1
|
Helfrich-Förster C, Reinhard N. Mutual coupling of neurons in the circadian master clock: What we can learn from fruit flies. Neurobiol Sleep Circadian Rhythms 2025; 18:100112. [PMID: 39906412 PMCID: PMC11791320 DOI: 10.1016/j.nbscr.2025.100112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025] Open
Abstract
Circadian master clocks in the brain consist of multiple neurons that are organized into populations with different morphology, physiology, and neuromessenger content and presumably different functions. In most animals, these master clocks are distributed bilaterally, located in close proximity to the visual system, and synchronized by the eyes with the light-dark cycles of the environment. In mammals and cockroaches, each of the two master clocks consists of a core region that receives information from the eyes and a shell region from which most of the output projections originate, whereas in flies and several other insects, the master clocks are distributed in lateral and dorsal brain regions. In all cases, morning and evening clock neurons seem to exist, and the communication between them and other populations of clock neurons, as well as the connection across the two brain hemispheres, is a prerequisite for normal rhythmic function. Phenomena such as rhythm splitting, and internal desynchronization are caused by the "decoupling" of the master clocks in the two brain hemispheres or by the decoupling of certain clock neurons within the master clock of one brain hemisphere. Since the master clocks in flies contain relatively few neurons that are well characterized at the individual level, the fly is particularly well suited to study the communication between individual clock neurons. Here, we review the organization of the bilateral master clocks in the fly brain, with a focus on synaptic and paracrine connections between the multiple clock neurons, in comparison with other insects and mammals.
Collapse
Affiliation(s)
- Charlotte Helfrich-Förster
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Nils Reinhard
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| |
Collapse
|
2
|
Su K, Zeng D, Zhang W, Peng F, Cui B, Liu Q. Integrating cancer medicine into metabolic rhythms. Trends Endocrinol Metab 2025:S1043-2760(25)00053-0. [PMID: 40199622 DOI: 10.1016/j.tem.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025]
Abstract
Circadian rhythms are cell-intrinsic time-keeping mechanisms that allow organisms to adapt to 24-h environmental changes, ensuring coordinated physiological functions by aligning internal metabolic oscillations with external timing cues. Disruption of daily metabolic rhythms is associated with pathological events such as cancer development, yet the mechanisms by which perturbed metabolic rhythms contribute to tumorigenesis remain unclear. Herein we review how circadian clocks drive balanced rhythmic metabolism which in turn governs physiological functions of locomotor, immune, and neuroendocrine systems. Misaligned metabolic rhythms cause pathological states which further drive cancer initiation, progression, and metastasis. Restoring the balance of metabolic rhythms with chemical, hormonal, and behavioral interventions serves as a promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Keyu Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Deshun Zeng
- State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Weiru Zhang
- State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China.
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
3
|
Song Z, Yan M, Zhang S, Hu B, Qing X, Shao Z, Chen S, Lv X, Liu H. Implications of circadian disruption on intervertebral disc degeneration: The mediating role of sympathetic nervous system. Ageing Res Rev 2025; 104:102633. [PMID: 39701186 DOI: 10.1016/j.arr.2024.102633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024]
Abstract
The circadian clock orchestrates a broad spectrum of physiological processes, crucially modulating human biology across an approximate 24-hour cycle. The circadian disturbances precipitated by modern lifestyle contribute to the occurrence of low back pain (LBP), mainly ascribed to intervertebral disc degeneration (IVDD). The intervertebral disc (IVD) exhibits rhythmic physiological behaviors, with fluctuations in osmotic pressure and hydration levels that synchronized with the diurnal cycle of activity and rest. Over recent decades, advanced molecular biology techniques have shed light on the association between circadian molecules and IVD homeostasis. The complex interplay between circadian rhythm disruption and IVDD is becoming increasingly evident, with the sympathetic nervous system (SNS) emerging as a potential mediator. Synchronized with circadian rhythm through suprachiasmatic nucleus, the SNS regulates diverse physiological functions and metabolic processes, profoundly influences the structural and functional integrity of the IVD. This review synthesizes the current understanding of circadian regulation and sympathetic innervation of the IVD, highlighting advancements in the comprehension of their interactions. We elucidate the impact of circadian system on the physiological functions of IVD through the SNS, advocating for the adoption of chronotherapy as a brand-new and effective strategy to ameliorate IVDD and alleviate LBP.
Collapse
Affiliation(s)
- Zongmian Song
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Miaoheng Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shuo Zhang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Binwu Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiangcheng Qing
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Xiao Lv
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
4
|
Ambroziak W, Nencini S, Pohle J, Zuza K, Pino G, Lundh S, Araujo-Sousa C, Goetz LIL, Schrenk-Siemens K, Manoj G, Herrera MA, Acuna C, Siemens J. Thermally induced neuronal plasticity in the hypothalamus mediates heat tolerance. Nat Neurosci 2025; 28:346-360. [PMID: 39653806 PMCID: PMC11802458 DOI: 10.1038/s41593-024-01830-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/25/2024] [Indexed: 12/19/2024]
Abstract
Heat acclimation is an adaptive process that improves physiological performance and supports survival in the face of increasing environmental temperatures, but the underlying mechanisms are not well understood. Here we identified a discrete group of neurons in the mouse hypothalamic preoptic area (POA) that rheostatically increase their activity over the course of heat acclimation, a property required for mice to become heat tolerant. In non-acclimated mice, peripheral thermoafferent pathways via the parabrachial nucleus activate POA neurons and mediate acute heat-defense mechanisms. However, long-term heat exposure promotes the POA neurons to gain intrinsically warm-sensitive activity, independent of thermoafferent parabrachial input. This newly gained cell-autonomous warm sensitivity is required to recruit peripheral heat tolerance mechanisms in acclimated animals. This pacemaker-like, warm-sensitive activity is driven by a combination of increased sodium leak current and enhanced utilization of the NaV1.3 ion channel. We propose that this salient neuronal plasticity mechanism adaptively drives acclimation to promote heat tolerance.
Collapse
Affiliation(s)
- Wojciech Ambroziak
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Department of Translational Disease Understanding, Grünenthal GmbH, Aachen, Germany
| | - Sara Nencini
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Istituto Italiano di Tecnologia, Genoa, Italy
| | - Jörg Pohle
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Department of Translational Disease Understanding, Grünenthal GmbH, Aachen, Germany
| | - Kristina Zuza
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Gabriela Pino
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Sofia Lundh
- Department of Pathology and Imaging, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Carolina Araujo-Sousa
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Larissa I L Goetz
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | | | - Gokul Manoj
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Mildred A Herrera
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Claudio Acuna
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Jan Siemens
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| |
Collapse
|
5
|
Halabian A, Radahmadi M. The neurobiological mechanisms of photoperiod impact on brain functions: a comprehensive review. Rev Neurosci 2024; 35:933-958. [PMID: 39520288 DOI: 10.1515/revneuro-2024-0006] [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: 01/14/2024] [Accepted: 06/19/2024] [Indexed: 11/16/2024]
Abstract
Variations in day length, or photoperiodism, whether natural or artificial light, significantly impact biological, physiological, and behavioral processes within the brain. Both natural and artificial light sources are environmental factors that significantly influence brain functions and mental well-being. Photoperiodism is a phenomenon, occurring either over a 24 h cycle or seasonally and denotes all biological responses of humans and animals to these fluctuations in day and night length. Conversely, artificial light occurrence refers to the presence of light during nighttime hours and/or its absence during the daytime (unnaturally long and short days, respectively). Light at night, which is a form of light pollution, is prevalent in many societies, especially common in certain emergency occupations. Moreover, individuals with certain mental disorders, such as depression, often exhibit a preference for darkness over daytime light. Nevertheless, disturbances in light patterns can have negative consequences, impacting brain performance through similar mechanisms albeit with varying degrees of severity. Furthermore, changes in day length lead to alterations in the activity of receptors, proteins, ion channels, and molecular signaling pathways, all of which can impact brain health. This review aims to summarize the mechanisms by which day length influences brain functions through neural circuits, hormonal systems, neurochemical processes, cellular activity, and even molecular signaling pathways.
