest that BK current activation does not absolutely require CaV1.3 channels and may therefore also depend on other LTCC subtypes, such as CaV1.2.
Collapse
Affiliation(s)
- Beth A McNally
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Amber E Plante
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Andrea L Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
22
|
Jones JR, Chaturvedi S, Granados-Fuentes D, Herzog ED. Circadian neurons in the paraventricular nucleus entrain and sustain daily rhythms in glucocorticoids. Nat Commun 2021; 12:5763. [PMID: 34599158 PMCID: PMC8486846 DOI: 10.1038/s41467-021-25959-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 09/02/2021] [Indexed: 02/08/2023] Open
Abstract
Signals from the central circadian pacemaker, the suprachiasmatic nucleus (SCN), must be decoded to generate daily rhythms in hormone release. Here, we hypothesized that the SCN entrains rhythms in the paraventricular nucleus (PVN) to time the daily release of corticosterone. In vivo recording revealed a critical circuit from SCN vasoactive intestinal peptide (SCNVIP)-producing neurons to PVN corticotropin-releasing hormone (PVNCRH)-producing neurons. PVNCRH neurons peak in clock gene expression around midday and in calcium activity about three hours later. Loss of the clock gene Bmal1 in CRH neurons results in arrhythmic PVNCRH calcium activity and dramatically reduces the amplitude and precision of daily corticosterone release. SCNVIP activation reduces (and inactivation increases) corticosterone release and PVNCRH calcium activity, and daily SCNVIP activation entrains PVN clock gene rhythms by inhibiting PVNCRH neurons. We conclude that daily corticosterone release depends on coordinated clock gene and neuronal activity rhythms in both SCNVIP and PVNCRH neurons.
Collapse
Affiliation(s)
- Jeff R Jones
- Department of Biology, Washington University, St. Louis, St. Louis, MO, USA
- Department of Biology, Texas A&M University, College Station, College Station, TX, USA
| | - Sneha Chaturvedi
- Department of Biology, Washington University, St. Louis, St. Louis, MO, USA
| | | | - Erik D Herzog
- Department of Biology, Washington University, St. Louis, St. Louis, MO, USA.
| |
Collapse
|
23
|
Plante AE, Rao VP, Rizzo MA, Meredith AL. Comparative Ca 2+ channel contributions to intracellular Ca 2+ levels in the circadian clock. BIOPHYSICAL REPORTS 2021; 1:100005. [PMID: 35330949 PMCID: PMC8942421 DOI: 10.1016/j.bpr.2021.100005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/08/2021] [Indexed: 11/22/2022]
Abstract
Circadian rhythms in mammals are coordinated by the central clock in the brain, located in the suprachiasmatic nucleus (SCN). Multiple molecular and cellular signals display a circadian variation within SCN neurons, including intracellular Ca2+, but the mechanisms are not definitively established. SCN cytosolic Ca2+ levels exhibit a peak during the day, when both action potential firing and Ca2+ channel activity are increased, and are decreased at night, correlating with a reduction in firing rate. In this study, we employ a single-color fluorescence anisotropy reporter (FLARE), Venus FLARE-Cameleon, and polarization inverted selective-plane illumination microscopy to measure rhythmic changes in cytosolic Ca2+ in SCN neurons. Using this technique, the Ca2+ channel subtypes contributing to intracellular Ca2+ at the peak and trough of the circadian cycle were assessed using a pharmacological approach with Ca2+ channel inhibitors. Peak (218 ± 16 nM) and trough (172 ± 13 nM) Ca2+ levels were quantified, indicating a 1.3-fold circadian variance in Ca2+ concentration. Inhibition of ryanodine-receptor-mediated Ca2+ release produced a larger relative decrease in cytosolic Ca2+ at both time points compared to voltage-gated Ca2+channels. These results support the hypothesis that circadian Ca2+ rhythms in SCN neurons are predominantly driven by intracellular Ca2+ channels, although not exclusively so. The study provides a foundation for future experiments to probe Ca2+ signaling in a dynamic biological context using FLAREs.
Collapse
Affiliation(s)
- Amber E. Plante
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Vishnu P. Rao
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Megan A. Rizzo
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrea L. Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
24
|
Kshatri A, Cerrada A, Gimeno R, Bartolomé-Martín D, Rojas P, Giraldez T. Differential regulation of BK channels by fragile X mental retardation protein. J Gen Physiol 2021; 152:151651. [PMID: 32275741 PMCID: PMC7266151 DOI: 10.1085/jgp.201912502] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/12/2020] [Indexed: 01/28/2023] Open
Abstract
Fragile X mental retardation protein (FMRP) is an RNA-binding protein prominently expressed in neurons. Missense mutations or complete loss of FMRP can potentially lead to fragile X syndrome, a common form of inherited intellectual disability. In addition to RNA regulation, FMRP was also proposed to modulate neuronal function by direct interaction with the large conductance Ca2+- and voltage-activated potassium channel (BK) β4 regulatory subunits (BKβ4). However, the molecular mechanisms underlying FMRP regulation of BK channels were not studied in detail. We have used electrophysiology and super-resolution stochastic optical reconstruction microscopy (STORM) to characterize the effects of FMRP on pore-forming BKα subunits, as well as the association with regulatory subunits BKβ4. Our data indicate that, in the absence of coexpressed β4, FMRP alters the steady-state properties of BKα channels by decreasing channel activation and deactivation rates. Analysis using the Horrigan-Aldrich model revealed alterations in the parameters associated with channel opening (L0) and voltage sensor activation (J0). Interestingly, FMRP also altered the biophysical properties of BKαβ4 channels favoring channel opening, although not as dramatically as BKα. STORM experiments revealed clustered multi-protein complexes, consistent with FMRP interacting not only to BKαβ4 but also to BKα. Lastly, we found that a partial loss-of-function mutation in FMRP (R138Q) counteracts many of its functional effects on BKα and BKαβ4 channels. In summary, our data show that FMRP modulates the function of both BKα and BKαβ4 channels.