Collapse
Affiliation(s)
- Alireza Halabian
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western, Ontario, N6A 3K7 London, ON, Canada
| | - Maryam Radahmadi
- Department of Physiology, School of Medicine, 48455 Isfahan University of Medical Sciences , 81746-73461 Isfahan, Iran
| |
Collapse
|
6
|
Wegner S, Belle MDC, Chang P, Hughes ATL, Conibear AE, Muir C, Samuels RE, Piggins HD. Loss of neuropeptide signalling alters temporal expression of mouse suprachiasmatic neuronal state and excitability. Eur J Neurosci 2024; 60:6617-6633. [PMID: 39551976 PMCID: PMC11612845 DOI: 10.1111/ejn.16590] [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: 02/19/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Individual neurons of the hypothalamic suprachiasmatic nuclei (SCN) contain an intracellular molecular clock that drives these neurons to exhibit day-night variation in excitability. The neuropeptide vasoactive intestinal polypeptide (VIP) and its cognate receptor, VPAC2, are synthesized by SCN neurons and this intercellular VIP-VPAC2 receptor signal facilitates coordination of SCN neuronal activity and timekeeping. How the loss of VPAC2 receptor signalling affects the electrophysiological properties and states of SCN neurons as well as their responses to excitatory inputs is unclear. Here we used patch-clamp electrophysiology and made recordings of SCN neurons in brain slices prepared from transgenic animals that do not express VPAC2 receptors (Vipr2-/- mice) as well as animals that do (Vipr2+/+ mice). We report that while Vipr2+/+ neurons exhibit coordinated day-night variation in their electrical state, Vipr2-/- neurons lack this and instead manifest a range of states during both day and night. Further, at the population level, Vipr2+/+ neurons vary the membrane threshold potential at which they start to fire action potentials from day to night, while Vipr2-/- neurons do not. We provide evidence that Vipr2-/- neurons lack a component of voltage-gated sodium currents that contribute to SCN neuronal excitability. Moreover, we determine that this aberrant temporal control of neuronal state and excitability alters neuronal responses to a neurochemical mimic of the light-input pathway to the SCN. These results highlight the critical role VIP-VPAC2 receptor signalling plays in the temporal expression of individual neuronal states as well as appropriate ensemble activity and input gating of the SCN neural network.
Collapse
Affiliation(s)
- Sven Wegner
- Faculty of Biology, Medicine, and HealthUniversity of ManchesterManchesterUK
| | - Mino D. C. Belle
- Faculty of Biology, Medicine, and HealthUniversity of ManchesterManchesterUK
| | - Pi‐Shan Chang
- School of Physiology, Pharmacology, and NeuroscienceUniversity of BristolBristolUK
| | - Alun T. L. Hughes
- Faculty of Biology, Medicine, and HealthUniversity of ManchesterManchesterUK
- School of Biological and Environmental ScienceLiverpool John Moores UniversityLiverpoolUK
| | | | - Charlotte Muir
- School of Physiology, Pharmacology, and NeuroscienceUniversity of BristolBristolUK
| | - Rayna E. Samuels
- Faculty of Biology, Medicine, and HealthUniversity of ManchesterManchesterUK
| | - Hugh D. Piggins
- Faculty of Biology, Medicine, and HealthUniversity of ManchesterManchesterUK
- School of Physiology, Pharmacology, and NeuroscienceUniversity of BristolBristolUK
| |
Collapse
|
7
|
Cervera J, Manzanares JA, Levin M, Mafe S. Oscillatory phenomena in electrophysiological networks: The coupling between cell bioelectricity and transcription. Comput Biol Med 2024; 180:108964. [PMID: 39106669 DOI: 10.1016/j.compbiomed.2024.108964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/04/2024] [Accepted: 07/27/2024] [Indexed: 08/09/2024]
Abstract
Morphogenetic regulation during embryogenesis and regeneration rely on information transfer and coordination between different regions. Here, we explore theoretically the coupling between bioelectrical and transcriptional oscillations at the individual cell and multicellular levels. The simulations, based on a set of ion channels and intercellular gap junctions, show that bioelectrical and transcriptional waves can electrophysiologically couple distant regions of a model network in phase and antiphase oscillatory states that include synchronization phenomena. In this way, different multicellular regionalizations can be encoded by cell potentials that oscillate between depolarized and polarized states, thus allowing a spatio-temporal coding. Because the electric potential patterns characteristic of development and regeneration are correlated with the spatial distributions of signaling ions and molecules, bioelectricity can act as a template for slow biochemical signals following a hierarchy of experimental times. In particular, bioelectrical gradients that couple cell potentials to transcription rates give to each single cell a rough idea of its location in the multicellular ensemble, thus controlling local differentiation processes that switch on and off crucial parts of the genome.
Collapse
Affiliation(s)
- Javier Cervera
- Dept. Termodinàmica, Facultat de Física, Universitat de València, 46100, Burjassot, Spain.
| | - José A Manzanares
- Dept. Termodinàmica, Facultat de Física, Universitat de València, 46100, Burjassot, Spain
| | - Michael Levin
- Dept. of Biology, Tufts University, Medford, MA, 02155, USA; Allen Discovery Center at Tufts University, Medford, MA, 02155, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Salvador Mafe
- Dept. Termodinàmica, Facultat de Física, Universitat de València, 46100, Burjassot, Spain; Allen Discovery Center at Tufts University, Medford, MA, 02155, USA
| |
Collapse
|
8
|
Liang X, Liang X, Zhao Y, Ding Y, Zhu X, Zhou J, Qiu J, Shen X, Xie W. Dysregulation of the Suprachiasmatic Nucleus Disturbs the Circadian Rhythm and Aggravates Epileptic Seizures by Inducing Hippocampal GABAergic Dysfunction in C57BL/6 Mice. J Pineal Res 2024; 76:e12993. [PMID: 39054842 DOI: 10.1111/jpi.12993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
The interplay between circadian rhythms and epilepsy has gained increasing attention. The suprachiasmatic nucleus (SCN), which acts as the master circadian pacemaker, regulates physiological and behavioral rhythms through its complex neural networks. However, the exact role of the SCN and its Bmal1 gene in the development of epilepsy remains unclear. In this study, we utilized a lithium-pilocarpine model to induce epilepsy in mice and simulated circadian disturbances by creating lesions in the SCN and specifically knocking out the Bmal1 gene in the SCN neurons. We observed that the pilocarpine-induced epileptic mice experienced increased daytime seizure frequency, irregular oscillations in core body temperature, and circadian gene alterations in both the SCN and the hippocampus. Additionally, there was enhanced activation of GABAergic projections from the SCN to the hippocampus. Notably, SCN lesions intensified seizure activity, concomitant with hippocampal neuronal damage and GABAergic signaling impairment. Further analyses using the Gene Expression Omnibus database and gene set enrichment analysis indicated reduced Bmal1 expression in patients with medial temporal lobe epilepsy, potentially affecting GABA receptor pathways. Targeted deletion of Bmal1 in SCN neurons exacerbated seizures and pathology in epilepsy, as well as diminished hippocampal GABAergic efficacy. These results underscore the crucial role of the SCN in modulating circadian rhythms and GABAergic function in the hippocampus, aggravating the severity of seizures. This study provides significant insights into how circadian rhythm disturbances can influence neuronal dysfunction and epilepsy, highlighting the therapeutic potential of targeting SCN and the Bmal1 gene within it in epilepsy management.
Collapse
Affiliation(s)
- Xiaoshan Liang
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaotao Liang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yunyan Zhao
- Department of Critical Care Medicine, The Afflliated Traditional Chinese Medicine Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuewen Ding
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoyu Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jieli Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jing Qiu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoqin Shen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wei Xie
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Masliukov PM. Functional properties of aged hypothalamic cells. VITAMINS AND HORMONES 2024; 127:207-243. [PMID: 39864942 DOI: 10.1016/bs.vh.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The hypothalamus, in addition to controlling the main body's vital functions, is also involved in aging regulation. The aging process in the hypothalamus is accompanied by disturbed intracellular pathways, including Ca2+ signaling and neuronal excitability in the brain. Intrinsic electrophysiological properties of individual neurons and synaptic transmission between cells is disrupted in the central nervous system of old animals. However, changes in neuronal excitability and excitation/inhibition balance with aging are specific to the type of neurons, brain region, and species. Glia-neuron interactions play a significant role in the brain and undergo remodeling accompanied by advanced loss of function with aging. In the current review, I have summarized the current understanding of the changes in the brain and especially in the hypothalamus with aging.
Collapse
Affiliation(s)
- Petr M Masliukov
- Department Normal Physiology, Yaroslavl State Medical University, Yaroslavl, Russia.
| |
Collapse
|
10
|
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
|
11
|
Rodan AR. Circadian Rhythm Regulation by Pacemaker Neuron Chloride Oscillation in Flies. Physiology (Bethesda) 2024; 39:0. [PMID: 38411570 PMCID: PMC11368518 DOI: 10.1152/physiol.00006.2024] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/28/2024] Open
Abstract
Circadian rhythms in physiology and behavior sync organisms to external environmental cycles. Here, circadian oscillation in intracellular chloride in central pacemaker neurons of the fly, Drosophila melanogaster, is reviewed. Intracellular chloride links SLC12 cation-coupled chloride transporter function with kinase signaling and the regulation of inwardly rectifying potassium channels.
Collapse
Affiliation(s)
- Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah, United States
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, United States
- Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, United States
| |
Collapse
|
12
|
Ramirez P, Portillo S, Cervera J, Bisquert J, Mafe S. Memristive arrangements of nanofluidic pores. Phys Rev E 2024; 109:044803. [PMID: 38755814 DOI: 10.1103/physreve.109.044803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/29/2024] [Indexed: 05/18/2024]
Abstract
We demonstrate that nanofluidic diodes in multipore membranes show a memristive behavior that can be controlled not only by the amplitude and frequency of the external signal but also by series and parallel arrangements of the membranes. Each memristor consists of a polymeric membrane with conical nanopores that allow current rectification due to the electrical interaction between the ionic solution and the pore surface charges. This surface charge-regulated ionic transport shows a rich nonlinear physics, including memory and inductive effects, which are characterized here by the current-voltage curves and electrical impedance spectroscopy. Also, neuromorphiclike potentiation of the membrane conductance following voltage pulses (spikes) is observed. The multipore membrane with nanofluidic diodes shows physical concepts that should have application for information processing and signal conversion in iontronics hybrid devices.
Collapse
Affiliation(s)
- Patricio Ramirez
- Departament de Física Aplicada, Universitat Politècnica de València, E-46022 València, Spain
| | - Sergio Portillo
- Departament de Física de la Terra i Termodinàmica, Universitat de València, E-46100 Burjassot, Spain
| | - Javier Cervera
- Departament de Física de la Terra i Termodinàmica, Universitat de València, E-46100 Burjassot, Spain
| | - Juan Bisquert
- Institute of Advanced Materials (INAM), Universitat Jaume I, 12006 Castelló, Spain
| | - Salvador Mafe
- Departament de Física de la Terra i Termodinàmica, Universitat de València, E-46100 Burjassot, Spain
- Allen Discovery Center at Tufts University, Medford, Massachusetts 02155, USA
| |
Collapse
|
13
|
Okhuarobo A, Kreifeldt M, Gandhi PJ, Lopez C, Martinez B, Fleck K, Bajo M, Bhattacharyya P, Dopico AM, Roberto M, Roberts AJ, Homanics GE, Contet C. Ethanol's interaction with BK channel α subunit residue K361 does not mediate behavioral responses to alcohol in mice. Mol Psychiatry 2024; 29:529-542. [PMID: 38135755 PMCID: PMC11116116 DOI: 10.1038/s41380-023-02346-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023]
Abstract
Large conductance potassium (BK) channels are among the most sensitive molecular targets of ethanol and genetic variations in the channel-forming α subunit have been nominally associated with alcohol use disorders. However, whether the action of ethanol at BK α influences the motivation to drink alcohol remains to be determined. To address this question, we first tested the effect of systemically administered BK channel modulators on voluntary alcohol consumption in C57BL/6J males. Penitrem A (blocker) exerted dose-dependent effects on moderate alcohol intake, while paxilline (blocker) and BMS-204352 (opener) were ineffective. Because pharmacological manipulations are inherently limited by non-specific effects, we then sought to investigate the behavioral relevance of ethanol's direct interaction with BK α by introducing in the mouse genome a point mutation known to render BK channels insensitive to ethanol while preserving their physiological function. The BK α K361N substitution prevented ethanol from reducing spike threshold in medial habenula neurons. However, it did not alter acute responses to ethanol in vivo, including ataxia, sedation, hypothermia, analgesia, and conditioned place preference. Furthermore, the mutation did not have reproducible effects on alcohol consumption in limited, continuous, or intermittent access home cage two-bottle choice paradigms conducted in both males and females. Notably, in contrast to previous observations made in mice missing BK channel auxiliary β subunits, the BK α K361N substitution had no significant impact on ethanol intake escalation induced by chronic intermittent alcohol vapor inhalation. It also did not affect the metabolic and locomotor consequences of chronic alcohol exposure. Altogether, these data suggest that the direct interaction of ethanol with BK α does not mediate the alcohol-related phenotypes examined here in mice.