Collapse
Affiliation(s)
- Aravind Kshatri
- Departamento de Ciencias Medicas Basicas-Fisiologia, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Alejandro Cerrada
- Departamento de Ciencias Medicas Basicas-Fisiologia, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Roger Gimeno
- Departamento de Ciencias Medicas Basicas-Fisiologia, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - David Bartolomé-Martín
- Departamento de Ciencias Medicas Basicas-Fisiologia, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Patricio Rojas
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Teresa Giraldez
- Departamento de Ciencias Medicas Basicas-Fisiologia, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologías Biomedicas, Universidad de La Laguna, Tenerife, Spain
| |
Collapse
|
25
|
Abstract
K+ channels enable potassium to flow across the membrane with great selectivity. There are four K+ channel families: voltage-gated K (Kv), calcium-activated (KCa), inwardly rectifying K (Kir), and two-pore domain potassium (K2P) channels. All four K+ channels are formed by subunits assembling into a classic tetrameric (4x1P = 4P for the Kv, KCa, and Kir channels) or tetramer-like (2x2P = 4P for the K2P channels) architecture. These subunits can either be the same (homomers) or different (heteromers), conferring great diversity to these channels. They share a highly conserved selectivity filter within the pore but show different gating mechanisms adapted for their function. K+ channels play essential roles in controlling neuronal excitability by shaping action potentials, influencing the resting membrane potential, and responding to diverse physicochemical stimuli, such as a voltage change (Kv), intracellular calcium oscillations (KCa), cellular mediators (Kir), or temperature (K2P).
Collapse
|
26
|
Plante AE, Whitt JP, Meredith AL. BK channel activation by L-type Ca 2+ channels Ca V1.2 and Ca V1.3 during the subthreshold phase of an action potential. J Neurophysiol 2021; 126:427-439. [PMID: 34191630 DOI: 10.1152/jn.00089.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mammalian circadian (24 h) rhythms are timed by the pattern of spontaneous action potential firing in the suprachiasmatic nucleus (SCN). This oscillation in firing is produced through circadian regulation of several membrane currents, including large-conductance Ca2+- and voltage-activated K+ (BK) and L-type Ca2+ channel (LTCC) currents. During the day steady-state BK currents depend mostly on LTCCs for activation, whereas at night they depend predominantly on ryanodine receptors (RyRs). However, the contribution of these Ca2+ channels to BK channel activation during action potential firing has not been thoroughly investigated. In this study, we used a pharmacological approach to determine that both LTCCs and RyRs contribute to the baseline membrane potential of SCN action potential waveforms, as well as action potential-evoked BK current, during the day and night, respectively. Since the baseline membrane potential is a major determinant of circadian firing rate, we focused on the LTCCs contributing to low voltage activation of BK channels during the subthreshold phase. For these experiments, two LTCC subtypes found in SCN (CaV1.2 and CaV1.3) were coexpressed with BK channels in heterologous cells, where their differential contributions could be separately measured. CaV1.3 channels produced currents that were shifted to more hyperpolarized potentials compared with CaV1.2, resulting in increased subthreshold Ca2+ and BK currents during an action potential command. These results show that although multiple Ca2+ sources in SCN can contribute to the activation of BK current during an action potential, specific BK-CaV1.3 partnerships may optimize the subthreshold BK current activation that is critical for firing rate regulation.NEW & NOTEWORTHY BK K+ channels are important regulators of firing. Although Ca2+ channels are required for their activation in excitable cells, it is not well understood how BK channels activate using these Ca2+ sources during an action potential. This study demonstrates the differences in BK current activated by CaV1.2 and CaV1.3 channels in clock neurons and heterologous cells. The results define how specific ion channel partnerships can be engaged during distinct phases of the action potential.
Collapse
Affiliation(s)
- Amber E Plante
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Joshua P Whitt
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrea L Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
27
|
Bano-Otalora B, Martial F, Harding C, Bechtold DA, Allen AE, Brown TM, Belle MDC, Lucas RJ. Bright daytime light enhances circadian amplitude in a diurnal mammal. Proc Natl Acad Sci U S A 2021; 118:e2100094118. [PMID: 34031246 PMCID: PMC8179182 DOI: 10.1073/pnas.2100094118] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mammalian circadian rhythms are orchestrated by a master pacemaker in the hypothalamic suprachiasmatic nuclei (SCN), which receives information about the 24 h light-dark cycle from the retina. The accepted function of this light signal is to reset circadian phase in order to ensure appropriate synchronization with the celestial day. Here, we ask whether light also impacts another key property of the circadian oscillation, its amplitude. To this end, we measured circadian rhythms in behavioral activity, body temperature, and SCN electrophysiological activity in the diurnal murid rodent Rhabdomys pumilio following stable entrainment to 12:12 light-dark cycles at four different daytime intensities (ranging from 18 to 1,900 lx melanopic equivalent daylight illuminance). R. pumilio showed strongly diurnal activity and body temperature rhythms in all conditions, but measures of rhythm robustness were positively correlated with daytime irradiance under both entrainment and subsequent free run. Whole-cell and extracellular recordings of electrophysiological activity in ex vivo SCN revealed substantial differences in electrophysiological activity between dim and bright light conditions. At lower daytime irradiance, daytime peaks in SCN spontaneous firing rate and membrane depolarization were substantially depressed, leading to an overall marked reduction in the amplitude of circadian rhythms in spontaneous activity. Our data reveal a previously unappreciated impact of daytime light intensity on SCN physiology and the amplitude of circadian rhythms and highlight the potential importance of daytime light exposure for circadian health.
Collapse
Affiliation(s)
- Beatriz Bano-Otalora
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Franck Martial
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Court Harding
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - David A Bechtold
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Annette E Allen
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Timothy M Brown
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Mino D C Belle
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX4 4PS, United Kingdom
| | - Robert J Lucas
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| |
Collapse
|
28
|
Buijink MR, Michel S. A multi-level assessment of the bidirectional relationship between aging and the circadian clock. J Neurochem 2021; 157:73-94. [PMID: 33370457 PMCID: PMC8048448 DOI: 10.1111/jnc.15286] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022]
Abstract
The daily temporal order of physiological processes and behavior contribute to the wellbeing of many organisms including humans. The central circadian clock, which coordinates the timing within our body, is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Like in other parts of the brain, aging impairs the SCN function, which in turn promotes the development and progression of aging-related diseases. We here review the impact of aging on the different levels of the circadian clock machinery-from molecules to organs-with a focus on the role of the SCN. We find that the molecular clock is less effected by aging compared to other cellular components of the clock. Proper rhythmic regulation of intracellular signaling, ion channels and neuronal excitability of SCN neurons are greatly disturbed in aging. This suggests a disconnection between the molecular clock and the electrophysiology of these cells. The neuronal network of the SCN is able to compensate for some of these cellular deficits. However, it still results in a clear reduction in the amplitude of the SCN electrical rhythm, suggesting a weakening of the output timing signal. Consequently, other brain areas and organs not only show aging-related deficits in their own local clocks, but also receive a weaker systemic timing signal. The negative spiral completes with the weakening of positive feedback from the periphery to the SCN. Consequently, chronotherapeutic interventions should aim at strengthening overall synchrony in the circadian system using life-style and/or pharmacological approaches.