Collapse
Affiliation(s)
- Agbonlahor Okhuarobo
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Max Kreifeldt
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Pauravi J Gandhi
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Catherine Lopez
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Briana Martinez
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Kiera Fleck
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Michal Bajo
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | | | - Alex M Dopico
- University of Tennessee Health Science Center, Department of Pharmacology, Addiction Science, and Toxicology, Memphis, TN, USA
| | - Marisa Roberto
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Amanda J Roberts
- The Scripps Research Institute, Animals Models Core Facility, La Jolla, CA, USA
| | - Gregg E Homanics
- University of Pittsburgh, Department of Anesthesiology and Perioperative Medicine, Pittsburgh, PA, USA
| | - Candice Contet
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA.
| |
Collapse
|
14
|
Stengl M, Schneider AC. Contribution of membrane-associated oscillators to biological timing at different timescales. Front Physiol 2024; 14:1243455. [PMID: 38264332 PMCID: PMC10803594 DOI: 10.3389/fphys.2023.1243455] [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: 06/20/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024] Open
Abstract
Environmental rhythms such as the daily light-dark cycle selected for endogenous clocks. These clocks predict regular environmental changes and provide the basis for well-timed adaptive homeostasis in physiology and behavior of organisms. Endogenous clocks are oscillators that are based on positive feedforward and negative feedback loops. They generate stable rhythms even under constant conditions. Since even weak interactions between oscillators allow for autonomous synchronization, coupling/synchronization of oscillators provides the basis of self-organized physiological timing. Amongst the most thoroughly researched clocks are the endogenous circadian clock neurons in mammals and insects. They comprise nuclear clockworks of transcriptional/translational feedback loops (TTFL) that generate ∼24 h rhythms in clock gene expression entrained to the environmental day-night cycle. It is generally assumed that this TTFL clockwork drives all circadian oscillations within and between clock cells, being the basis of any circadian rhythm in physiology and behavior of organisms. Instead of the current gene-based hierarchical clock model we provide here a systems view of timing. We suggest that a coupled system of autonomous TTFL and posttranslational feedback loop (PTFL) oscillators/clocks that run at multiple timescales governs adaptive, dynamic homeostasis of physiology and behavior. We focus on mammalian and insect neurons as endogenous oscillators at multiple timescales. We suggest that neuronal plasma membrane-associated signalosomes constitute specific autonomous PTFL clocks that generate localized but interlinked oscillations of membrane potential and intracellular messengers with specific endogenous frequencies. In each clock neuron multiscale interactions of TTFL and PTFL oscillators/clocks form a temporally structured oscillatory network with a common complex frequency-band comprising superimposed multiscale oscillations. Coupling between oscillator/clock neurons provides the next level of complexity of an oscillatory network. This systemic dynamic network of molecular and cellular oscillators/clocks is suggested to form the basis of any physiological homeostasis that cycles through dynamic homeostatic setpoints with a characteristic frequency-band as hallmark. We propose that mechanisms of homeostatic plasticity maintain the stability of these dynamic setpoints, whereas Hebbian plasticity enables switching between setpoints via coupling factors, like biogenic amines and/or neuropeptides. They reprogram the network to a new common frequency, a new dynamic setpoint. Our novel hypothesis is up for experimental challenge.
Collapse
Affiliation(s)
- Monika Stengl
- Department of Biology, Animal Physiology/Neuroethology, University of Kassel, Kassel, Germany
| | | |
Collapse
|
15
|
Hearn JI, Alhilali M, Kim M, Kalev-Zylinska ML, Poulsen RC. N-methyl-D-aspartate receptor regulates the circadian clock in megakaryocytic cells and impacts cell proliferation through BMAL1. Platelets 2023; 34:2206918. [PMID: 37183795 DOI: 10.1080/09537104.2023.2206918] [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: 08/13/2022] [Revised: 02/16/2023] [Accepted: 04/17/2023] [Indexed: 05/16/2023]
Abstract
Peripheral circadian clocks control cell proliferation and survival, but little is known about their role and regulation in megakaryocytic cells. N-methyl-D-aspartate receptor (NMDAR) regulates the central clock in the brain. The purpose of this study was to determine whether NMDAR regulates the megakaryocytic cell clock and whether the megakaryocytic clock regulates cell proliferation and cell death. We found that both the Meg-01 megakaryocytic cell line and native murine megakaryocytes expressed circadian clock genes. Megakaryocyte-directed deletion of Grin1 in mice caused significant disruption of the circadian rhythm pathway at the transcriptional level and increased expression of BMAL1 at the protein level. Similarly, both pharmacological (MK-801) and genetic (GRIN-/-) inhibition of NMDAR in Meg-01 cells in vitro resulted in widespread changes in clock gene expression including increased expression of BMAL1, the core clock transcription factor. BMAL1 overexpression reduced Meg-01 cell proliferation and altered the time-dependent expression of the cell cycle regulators MYC and WEE1, whereas BMAL1 knockdown led to increased cell death in Meg-01-GRIN1-/- cells. Our results demonstrate that NMDAR regulates the circadian clock in megakaryocytic cells and that the circadian clock component BMAL1 contributes to the control of Meg-01 cell proliferation and survival.
Collapse
Affiliation(s)
- James I Hearn
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Mariam Alhilali
- Department of Medicine, School of Medicine, University of Auckland, Auckland, New Zealand
| | - Minah Kim
- Department of Medicine, School of Medicine, University of Auckland, Auckland, New Zealand
| | - Maggie L Kalev-Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- Department of Pathology and Laboratory Medicine, Haematology Laboratory, Auckland City Hospital, Auckland, New Zealand
| | - Raewyn C Poulsen
- Department of Medicine, School of Medicine, University of Auckland, Auckland, New Zealand
- Department of Pharmacology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
16
|
Ramirez P, Gómez V, Cervera J, Mafe S, Bisquert J. Synaptical Tunability of Multipore Nanofluidic Memristors. J Phys Chem Lett 2023:10930-10934. [PMID: 38033300 DOI: 10.1021/acs.jpclett.3c02796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
We demonstrate a multipore nanofluidic memristor with conical pores showcasing a wide range of hysteresis and memristor properties that provide functionalities for brainlike computation in neuromorphic applications. Leveraging the interplay between the charged functional groups on the pore surfaces and the confined ionic solution, the memristor characteristics are modulated through the electrolyte type, ionic concentrations, and pH levels of the aqueous solution. The multipore membrane mimics the functional characteristics of biological ion channels and displays synaptical potentiation and depression. Furthermore, this property can be inverted in polarity by chemically varying the pH level. The ability to modulate memory effects by ionic conductivity holds promise for enhancing signal information processing capabilities.