Collapse
Affiliation(s)
- M. Renate Buijink
- Department of Cellular and Chemical BiologyLaboratory for NeurophysiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Stephan Michel
- Department of Cellular and Chemical BiologyLaboratory for NeurophysiologyLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
29
|
Sominsky L, Dangel T, Malik S, De Luca SN, Singewald N, Spencer SJ. Microglial ablation in rats disrupts the circadian system. FASEB J 2021; 35:e21195. [PMID: 33200466 DOI: 10.1096/fj.202001555rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 01/26/2023]
Abstract
Microglia, the key neuroimmune cells of the central nervous system, are best known for their function in defending an individual from pathogens and injury. Recent findings, including our own, suggest microglia also have several immune-independent roles, including in regulating satiety, promoting memory, and modifying pain responses. Many of these microglia-associated functions are affected by circadian rhythmicity, thus, varying substantially depending upon the time of day. To gain further insight into this link, we used a Cx3cr1-Dtr transgenic Wistar rat model to acutely deplete microglia and examined if this could lead to a disruption in diurnal temperature, metabolism, and activity measures. We also examined if differences in the physiological rhythms corresponded with changes in the expression of key circadian rhythm-regulating genes and proteins. Our data show that in the absence of microglia there is a pronounced disruption of diurnal rhythms in several domains consistent with a shift toward the inactive phase, in conjunction with changes in circadian rhythm-regulating genes and proteins. These data suggest microglia are involved in the regulation of circadian rhythms and indicate an exciting potential to manipulate these cells to improve disrupted circadian rhythms such as with shift-work or jet-lag.
Collapse
Affiliation(s)
- Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Tamara Dangel
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| | - Sajida Malik
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Simone N De Luca
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
30
|
Lalic T, Steponenaite A, Wei L, Vasudevan SR, Mathie A, Peirson SN, Lall GS, Cader MZ. TRESK is a key regulator of nocturnal suprachiasmatic nucleus dynamics and light adaptive responses. Nat Commun 2020; 11:4614. [PMID: 32929069 PMCID: PMC7490422 DOI: 10.1038/s41467-020-17978-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 07/24/2020] [Indexed: 11/25/2022] Open
Abstract
The suprachiasmatic nucleus (SCN) is a complex structure dependent upon multiple mechanisms to ensure rhythmic electrical activity that varies between day and night, to determine circadian adaptation and behaviours. SCN neurons are exposed to glutamate from multiple sources including from the retino-hypothalamic tract and from astrocytes. However, the mechanism preventing inappropriate post-synaptic glutamatergic effects is unexplored and unknown. Unexpectedly we discovered that TRESK, a calcium regulated two-pore potassium channel, plays a crucial role in this system. We propose that glutamate activates TRESK through NMDA and AMPA mediated calcium influx and calcineurin activation to then oppose further membrane depolarisation and rising intracellular calcium. Hence, in the absence of TRESK, glutamatergic activity is unregulated leading to membrane depolarisation, increased nocturnal SCN firing, inverted basal calcium levels and impaired sensitivity in light induced phase delays. Our data reveals TRESK plays an essential part in SCN regulatory mechanisms and light induced adaptive behaviours. The suprachiasmatic nucleus (SCN) ensures rhythmic electrical activity that varies between day and night to determine circadian behaviours. The authors show that TRESK channels provide a feedback mechanism to maintain the SCN in the appropriate state for nocturnal light-induced behavioural changes.
Collapse
Affiliation(s)
- Tatjana Lalic
- Translational Molecular Neuroscience Group, Weatherall Institute of Molecular Medicine, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| | - Aiste Steponenaite
- Medway School of Pharmacy, University of Kent and University of Greenwich, Anson Building, Central Avenue, Chatham, Kent, ME4 4TB, UK
| | - Liting Wei
- Translational Molecular Neuroscience Group, Weatherall Institute of Molecular Medicine, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Alistair Mathie
- Medway School of Pharmacy, University of Kent and University of Greenwich, Anson Building, Central Avenue, Chatham, Kent, ME4 4TB, UK
| | - Stuart N Peirson
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Gurprit S Lall
- Medway School of Pharmacy, University of Kent and University of Greenwich, Anson Building, Central Avenue, Chatham, Kent, ME4 4TB, UK.
| | - M Zameel Cader
- Translational Molecular Neuroscience Group, Weatherall Institute of Molecular Medicine, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| |
Collapse
|
31
|
Hartsock MJ, Spencer RL. Memory and the circadian system: Identifying candidate mechanisms by which local clocks in the brain may regulate synaptic plasticity. Neurosci Biobehav Rev 2020; 118:134-162. [PMID: 32712278 DOI: 10.1016/j.neubiorev.2020.07.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 07/14/2020] [Accepted: 07/18/2020] [Indexed: 12/11/2022]
Abstract
The circadian system is an endogenous biological network responsible for coordinating near-24-h cycles in behavior and physiology with daily timing cues from the external environment. In this review, we explore how the circadian system regulates memory formation, retention, and recall. Circadian rhythms in these memory processes may arise through several endogenous pathways, and recent work highlights the importance of genetic timekeepers found locally within tissues, called local clocks. We evaluate the circadian memory literature for evidence of local clock involvement in memory, identifying potential nodes for direct interactions between local clock components and mechanisms of synaptic plasticity. Our discussion illustrates how local clocks may pervasively modulate neuronal plastic capacity, a phenomenon that we designate here as circadian metaplasticity. We suggest that this function of local clocks supports the temporal optimization of memory processes, illuminating the potential for circadian therapeutic strategies in the prevention and treatment of memory impairment.