Collapse
Affiliation(s)
- Patricio Ramirez
- Dept. de Física Aplicada, Universitat Politècnica de València, E-46022 València, Spain
| | - Vicente Gómez
- Dept. de Física Aplicada, Universitat Politècnica de València, E-46022 València, Spain
| | - Javier Cervera
- Dept. de Física de la Terra i Termodinàmica, Universitat de València, E-46100 Burjassot, Spain
| | - Salvador Mafe
- Dept. de Física Aplicada, Universitat Politècnica de València, E-46022 València, Spain
- Dept. de Física de la Terra i Termodinàmica, Universitat de València, E-46100 Burjassot, Spain
| | - Juan Bisquert
- Institute of Advanced Materials (INAM), Universitat Jaume I, 12006 Castelló, Spain
| |
Collapse
|
17
|
North KC, Shaw AA, Bukiya AN, Dopico AM. Progesterone activation of β 1-containing BK channels involves two binding sites. Nat Commun 2023; 14:7248. [PMID: 37945687 PMCID: PMC10636063 DOI: 10.1038/s41467-023-42827-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Progesterone (≥1 µM) is used in recovery of cerebral ischemia, an effect likely contributed to by cerebrovascular dilation. The targets of this progesterone action are unknown. We report that micromolar (µM) progesterone activates mouse cerebrovascular myocyte BK channels; this action is lost in β1-/- mice myocytes and in lipid bilayers containing BK α subunit homomeric channels but sustained on β1/β4-containing heteromers. Progesterone binds to both regulatory subunits, involving two steroid binding sites conserved in β1-β4: high-affinity (sub-µM), which involves Trp87 in β1 loop, and low-affinity (µM) defined by TM1 Tyr32 and TM2 Trp163. Thus progesterone, but not its oxime, bridges TM1-TM2. Mutation of the high-affinity site blunts channel activation by progesterone underscoring a permissive role of the high-affinity site: progesterone binding to this site enables steroid binding at the low-affinity site, which activates the channel. In support of our model, cerebrovascular dilation evoked by μM progesterone is lost by mutating Tyr32 or Trp163 in β1 whereas these mutations do not affect alcohol-induced cerebrovascular constriction. Furthermore, this alcohol action is effectively counteracted both in vitro and in vivo by progesterone but not by its oxime.
Collapse
Affiliation(s)
- Kelsey C North
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Andrew A Shaw
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA.
| |
Collapse
|
18
|
Cervera J, Levin M, Mafe S. Correcting instructive electric potential patterns in multicellular systems: External actions and endogenous processes. Biochim Biophys Acta Gen Subj 2023; 1867:130440. [PMID: 37527731 DOI: 10.1016/j.bbagen.2023.130440] [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: 04/15/2023] [Revised: 06/19/2023] [Accepted: 07/28/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND Transmembrane electrical potential differences in cells modulate the spatio-temporal distribution of signaling ions and molecules that are instructive for downstream signaling pathways in multicellular systems. The local coupling between bioelectricity and protein transcription patterns allows dynamic subsystems (modules) of cells that share the same bioelectrical state to show similar biochemical downstream processes. METHODS We simulate theoretically how the integration-segregation pattern formed by the different multicellular modules that define a biosystem can be controlled by multicellular potentials. To this end, we couple together the model equations of the bioelectrical network to those of the genetic network. RESULTS The coupling provided by the intercellular junctions and the external microenvironment allows the restoration of the target bioelectrical pattern by changing the transcription rate of specific ion channels, the post-translational blocking of these channels, and changes in the environmental ionic concentrations. CONCLUSIONS The simulations show that the single-cell feedback between bioelectrical and transcriptional processes, together with the coupling provided by the intercellular junctions and the environment, can correct large-scale patterns by means of suitable external actions. GENERAL SIGNIFICANCE This study provides a theoretical advancement in the understanding of how the multicellular bioelectric coupling may guide repolarizing interventions for regenerating a tissue, with potential implications in biomedicine.
Collapse
Affiliation(s)
- Javier Cervera
- Dept. Termodinàmica, Facultat de Física, Universitat de València, E-46100 Burjassot, Spain.
| | - Michael Levin
- Dept. of Biology and Allen Discovery Center at Tufts University, Medford, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, USA
| | - Salvador Mafe
- Dept. Termodinàmica, Facultat de Física, Universitat de València, E-46100 Burjassot, Spain
| |
Collapse
|
19
|
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
|
20
|
Short B. Kv12 channels flick the switch. J Gen Physiol 2023; 155:e202313459. [PMID: 37584659 PMCID: PMC10433146 DOI: 10.1085/jgp.202313459] [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: 08/17/2023] Open
Abstract
JGP study (Hermanstyne et al. 2023. J. Gen. Physiol.https://doi.org/10.1085/jgp.202213310) shows that Kv12-encoded K+ currents reduce the repetitive firing rates of SCN neurons at night, thereby regulating daily oscillations in the master circadian pacemaker.
Collapse
|
21
|
Thirouin ZS, Gizowski C, Murtaz A, Bourque CW. Sex-specific differences in the circadian pattern of action potential firing by rat suprachiasmatic nucleus vasopressin neurons. J Neuroendocrinol 2023; 35:e13273. [PMID: 37132408 DOI: 10.1111/jne.13273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 05/04/2023]
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus serves as the master circadian clock in mammals. Most SCN neurons express the inhibitory neurotransmitter GABA (gamma amino butyric acid) along with a peptide cotransmitter. Notably, the neuropeptides vasopressin (VP) and vasoactive intestinal peptide (VIP) define two prominent clusters within the SCN: those located in the ventral core (VIP) and those forming the dorsomedial "shell" of the nucleus (VP). Axons emerging from VP neurons in the shell are thought to mediate much of the SCN's output to other brain regions as well as VP release into the cerebrospinal fluid (CSF). Previous work has shown that VP release by SCN neurons is activity dependent and SCN VP neurons fire action potentials at a higher rate during the light phase. Accordingly, CSF VP levels are higher during daytime. Interestingly, the amplitude of the CSF VP rhythm is greater in males than females, suggesting the existence of sex differences in the electrical activity of SCN VP neurons. Here we investigated this hypothesis by performing cell-attached recordings from 1070 SCN VP neurons across the entire circadian cycle in both sexes of transgenic rats that express green fluorescent protein (GFP) driven by the VP gene promoter. Using an immunocytochemical approach we confirmed that >60% of SCN VP neurons display visible GFP. Recordings in acute coronal slices revealed that VP neurons display a striking circadian pattern of action potential firing, but the characteristics of this activity cycle differ in males and females. Specifically, neurons in males reached a significantly higher peak firing frequency during subjective daytime compared to females and the acrophase occurred ~1 h earlier in females. Peak firing rates in females were not significantly different at various phases of the estrous cycle.