Collapse
Affiliation(s)
- Matthew J Hartsock
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado 80309, United States.
| | - Robert L Spencer
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado 80309, United States.
| |
Collapse
|
32
|
Re CJ, Batterman AI, Gerstner JR, Buono RJ, Ferraro TN. The Molecular Genetic Interaction Between Circadian Rhythms and Susceptibility to Seizures and Epilepsy. Front Neurol 2020; 11:520. [PMID: 32714261 PMCID: PMC7344275 DOI: 10.3389/fneur.2020.00520] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/12/2020] [Indexed: 12/19/2022] Open
Abstract
Seizure patterns observed in patients with epilepsy suggest that circadian rhythms and sleep/wake mechanisms play some role in the disease. This review addresses key topics in the relationship between circadian rhythms and seizures in epilepsy. We present basic information on circadian biology, but focus on research studying the influence of both the time of day and the sleep/wake cycle as independent but related factors on the expression of seizures in epilepsy. We review studies investigating how seizures and epilepsy disrupt expression of core clock genes, and how disruption of clock mechanisms impacts seizures and the development of epilepsy. We focus on the overlap between mechanisms of circadian-associated changes in SCN neuronal excitability and mechanisms of epileptogenesis as a means of identifying key pathways and molecules that could represent new targets or strategies for epilepsy therapy. Finally, we review the concept of chronotherapy and provide a perspective regarding its application to patients with epilepsy based on their individual characteristics (i.e., being a “morning person” or a “night owl”). We conclude that better understanding of the relationship between circadian rhythms, neuronal excitability, and seizures will allow both the identification of new therapeutic targets for treating epilepsy as well as more effective treatment regimens using currently available pharmacological and non-pharmacological strategies.
Collapse
Affiliation(s)
- Christopher J Re
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| | - Alexander I Batterman
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| | - Jason R Gerstner
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Russell J Buono
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| | - Thomas N Ferraro
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| |
Collapse
|
33
|
An K, Zhao H, Miao Y, Xu Q, Li YF, Ma YQ, Shi YM, Shen JW, Meng JJ, Yao YG, Zhang Z, Chen JT, Bao J, Zhang M, Xue T. A circadian rhythm-gated subcortical pathway for nighttime-light-induced depressive-like behaviors in mice. Nat Neurosci 2020; 23:869-880. [PMID: 32483349 DOI: 10.1038/s41593-020-0640-8] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/10/2020] [Indexed: 12/11/2022]
Abstract
Besides generating vision, light modulates various physiological functions, including mood. While light therapy applied in the daytime is known to have anti-depressive properties, excessive light exposure at night has been reportedly associated with depressive symptoms. The neural mechanisms underlying this day-night difference in the effects of light are unknown. Using a light-at-night (LAN) paradigm in mice, we showed that LAN induced depressive-like behaviors without disturbing the circadian rhythm. This effect was mediated by a neural pathway from retinal melanopsin-expressing ganglion cells to the dorsal perihabenular nucleus (dpHb) to the nucleus accumbens (NAc). Importantly, the dpHb was gated by the circadian rhythm, being more excitable at night than during the day. This indicates that the ipRGC→dpHb→NAc pathway preferentially conducts light signals at night, thereby mediating LAN-induced depressive-like behaviors. These findings may be relevant when considering the mental health effects of the prevalent nighttime illumination in the industrial world.
Collapse
Affiliation(s)
- Kai An
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huan Zhao
- College of Biology, Food and Environment, Hefei University, Hefei, China.
| | - Ying Miao
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Qi Xu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yu-Fei Li
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yu-Qian Ma
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yi-Ming Shi
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jia-Wei Shen
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jian-Jun Meng
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhi Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ju-Tao Chen
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jin Bao
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Mei Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tian Xue
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. .,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China. .,Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
34
|
Abstract
Ca2+- and voltage-gated K+ channels of large conductance (BK channels) are expressed in a diverse variety of both excitable and inexcitable cells, with functional properties presumably uniquely calibrated for the cells in which they are found. Although some diversity in BK channel function, localization, and regulation apparently arises from cell-specific alternative splice variants of the single pore-forming α subunit ( KCa1.1, Kcnma1, Slo1) gene, two families of regulatory subunits, β and γ, define BK channels that span a diverse range of functional properties. We are just beginning to unravel the cell-specific, physiological roles served by BK channels of different subunit composition.
Collapse
Affiliation(s)
- Vivian Gonzalez-Perez
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| |
Collapse
|
35
|
Harvey JRM, Plante AE, Meredith AL. Ion Channels Controlling Circadian Rhythms in Suprachiasmatic Nucleus Excitability. Physiol Rev 2020; 100:1415-1454. [PMID: 32163720 DOI: 10.1152/physrev.00027.2019] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Animals synchronize to the environmental day-night cycle by means of an internal circadian clock in the brain. In mammals, this timekeeping mechanism is housed in the suprachiasmatic nucleus (SCN) of the hypothalamus and is entrained by light input from the retina. One output of the SCN is a neural code for circadian time, which arises from the collective activity of neurons within the SCN circuit and comprises two fundamental components: 1) periodic alterations in the spontaneous excitability of individual neurons that result in higher firing rates during the day and lower firing rates at night, and 2) synchronization of these cellular oscillations throughout the SCN. In this review, we summarize current evidence for the identity of ion channels in SCN neurons and the mechanisms by which they set the rhythmic parameters of the time code. During the day, voltage-dependent and independent Na+ and Ca2+ currents, as well as several K+ currents, contribute to increased membrane excitability and therefore higher firing frequency. At night, an increase in different K+ currents, including Ca2+-activated BK currents, contribute to membrane hyperpolarization and decreased firing. Layered on top of these intrinsically regulated changes in membrane excitability, more than a dozen neuromodulators influence action potential activity and rhythmicity in SCN neurons, facilitating both synchronization and plasticity of the neural code.
Collapse
Affiliation(s)
- Jenna R M Harvey
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Amber E Plante
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrea L Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
36
|
Ahuja AS, Rozen TD, Atwal PS. A sleep modulated Channelopathy: a novel CACNA1A pathogenic variant identified in episodic Ataxia type 2 and a potential link to sleep alleviated migraine. BMC Neurol 2019; 19:246. [PMID: 31640633 PMCID: PMC6806495 DOI: 10.1186/s12883-019-1491-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 10/01/2019] [Indexed: 11/25/2022] Open
Abstract
Background To describe a patient with sleep alleviated episodic ataxia type 2 with a novel CACNA1A pathogenic variant and provide a possible link to sleep responsive migraine. Case presentation A 26-year-old woman with recurrent attacks of dizziness, nausea, vomiting, ataxia and dysarthria presented for a possible diagnosis of vestibular migraine. Unique to her attacks was if she could fall asleep for as little as 15 min the spells would subside. If however she remained awake the attacks would continue unabated. A presumed diagnosis of episodic ataxia type 2 was made and she became attack free on acetazolamide without recurrence. Genetic testing demonstrated a novel pathogenic variant in CACNA1A on chromosome 19. This pathogenic variant has not been previously reported in the literature and is suggested to truncate the CACNA1A polypeptide by introducing a premature stop codon. Conclusion A case of episodic ataxia type 2 with a novel pathogenic variant in CACNA1A is described. Interestingly, the patient’s symptoms would completely alleviate with sleep which suggests a sleep modulated channelopathy. The mechanisms by which sleep could potentially alter this pathogenic variant are hypothesized. A potential link to sleep alleviated migraine is suggested. Further study of this novel pathogenic variant may help us understand not only how sleep can modulate episodic ataxia type 2, but also migraine.
Collapse
|
37
|
Clusin WT, Wu TH, Shi LF, Kao PN. Further studies of ion channels in the electroreceptor of the skate through deep sequencing, cloning and cross species comparisons. Gene 2019; 718:143989. [PMID: 31326551 DOI: 10.1016/j.gene.2019.143989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 06/23/2019] [Accepted: 07/17/2019] [Indexed: 11/25/2022]
Abstract
Our comparative studies seek to understand the structure and function of ion channels in cartilaginous fish that can detect very low voltage gradients in seawater. The principal channels of the electroreceptor include a calcium activated K channel whose α subunit is Kcnma1, and a voltage-dependent calcium channel, Cacna1d. It has also been suggested based on physiological and pharmacological evidence that a voltage-gated K channel is present in the basal membranes of the receptor cells which modulates synaptic transmitter release. Large conductance calcium-activated K channels (BK) are comprised of four α subunits, encoded by Kcnma1 and modulatory β subunits of the Kcnmb class. We recently cloned and published the skate Kcnma1 gene and most of Kcnmb4 using purified mRNA of homogenized electroreceptors. Bellono et al. have recently performed RNA sequencing (RNA-seq) on purified mRNA from skate electroreceptors and found several ion channels including Kcnma1. We searched the Bellono et al. RNA-seq repository for additional channels and subunits. Our most significant findings are the presence of two Shaker type voltage dependent K channel sequences which are grouped together as isoforms in the data repository. The larger of these is a skate ortholog of the voltage dependent fast potassium channel Kv1.1, which is expressed at appreciable levels. The second ortholog is similar to Kv1.5 but has fewer N-terminal amino acids than other species. The sequence for Kv1.5 in the skate is very strongly aligned with the recently reported sequence for potassium channels in the electroreceptors of the cat shark, S. retifer, which also modulate synaptic transmission. The latter channel was designated as Kv1.3 in the initial report, but we suggest that these channels are actually orthologs of each other, and that Kv1.5 is the prevailing designation. We also found a beta subunit sequence (Kcnab2) which may co-assemble with one or both of the voltage gated channels. The new channels and subunits were verified by RT-PCR and the Kv1.1 sequence was confirmed by cloning. We also searched the RNA-seq repository for accessory subunits of Kcnma1, and found a computer-generated assembly that contained a complete sequence of its β subunit, Kcnmb2. Skate Kcnmb2 has a total of 279 amino acids, with 51 novel amino acids at the N-terminus which may play a specific physiological role. This sequence was confirmed by PCR and cloning. However, skate Kcnmb2 is expressed at low levels in the electroreceptor compared to Kcnma1 and skate Kcnmb1 is absent. The evolutionary origin of the newly described K channels and their subunits was studied by alignments with mammalian sequences, including human, and also those in related fish: the whale shark (R. typus), the ghost shark (C.milii), and (S. retifer). There are also orthologous K channels of the lamprey, which has electroreceptors. Tree building and bootstrap programs were used to confirm phylogenetic inferences. Further research should focus on the subcellular locations of these channels, their gating behavior, and the effects of accessory subunits on gating.
Collapse
Affiliation(s)
- William T Clusin
- Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, United States of America.
| | - Ting-Hsuan Wu
- Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, United States of America
| | - Ling-Fang Shi
- Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, United States of America
| | - Peter N Kao
- Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, United States of America
| |
Collapse
|
38
|
Smith P, Buhl E, Tsaneva-Atanasova K, Hodge JJL. Shaw and Shal voltage-gated potassium channels mediate circadian changes in Drosophila clock neuron excitability. J Physiol 2019; 597:5707-5722. [PMID: 31612994 DOI: 10.1113/jp278826] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/03/2019] [Indexed: 01/08/2023] Open
Abstract
As in mammals, Drosophila circadian clock neurons display rhythms of activity with higher action potential firing rates and more positive resting membrane potentials during the day. This rhythmic excitability has been widely observed but, critically, its regulation remains unresolved. We have characterized and modelled the changes underlying these electrical activity rhythms in the lateral ventral clock neurons (LNvs). We show that currents mediated by the voltage-gated potassium channels Shaw (Kv3) and Shal (Kv4) oscillate in a circadian manner. Disruption of these channels, by expression of dominant negative (DN) subunits, leads to changes in circadian locomotor activity and shortens lifespan. LNv whole-cell recordings then show that changes in Shaw and Shal currents drive changes in action potential firing rate and that these rhythms are abolished when the circadian molecular clock is stopped. A whole-cell biophysical model using Hodgkin-Huxley equations can recapitulate these changes in electrical activity. Based on this model and by using dynamic clamp to manipulate clock neurons directly, we can rescue the pharmacological block of Shaw and Shal, restore the firing rhythm, and thus demonstrate the critical importance of Shaw and Shal. Together, these findings point to a key role for Shaw and Shal in controlling circadian firing of clock neurons and show that changes in clock neuron currents can account for this. Moreover, with dynamic clamp we can switch the LNvs between morning-like and evening-like states of electrical activity. We conclude that changes in Shaw and Shal underlie the daily oscillation in LNv firing rate.
Collapse
Affiliation(s)
- Philip Smith
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Edgar Buhl
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Living Systems Institute, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - James J L Hodge
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| |
Collapse
|
39
|
Muheim CM, Spinnler A, Sartorius T, Dürr R, Huber R, Kabagema C, Ruth P, Brown SA. Dynamic- and Frequency-Specific Regulation of Sleep Oscillations by Cortical Potassium Channels. Curr Biol 2019; 29:2983-2992.e3. [PMID: 31474531 DOI: 10.1016/j.cub.2019.07.056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 06/15/2019] [Accepted: 07/17/2019] [Indexed: 10/26/2022]
Abstract
Primary electroencephalographic (EEG) features of sleep arise in part from thalamocortical neural assemblies, and cortical potassium channels have long been thought to play a critical role. We have exploited the regionally dynamic nature of sleep EEG to develop a novel screening strategy and used it to conduct an adeno-associated virus (AAV)-mediated RNAi screen for cellular roles of 31 different voltage-gated potassium channels in modulating cortical EEG features across the circadian sleep-wake cycle. Surprisingly, a majority of channels modified only electroencephalographic frequency bands characteristic of sleep, sometimes diurnally or even in specific vigilance states. Confirming our screen for one channel, we show that depletion of the KCa1.1 (or "BK") channel reduces EEG power in slow-wave sleep by slowing neuronal repolarization. Strikingly, this reduction completely abolishes transcriptomic changes between sleep and wake. Thus, our data establish an unexpected connection between transcription and EEG power controlled by specific potassium channels. We postulate that additive dynamic roles of individual potassium channels could integrate different influences upon sleep and wake within single neurons.