Collapse
Affiliation(s)
- Zahra S Thirouin
- Center for Research in Neuroscience, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Claire Gizowski
- Center for Research in Neuroscience, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Anzala Murtaz
- Center for Research in Neuroscience, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Charles W Bourque
- Center for Research in Neuroscience, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| |
Collapse
|
22
|
Tang Q, Godschall E, Brennan CD, Zhang Q, Abraham-Fan RJ, Williams SP, Güngül TB, Onoharigho R, Buyukaksakal A, Salinas R, Sajonia IR, Olivieri JJ, Calhan OY, Deppmann CD, Campbell JN, Podyma B, Güler AD. Leptin receptor neurons in the dorsomedial hypothalamus input to the circadian feeding network. SCIENCE ADVANCES 2023; 9:eadh9570. [PMID: 37624889 PMCID: PMC10456850 DOI: 10.1126/sciadv.adh9570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023]
Abstract
Salient cues, such as the rising sun or availability of food, entrain biological clocks for behavioral adaptation. The mechanisms underlying entrainment to food availability remain elusive. Using single-nucleus RNA sequencing during scheduled feeding, we identified a dorsomedial hypothalamus leptin receptor-expressing (DMHLepR) neuron population that up-regulates circadian entrainment genes and exhibits calcium activity before an anticipated meal. Exogenous leptin, silencing, or chemogenetic stimulation of DMHLepR neurons disrupts the development of molecular and behavioral food entrainment. Repetitive DMHLepR neuron activation leads to the partitioning of a secondary bout of circadian locomotor activity that is in phase with the stimulation and dependent on an intact suprachiasmatic nucleus (SCN). Last, we found a DMHLepR neuron subpopulation that projects to the SCN with the capacity to influence the phase of the circadian clock. This direct DMHLepR-SCN connection is well situated to integrate the metabolic and circadian systems, facilitating mealtime anticipation.
Collapse
Affiliation(s)
- Qijun Tang
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Elizabeth Godschall
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Charles D. Brennan
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Qi Zhang
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | | | - Sydney P. Williams
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Taha Buğra Güngül
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Roberta Onoharigho
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Aleyna Buyukaksakal
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Ricardo Salinas
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Isabelle R. Sajonia
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Joey J. Olivieri
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - O. Yipkin Calhan
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Christopher D. Deppmann
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
- Program in Fundamental Neuroscience, Charlottesville, VA 22904, USA
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22904, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - John N. Campbell
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Brandon Podyma
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Ali D. Güler
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
- Program in Fundamental Neuroscience, Charlottesville, VA 22904, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
23
|
Thangaleela S, Sivamaruthi BS, Kesika P, Mariappan S, Rashmi S, Choeisoongnern T, Sittiprapaporn P, Chaiyasut C. Neurological Insights into Sleep Disorders in Parkinson's Disease. Brain Sci 2023; 13:1202. [PMID: 37626558 PMCID: PMC10452387 DOI: 10.3390/brainsci13081202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/07/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Parkinson's disease (PD) is a common multidimensional neurological disorder characterized by motor and non-motor features and is more prevalent in the elderly. Sleep disorders and cognitive disturbances are also significant characteristics of PD. Sleep is an important physiological process for normal human cognition and physical functioning. Sleep deprivation negatively impacts human physical, mental, and behavioral functions. Sleep disturbances include problems falling asleep, disturbances occurring during sleep, abnormal movements during sleep, insufficient sleep, and excessive sleep. The most recognizable and known sleep disorders, such as rapid-eye-movement behavior disorder (RBD), insomnia, excessive daytime sleepiness (EDS), restless legs syndrome (RLS), sleep-related breathing disorders (SRBDs), and circadian-rhythm-related sleep-wake disorders (CRSWDs), have been associated with PD. RBD and associated emotional disorders are common non-motor symptoms of PD. In individuals, sleep disorders and cognitive impairment are important prognostic factors for predicting progressing neurodegeneration and developing dementia conditions in PD. Studies have focused on RBD and its associated neurological changes and functional deficits in PD patients. Other risks, such as cognitive decline, anxiety, and depression, are related to RBD. Sleep-disorder diagnosis is challenging, especially in identifying the essential factors that disturb the sleep-wake cycle and the co-existence of other concomitant sleep issues, motor symptoms, and breathing disorders. Focusing on sleep patterns and their disturbances, including genetic and other neurochemical changes, helps us to better understand the central causes of sleep alterations and cognitive functions in PD patients. Relations between α-synuclein aggregation in the brain and gender differences in sleep disorders have been reported. The existing correlation between sleep disorders and levels of α-synuclein in the cerebrospinal fluid indicates the risk of progression of synucleinopathies. Multidirectional approaches are required to correlate sleep disorders and neuropsychiatric symptoms and diagnose sensitive biomarkers for neurodegeneration. The evaluation of sleep pattern disturbances and cognitive impairment may aid in the development of novel and effective treatments for PD.
Collapse
Affiliation(s)
- Subramanian Thangaleela
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (B.S.S.); (P.K.)
| | - Bhagavathi Sundaram Sivamaruthi
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (B.S.S.); (P.K.)
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Periyanaina Kesika
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (B.S.S.); (P.K.)
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Subramanian Rashmi
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (B.S.S.); (P.K.)
| | - Thiwanya Choeisoongnern
- Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand
| | - Phakkharawat Sittiprapaporn
- Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand
| | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (B.S.S.); (P.K.)
| |
Collapse
|
24
|
Yang JJ, Huang RC. Afterhyperpolarization potential modulated by local [K +] o in K + diffusion-restricted extracellular space in the central clock of suprachiasmatic nucleus. Biomed J 2023; 46:100551. [PMID: 35863667 PMCID: PMC10345224 DOI: 10.1016/j.bj.2022.07.005] [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: 03/06/2022] [Revised: 05/31/2022] [Accepted: 07/14/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Intercellular coupling is essential for the suprachiasmatic nucleus (SCN) to serve as a coherent central clock. Synaptic release of neurotransmitters and neuropeptides is critical for synchronizing SCN neurons. However, intercellular coupling via non-synaptic mechanisms has also been demonstrated. In particular, the abundant perikaryal appositions with morphological specializations in the narrow extracellular space (ECS) may hinder molecular diffusion to allow for ion accumulation or depletion. METHODS The SCN neurons were recorded in the whole-cell current-clamp mode, with pipette filled with high (26 mM)-Na+ or low (6 mM)-Na+ solution. RESULTS Cells recorded with high-Na+ pipette solution could fire spontaneous action potentials (AP) with peak AHP more negative than the calculated value of K+ equilibrium potential (EK) and with peak AP more positive than calculated ENa. Cells recorded with low-Na+ pipette solution could also have peak AHP more negative than calculated EK. In contrast, the resting membrane potential (RMP) was always less negative to calculated EK. The distribution and the averaged amplitude of peak AHP, peak AP, or RMP was similar between cells recorded with high-Na+ and low-Na+ solution pipette. In a number of cells, the peak AHP could increase from more positive to become more negative than calculated EK spontaneously or after treatments to hyperpolarize the RMP. TTX blocked the Na+ -dependent APs and tetraethylammonium (TEA), but not Ba2+ or Cd2+, markedly reduced the peak AHP. Perforated-patch cells could also but rarely fire APs with peak AHP more negative than calculated EK. CONCLUSION The result of peak AHP negative to calculated EK indicates that local [K+]o sensed by the TEA-sensitive AHP K+ channels must be lower than bulk [K+]o, most likely due to K+ clearance from K+ diffusion-restricted ECS by the Na+/K+-ATPase. The K+ diffusion-restricted ECS may allow for K+-mediated ionic interactions among neurons to regulate SCN excitability.