Collapse
Affiliation(s)
- Christine M Muheim
- Chronobiology and Sleep Research Group, Institute of Pharmacology and Toxicology, University of Zürich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Andrea Spinnler
- Chronobiology and Sleep Research Group, Institute of Pharmacology and Toxicology, University of Zürich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Tina Sartorius
- Institute of Pharmacy, Department of Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Auf der Morgenstelle 8, Tübingen 72076, Germany
| | - Roland Dürr
- Chronobiology and Sleep Research Group, Institute of Pharmacology and Toxicology, University of Zürich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Reto Huber
- University Children's Hospital Zurich, University of Zürich, Steinwiesstrasse 75, Zürich 8032, Switzerland
| | - Clement Kabagema
- Institute of Pharmacy, Department of Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Auf der Morgenstelle 8, Tübingen 72076, Germany
| | - Peter Ruth
- Institute of Pharmacy, Department of Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Auf der Morgenstelle 8, Tübingen 72076, Germany
| | - Steven A Brown
- Chronobiology and Sleep Research Group, Institute of Pharmacology and Toxicology, University of Zürich, Winterthurerstrasse 190, Zürich 8057, Switzerland.
| |
Collapse
|
40
|
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is remarkable. Despite numbering only about 10,000 neurons on each side of the third ventricle, the SCN is our principal circadian clock, directing the daily cycles of behaviour and physiology that set the tempo of our lives. When this nucleus is isolated in organotypic culture, its autonomous timing mechanism can persist indefinitely, with precision and robustness. The discovery of the cell-autonomous transcriptional and post-translational feedback loops that drive circadian activity in the SCN provided a powerful exemplar of the genetic specification of complex mammalian behaviours. However, the analysis of circadian time-keeping is moving beyond single cells. Technical and conceptual advances, including intersectional genetics, multidimensional imaging and network theory, are beginning to uncover the circuit-level mechanisms and emergent properties that make the SCN a uniquely precise and robust clock. However, much remains unknown about the SCN, not least the intrinsic properties of SCN neurons, its circuit topology and the neuronal computations that these circuits support. Moreover, the convention that the SCN is a neuronal clock has been overturned by the discovery that astrocytes are an integral part of the timepiece. As a test bed for examining the relationships between genes, cells and circuits in sculpting complex behaviours, the SCN continues to offer powerful lessons and opportunities for contemporary neuroscience.
Collapse
|
41
|
Colwell CS, Donlea J. Temporal Coding of Sleep. Cell 2019; 175:1177-1179. [PMID: 30445036 DOI: 10.1016/j.cell.2018.10.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In Drosophila, well-delineated circuits control circadian rhythms, but the electrophysiological patterns that occur within these circuits are not well understood. In this issue, Tabuchi et al. clarify the temporal coding within a circuit, linking patterns of neural activity to sleep behavior.
Collapse
Affiliation(s)
- Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Jeffrey Donlea
- Department of Neurobiology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
42
|
Bailey CS, Moldenhauer HJ, Park SM, Keros S, Meredith AL. KCNMA1-linked channelopathy. J Gen Physiol 2019; 151:1173-1189. [PMID: 31427379 PMCID: PMC6785733 DOI: 10.1085/jgp.201912457] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022] Open
Abstract
Bailey et al. review a new neurological channelopathy associated with KCNMA1, encoding the BK voltage- and Ca2+-activated K+ channel. KCNMA1 encodes the pore-forming α subunit of the “Big K+” (BK) large conductance calcium and voltage-activated K+ channel. BK channels are widely distributed across tissues, including both excitable and nonexcitable cells. Expression levels are highest in brain and muscle, where BK channels are critical regulators of neuronal excitability and muscle contractility. A global deletion in mouse (KCNMA1−/−) is viable but exhibits pathophysiology in many organ systems. Yet despite the important roles in animal models, the consequences of dysfunctional BK channels in humans are not well characterized. Here, we summarize 16 rare KCNMA1 mutations identified in 37 patients dating back to 2005, with an array of clinically defined pathological phenotypes collectively referred to as “KCNMA1-linked channelopathy.” These mutations encompass gain-of-function (GOF) and loss-of-function (LOF) alterations in BK channel activity, as well as several variants of unknown significance (VUS). Human KCNMA1 mutations are primarily associated with neurological conditions, including seizures, movement disorders, developmental delay, and intellectual disability. Due to the recent identification of additional patients, the spectrum of symptoms associated with KCNMA1 mutations has expanded but remains primarily defined by brain and muscle dysfunction. Emerging evidence suggests the functional BK channel alterations produced by different KCNMA1 alleles may associate with semi-distinct patient symptoms, such as paroxysmal nonkinesigenic dyskinesia (PNKD) with GOF and ataxia with LOF. However, due to the de novo origins for the majority of KCNMA1 mutations identified to date and the phenotypic variability exhibited by patients, additional evidence is required to establish causality in most cases. The symptomatic picture developing from patients with KCNMA1-linked channelopathy highlights the importance of better understanding the roles BK channels play in regulating cell excitability. Establishing causality between KCNMA1-linked BK channel dysfunction and specific patient symptoms may reveal new treatment approaches with the potential to increase therapeutic efficacy over current standard regimens.
Collapse
Affiliation(s)
- Cole S Bailey
- Dept. of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Hans J Moldenhauer
- Dept. of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Su Mi Park
- Dept. of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Sotirios Keros
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, SD
| | - Andrea L Meredith
- Dept. of Physiology, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
43
|
Smarr B, Cutler T, Loh DH, Kudo T, Kuljis D, Kriegsfeld L, Ghiani CA, Colwell CS. Circadian dysfunction in the Q175 model of Huntington's disease: Network analysis. J Neurosci Res 2019; 97:1606-1623. [PMID: 31359503 DOI: 10.1002/jnr.24505] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/29/2019] [Accepted: 07/16/2019] [Indexed: 12/19/2022]
Abstract
Disturbances in sleep/wake cycle are a common complaint of individuals with Huntington's disease (HD) and are displayed by HD mouse models. The underlying mechanisms, including the possible role of the circadian timing system, have been the topic of a number of recent studies. The (z)Q175 mouse is a knock-in model in which the human exon 1 sequence of the huntingtin gene is inserted into the mouse DNA with approximately 190 CAG repeats. Among the numerous models available, the heterozygous Q175 offers strong construct validity with a single copy of the mutation, genetic precision of the insertion and control of mutation copy number. In this review, we will summarize the evidence that this model exhibits disrupted diurnal and circadian rhythms in locomotor activity. We found overwhelming evidence for autonomic dysfunction including blunted daily rhythms in heart rate and core body temperature (CBT), reduced heart rate variability, and almost a complete failure of the sympathetic arm of the autonomic nervous system to function during the baroreceptor reflex. Mechanistically, the Q175 mouse model exhibits deficits in the neural output of the central circadian clock, the suprachiasmatic nucleus along with an enhancement of at least one type of potassium current in these neurons. Finally, we report a novel network analysis examining the phase coherence between activity, CBT, and cardiovascular measures. Such analyses found that even young Q175 mutants (heterozygous or homozygous) show coherence degradation, and suggests that loss of phase coherence is a variable that should be considered as a possible biomarker for HD.
Collapse
Affiliation(s)
- Benjamin Smarr
- Department of Psychology, University of California Berkeley, Berkeley, California.,Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California
| | - Tamara Cutler
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Dawn H Loh
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Takashi Kudo
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Dika Kuljis
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Lance Kriegsfeld
- Department of Psychology, University of California Berkeley, Berkeley, California.,Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California
| | - Cristina A Ghiani
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Department of Pathology, University of California Los Angeles, Los Angeles, California.,Laboratory Medicine, University of California Los Angeles, Los Angeles, California
| | - Christopher S Colwell
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
44
|
An Epilepsy-Associated KCNT1 Mutation Enhances Excitability of Human iPSC-Derived Neurons by Increasing Slack K Na Currents. J Neurosci 2019; 39:7438-7449. [PMID: 31350261 DOI: 10.1523/jneurosci.1628-18.2019] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/08/2019] [Accepted: 07/17/2019] [Indexed: 12/26/2022] Open
Abstract
Mutations in the KCNT1 (Slack, KNa1.1) sodium-activated potassium channel produce severe epileptic encephalopathies. Expression in heterologous systems has shown that the disease-causing mutations give rise to channels that have increased current amplitude. It is not known, however, whether such gain of function occurs in human neurons, nor whether such increased KNa current is expected to suppress or increase the excitability of cortical neurons. Using genetically engineered human induced pluripotent stem cell (iPSC)-derived neurons, we have now found that sodium-dependent potassium currents are increased several-fold in neurons bearing a homozygous P924L mutation. In current-clamp recordings, the increased KNa current in neurons with the P924L mutation acts to shorten the duration of action potentials and to increase the amplitude of the afterhyperpolarization that follows each action potential. Strikingly, the number of action potentials that were evoked by depolarizing currents as well as maximal firing rates were increased in neurons expressing the mutant channel. In networks of spontaneously active neurons, the mean firing rate, the occurrence of rapid bursts of action potentials, and the intensity of firing during the burst were all increased in neurons with the P924L Slack mutation. The feasibility of an increased KNa current to increase firing rates independent of any compensatory changes was validated by numerical simulations. Our findings indicate that gain-of-function in Slack KNa channels causes hyperexcitability in both isolated neurons and in neural networks and occurs by a cell-autonomous mechanism that does not require network interactions.SIGNIFICANCE STATEMENT KCNT1 mutations lead to severe epileptic encephalopathies for which there are no effective treatments. This study is the first demonstration that a KCNT1 mutation increases the Slack current in neurons. It also provides the first explanation for how this increased potassium current induces hyperexcitability, which could be the underlining factor causing seizures.
Collapse
|
45
|
McNally BA, Plante AE, Meredith AL. Diurnal properties of voltage-gated Ca 2+ currents in suprachiasmatic nucleus and roles in action potential firing. J Physiol 2019; 598:1775-1790. [PMID: 31177540 DOI: 10.1113/jp278327] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 06/06/2019] [Indexed: 02/04/2023] Open
Abstract
KEY POINTS Circadian oscillations in spontaneous action potential firing in the suprachiasmatic nucleus (SCN) translate time-of-day throughout the mammalian brain. The ion channels that regulate the circadian pattern of SCN firing have not been comprehensively identified. Ca2+ channels regulate action potential activity across many types of excitable cells, and the activity of L-, N-, P/Q- and R-type channels are required for normal daytime firing frequency in SCN neurons and circuit rhythms. Only the L-type Ca2+ current exhibits a day versus night difference in current magnitude, providing insight into the mechanism that produces rhythmic action potential firing in SCN. ABSTRACT The mammalian circadian clock encodes time via rhythmic action potential activity in the suprachiasmatic nucleus (SCN) of the hypothalamus, which governs daily rhythms in physiology and behaviour. SCN neurons exhibit 24 h oscillations in spontaneous firing, with higher firing during day compared to night. Several ionic currents have been identified that regulate SCN firing, including voltage-gated Ca2+ currents, but the circadian regulation of distinct voltage-gated Ca2+ channel (VGCC) components has not been comprehensively addressed. In this study, whole-cell L- (nimodipine-sensitive), N- and P/Q- (ω-agatoxin IVA, ω-conotoxin GVIA, ω-conotoxin MVIIC-sensitive), R- (Ni2+ -sensitive) and T-type (TTA-P2-sensitive) currents were recorded from day and night SCN slices. Using standard voltage protocols, Ni2+ -sensitive currents comprised the largest proportion of total VGCC current, followed by nimodipine-, ω-agatoxin IVA-, ω-conotoxin GVIA- and TTA-P2-sensitive currents. Only the nimodipine-sensitive current exhibited a diurnal difference in magnitude, with daytime current larger than night. No diurnal variation was observed for the other Ca2+ current subtypes. The difference in nimodipine-sensitive current was due to larger peak current activated during the day, not differences in inactivation, and was eliminated by Bay K8644. Blocking L-type channels decreased firing selectively during the day, consistent with higher current magnitudes, and reduced SCN circuit rhythmicity recorded by multi-electrode arrays. Yet blocking N-, P/Q- and R-type channels also decreased daytime firing, with little effect at night, and decreased circuit rhythmicity. These data identify a unique diurnal regulation of L-type current among the major VGCC subtypes in SCN neurons, but also reveal that diurnal modulation is not required for time-of-day-specific effects on firing and circuit rhythmicity.