Collapse
Affiliation(s)
- Jyh-Jeen Yang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Rong-Chi Huang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| |
Collapse
|
25
|
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
|
26
|
Patton AP, Morris EL, McManus D, Wang H, Li Y, Chin JW, Hastings MH. Astrocytic control of extracellular GABA drives circadian timekeeping in the suprachiasmatic nucleus. Proc Natl Acad Sci U S A 2023; 120:e2301330120. [PMID: 37186824 PMCID: PMC10214171 DOI: 10.1073/pnas.2301330120] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
The hypothalamic suprachiasmatic nucleus (SCN) is the master mammalian circadian clock. Its cell-autonomous timing mechanism, a transcriptional/translational feedback loop (TTFL), drives daily peaks of neuronal electrical activity, which in turn control circadian behavior. Intercellular signals, mediated by neuropeptides, synchronize and amplify TTFL and electrical rhythms across the circuit. SCN neurons are GABAergic, but the role of GABA in circuit-level timekeeping is unclear. How can a GABAergic circuit sustain circadian cycles of electrical activity, when such increased neuronal firing should become inhibitory to the network? To explore this paradox, we show that SCN slices expressing the GABA sensor iGABASnFR demonstrate a circadian oscillation of extracellular GABA ([GABA]e) that, counterintuitively, runs in antiphase to neuronal activity, with a prolonged peak in circadian night and a pronounced trough in circadian day. Resolving this unexpected relationship, we found that [GABA]e is regulated by GABA transporters (GATs), with uptake peaking during circadian day, hence the daytime trough and nighttime peak. This uptake is mediated by the astrocytically expressed transporter GAT3 (Slc6a11), expression of which is circadian-regulated, being elevated in daytime. Clearance of [GABA]e in circadian day facilitates neuronal firing and is necessary for circadian release of the neuropeptide vasoactive intestinal peptide, a critical regulator of TTFL and circuit-level rhythmicity. Finally, we show that genetic complementation of the astrocytic TTFL alone, in otherwise clockless SCN, is sufficient to drive [GABA]e rhythms and control network timekeeping. Thus, astrocytic clocks maintain the SCN circadian clockwork by temporally controlling GABAergic inhibition of SCN neurons.
Collapse
Affiliation(s)
- Andrew P. Patton
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Emma L. Morris
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - David McManus
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Huan Wang
- State Key Laboratory of Membrane Biology, Peking University, School of Life Sciences, 100871Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University, School of Life Sciences, 100871Beijing, China
| | - Jason W. Chin
- PNAC Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Michael H. Hastings
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| |
Collapse
|
27
|
Geng Y, Li P, Butler A, Wang B, Salkoff L, Magleby KL. BK channels of five different subunit combinations underlie the de novo KCNMA1 G375R channelopathy. J Gen Physiol 2023; 155:e202213302. [PMID: 36995317 PMCID: PMC10067970 DOI: 10.1085/jgp.202213302] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/08/2023] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
The molecular basis of a severe developmental and neurological disorder associated with a de novo G375R variant of the tetrameric BK channel is unknown. Here, we address this question by recording from single BK channels expressed to mimic a G375R mutation heterozygous with a WT allele. Five different types of functional BK channels were expressed: 3% were consistent with WT, 12% with homotetrameric mutant, and 85% with three different types of hybrid (heterotetrameric) channels assembled from both mutant and WT subunits. All channel types except WT showed a marked gain-of-function in voltage activation and a smaller decrease-of-function in single-channel conductance, with both changes in function becoming more pronounced as the number of mutant subunits per tetrameric channel increased. The net cellular response from the five different types of channels comprising the molecular phenotype was a shift of -120 mV in the voltage required to activate half of the maximal current through BK channels, giving a net gain-of-function. The WT and homotetrameric mutant channels in the molecular phenotype were consistent with genetic codominance as each displayed properties of a channel arising from only one of the two alleles. The three types of hybrid channels in the molecular phenotype were consistent with partial dominance as their properties were intermediate between those of mutant and WT channels. A model in which BK channels randomly assemble from mutant and WT subunits, with each subunit contributing increments of activation and conductance, approximated the molecular phenotype of the heterozygous G375R mutation.
Collapse
Affiliation(s)
- Yanyan Geng
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Ping Li
- Department of Neuroscience, Washington University St. Louis, St. Louis, MO, USA
| | - Alice Butler
- Department of Neuroscience, Washington University St. Louis, St. Louis, MO, USA
| | - Bill Wang
- Department of Neuroscience, Washington University St. Louis, St. Louis, MO, USA
| | - Lawrence Salkoff
- Department of Neuroscience, Washington University St. Louis, St. Louis, MO, USA
- Department of Genetics, Washington University St. Louis, St. Louis, MO, USA
| | - Karl L. Magleby
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
28
|
Bafna A, Banks G, Hastings MH, Nolan PM. Dynamic modulation of genomic enhancer elements in the suprachiasmatic nucleus, the site of the mammalian circadian clock. Genome Res 2023; 33:673-688. [PMID: 37156620 PMCID: PMC10317116 DOI: 10.1101/gr.277581.122] [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: 12/16/2022] [Accepted: 05/03/2023] [Indexed: 05/10/2023]
Abstract
The mammalian suprachiasmatic nucleus (SCN), located in the ventral hypothalamus, synchronizes and maintains daily cellular and physiological rhythms across the body, in accordance with environmental and visceral cues. Consequently, the systematic regulation of spatiotemporal gene transcription in the SCN is vital for daily timekeeping. So far, the regulatory elements assisting circadian gene transcription have only been studied in peripheral tissues, lacking the critical neuronal dimension intrinsic to the role of the SCN as central brain pacemaker. By using histone-ChIP-seq, we identified SCN-enriched gene regulatory elements that associated with temporal gene expression. Based on tissue-specific H3K27ac and H3K4me3 marks, we successfully produced the first-ever SCN gene-regulatory map. We found that a large majority of SCN enhancers not only show robust 24-h rhythmic modulation in H3K27ac occupancy, peaking at distinct times of day, but also possess canonical E-box (CACGTG) motifs potentially influencing downstream cycling gene expression. To establish enhancer-gene relationships in the SCN, we conducted directional RNA-seq at six distinct times across the day and night, and studied the association between dynamically changing histone acetylation and gene transcript levels. About 35% of the cycling H3K27ac sites were found adjacent to rhythmic gene transcripts, often preceding the rise in mRNA levels. We also noted that enhancers encompass noncoding, actively transcribing enhancer RNAs (eRNAs) in the SCN, which in turn oscillate, along with cyclic histone acetylation, and correlate with rhythmic gene transcription. Taken together, these findings shed light on genome-wide pretranscriptional regulation operative in the central clock that confers its precise and robust oscillation necessary to orchestrate daily timekeeping in mammals.