Collapse
Affiliation(s)
- Beth A McNally
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Amber E Plante
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Andrea L Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
46
|
McCarthy MJ. Missing a beat: assessment of circadian rhythm abnormalities in bipolar disorder in the genomic era. Psychiatr Genet 2019; 29:29-36. [PMID: 30516584 DOI: 10.1097/ypg.0000000000000215] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Circadian rhythm abnormalities have been recognized as a central feature of bipolar disorder (BD) but a coherent biological explanation for them remains lacking. Using genetic mutation of 'clock genes', robust animal models of mania and depression have been developed that elucidate key aspects of circadian rhythms and the circadian clock-mood connection. However, translation of this knowledge into humans remains incomplete. In recent years, very large genome-wide association studies (GWAS) have been conducted and the genetic underpinnings of BD are beginning to emerge. However, these genetic studies in BD do not match well with the evidence from animal studies that implicate the circadian clock in mood regulation. Even larger GWAS have been conducted for circadian phenotypes including chronotype, rhythm amplitude, sleep duration, and insomnia. These studies have identified a diverse set of associated genes, including a minority with previously well-characterized functions in the circadian clock. Taken together, the data from recent GWAS of BD and circadian phenotypes indicate that the genetic organization of the circadian clock, both in health and in BD is complex. The findings from GWAS elucidate potentially novel circadian mechanism that may be partly distinct from those identified in animal models. Pleiotropy, epistasis and nongenetic factors may play important roles in regulating circadian rhythms, some of which may underlie circadian rhythm disturbances in BD.
Collapse
Affiliation(s)
- Michael J McCarthy
- Department of Psychiatry, Center for Circadian Biology, VA San Diego Healthcare System, University of California San Diego, San Diego, California, USA
| |
Collapse
|
47
|
Abstract
Ca2+- and voltage-gated K+ channels of large conductance (BK channels) are expressed in a diverse variety of both excitable and inexcitable cells, with functional properties presumably uniquely calibrated for the cells in which they are found. Although some diversity in BK channel function, localization, and regulation apparently arises from cell-specific alternative splice variants of the single pore-forming α subunit ( KCa1.1, Kcnma1, Slo1) gene, two families of regulatory subunits, β and γ, define BK channels that span a diverse range of functional properties. We are just beginning to unravel the cell-specific, physiological roles served by BK channels of different subunit composition.
Collapse
Affiliation(s)
- Vivian Gonzalez-Perez
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| |
Collapse
|
48
|
Paul JR, Davis JA, Goode LK, Becker BK, Fusilier A, Meador-Woodruff A, Gamble KL. Circadian regulation of membrane physiology in neural oscillators throughout the brain. Eur J Neurosci 2019; 51:109-138. [PMID: 30633846 DOI: 10.1111/ejn.14343] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 12/21/2022]
Abstract
Twenty-four-hour rhythmicity in physiology and behavior are driven by changes in neurophysiological activity that vary across the light-dark and rest-activity cycle. Although this neural code is most prominent in neurons of the primary circadian pacemaker in the suprachiasmatic nucleus (SCN) of the hypothalamus, there are many other regions in the brain where region-specific function and behavioral rhythmicity may be encoded by changes in electrical properties of those neurons. In this review, we explore the existing evidence for molecular clocks and/or neurophysiological rhythms (i.e., 24 hr) in brain regions outside the SCN. In addition, we highlight the brain regions that are ripe for future investigation into the critical role of circadian rhythmicity for local oscillators. For example, the cerebellum expresses rhythmicity in over 2,000 gene transcripts, and yet we know very little about how circadian regulation drives 24-hr changes in the neural coding responsible for motor coordination. Finally, we conclude with a discussion of how our understanding of circadian regulation of electrical properties may yield insight into disease mechanisms which may lead to novel chronotherapeutic strategies in the future.
Collapse
Affiliation(s)
- Jodi R Paul
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jennifer A Davis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lacy K Goode
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bryan K Becker
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Allison Fusilier
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Aidan Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
49
|
Bukiya AN, Dopico AM. Regulation of BK Channel Activity by Cholesterol and Its Derivatives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1115:53-75. [DOI: 10.1007/978-3-030-04278-3_3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
50
|
Yamada Y, Prosser RA. Copper in the suprachiasmatic circadian clock: A possible link between multiple circadian oscillators. Eur J Neurosci 2018; 51:47-70. [PMID: 30269387 DOI: 10.1111/ejn.14181] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 09/05/2018] [Accepted: 09/17/2018] [Indexed: 01/07/2023]
Abstract
The mammalian circadian clock in the suprachiasmatic nucleus (SCN) is very robust, able to coordinate our daily physiological and behavioral rhythms with exquisite accuracy. Simultaneously, the SCN clock is highly sensitive to environmental timing cues such as the solar cycle. This duality of resiliency and sensitivity may be sustained in part by a complex intertwining of three cellular oscillators: transcription/translation, metabolic/redox, and membrane excitability. We suggest here that one of the links connecting these oscillators may be forged from copper (Cu). Cellular Cu levels are highly regulated in the brain and peripherally, and Cu affects cellular metabolism, redox state, cell signaling, and transcription. We have shown that both Cu chelation and application induce nighttime phase shifts of the SCN clock in vitro and that these treatments affect glutamate, N-methyl-D-aspartate receptor, and associated signaling processes differently. More recently we found that Cu induces mitogen-activated protein kinase-dependent phase shifts, while the mechanisms by which Cu removal induces phase shifts remain unclear. Lastly, we have found that two Cu transporters are expressed in the SCN, and that one of these transporters (ATP7A) exhibits a day/night rhythm. Our results suggest that Cu homeostasis is tightly regulated in the SCN, and that changes in Cu levels may serve as a time cue for the circadian clock. We discuss these findings in light of the existing literature and current models of multiple coupled circadian oscillators in the SCN.
Collapse
Affiliation(s)
- Yukihiro Yamada
- Department of Biochemistry & Cellular and Molecular Biology, NeuroNET Research Center, University of Tennessee, Knoxville, Tennessee
| | - Rebecca A Prosser
- Department of Biochemistry & Cellular and Molecular Biology, NeuroNET Research Center, University of Tennessee, Knoxville, Tennessee
| |
Collapse
|