Collapse
Affiliation(s)
- Akanksha Bafna
- Medical Research Council, Harwell Science Campus, Oxfordshire OX11 0RD, United Kingdom;
| | - Gareth Banks
- Medical Research Council, Harwell Science Campus, Oxfordshire OX11 0RD, United Kingdom
| | - Michael H Hastings
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, United Kingdom
| | - Patrick M Nolan
- Medical Research Council, Harwell Science Campus, Oxfordshire OX11 0RD, United Kingdom;
| |
Collapse
|
29
|
Ragozzino FJ, Peterson B, Karatsoreos IN, Peters JH. Circadian regulation of glutamate release pathways shapes synaptic throughput in the brainstem nucleus of the solitary tract (NTS). J Physiol 2023; 601:1881-1896. [PMID: 36975145 PMCID: PMC10192157 DOI: 10.1113/jp284370] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
Circadian regulation of autonomic reflex pathways pairs physiological function with the daily light cycle. The brainstem nucleus of the solitary tract (NTS) is a key candidate for rhythmic control of the autonomic nervous system. Here we investigated circadian regulation of NTS neurotransmission and synaptic throughput using patch-clamp electrophysiology in brainstem slices from mice. We found that spontaneous quantal glutamate release onto NTS neurons showed strong circadian rhythmicity, with the highest rate of release during the light phase and the lowest in the dark, that were sufficient to drive day/night differences in constitutive postsynaptic action potential firing. In contrast, afferent evoked action potential throughput was enhanced during the dark and diminished in the light. Afferent-driven synchronous release pathways showed a similar decrease in release probability that did not explain the enhanced synaptic throughput during the night. However, analysis of postsynaptic membrane properties revealed diurnal changes in conductance, which, when coupled with the circadian changes in glutamate release pathways, tuned synaptic throughput between the light and dark phases. These coordinated pre-/postsynaptic changes encode nuanced control over synaptic performance and pair NTS action potential firing and vagal throughput with time of day. KEY POINTS: Vagal afferent neurons relay information from peripheral organs to the brainstem nucleus of the solitary tract (NTS) to initiate autonomic reflex pathways as well as providing important controls of food intake, digestive function and energy balance. Vagally mediated reflexes and behaviours are under strong circadian regulation. Diurnal fluctuations in presynaptic vesicle release pathways and postsynaptic membrane conductances provide nuanced control over NTS action potential firing and vagal synaptic throughput. Coordinated pre-/postsynaptic changes represent a fundamental mechanism mediating daily changes in vagal afferent signalling and autonomic function.
Collapse
Affiliation(s)
- Forrest J. Ragozzino
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - BreeAnne Peterson
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Ilia N. Karatsoreos
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - James H. Peters
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
30
|
Piet R. Circadian and kisspeptin regulation of the preovulatory surge. Peptides 2023; 163:170981. [PMID: 36842628 DOI: 10.1016/j.peptides.2023.170981] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 02/28/2023]
Abstract
Fertility in mammals is ultimately controlled by a small population of neurons - the gonadotropin-releasing hormone (GnRH) neurons - located in the ventral forebrain. GnRH neurons control gonadal function through the release of GnRH, which in turn stimulates the secretion of the anterior pituitary gonadotropins luteinizing hormone (LH) and follicle-stimulating hormone (FSH). In spontaneous ovulators, ovarian follicle maturation eventually stimulates, via sex steroid feedback, the mid-cycle surge in GnRH and LH secretion that causes ovulation. The GnRH/LH surge is initiated in many species just before the onset of activity through processes controlled by the central circadian clock, ensuring that the neuroendocrine control of ovulation and sex behavior are coordinated. This review aims to give an overview of anatomical and functional studies that collectively reveal some of the mechanisms through which the central circadian clock regulates GnRH neurons and their afferent circuits to drive the preovulatory surge.
Collapse
Affiliation(s)
- Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States.
| |
Collapse
|
31
|
Cervera J, Ramirez P, Nasir S, Ali M, Ensinger W, Siwy ZS, Mafe S. Cation pumping against a concentration gradient in conical nanopores characterized by load capacitors. Bioelectrochemistry 2023; 152:108445. [PMID: 37086711 DOI: 10.1016/j.bioelechem.2023.108445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/24/2023]
Abstract
We study the cation transport against an external concentration gradient (cation pumping) that occurs in conical nanopores when zero-average oscillatory and white noise potentials are externally applied. This pumping, based on the electrically asymmetric nanostructure, is characterized here by a load capacitor arrangement. In the case of white noise signals, the conical nanopore acts as an electrical valve that allows extraction of order from chaos. No molecular carriers, specific ion pumps, and competitive ion-binding phenomena are required. The nanopore conductance on/off states mimic those of the voltage-gated ion channels in the cell membrane. These channels allow modulating membrane potentials and ionic concentration gradients along oscillatory pulses in circadian rhythms and the cell cycle. We show that the combination of asymmetric nanostructures with load capacitors can be useful for the understanding of nanofluidic processes based on bioelectrochemical gradients.
Collapse
Affiliation(s)
- Javier Cervera
- Departament de Física de la Terra i Termodinàmica, Universitat de València, E-46100 Burjassot, Spain.
| | - Patricio Ramirez
- Departament de Física Aplicada, Universitat Politècnica de València, E-46022 València, Spain
| | - Saima Nasir
- Materials Research Department, GSI Helmholtzzentrum für Schwerionenforschung, D-64291 Darmstadt, Germany; Department of Material- and Geo-Sciences, Technische Universität Darmstadt, D-64287 Darmstadt, Germany
| | - Mubarak Ali
- Materials Research Department, GSI Helmholtzzentrum für Schwerionenforschung, D-64291 Darmstadt, Germany; Department of Material- and Geo-Sciences, Technische Universität Darmstadt, D-64287 Darmstadt, Germany
| | - Wolfgang Ensinger
- Department of Material- and Geo-Sciences, Technische Universität Darmstadt, D-64287 Darmstadt, Germany
| | - Zuzanna S Siwy
- Department of Physics and Astronomy, Department of Chemistry, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Salvador Mafe
- Departament de Física de la Terra i Termodinàmica, Universitat de València, E-46100 Burjassot, Spain.
| |
Collapse
|
32
|
Stangherlin A. Ion dynamics and the regulation of circadian cellular physiology. Am J Physiol Cell Physiol 2023; 324:C632-C643. [PMID: 36689675 DOI: 10.1152/ajpcell.00378.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Circadian rhythms in physiology and behavior allow organisms to anticipate the daily environmental changes imposed by the rotation of our planet around its axis. Although these rhythms eventually manifest at the organismal level, a cellular basis for circadian rhythms has been demonstrated. Significant contributors to these cell-autonomous rhythms are daily cycles in gene expression and protein translation. However, recent data revealed cellular rhythms in other biological processes, including ionic currents, ion transport, and cytosolic ion abundance. Circadian rhythms in ion currents sustain circadian variation in action potential firing rate, which coordinates neuronal behavior and activity. Circadian regulation of metal ions abundance and dynamics is implicated in distinct cellular processes, from protein translation to membrane activity and osmotic homeostasis. In turn, studies showed that manipulating ion abundance affects the expression of core clock genes and proteins, suggestive of a close interplay. However, the relationship between gene expression cycles, ion dynamics, and cellular function is still poorly characterized. In this review, I will discuss the mechanisms that generate ion rhythms, the cellular functions they govern, and how they feed back to regulate the core clock machinery.
Collapse
Affiliation(s)
- Alessandra Stangherlin
- Faculty of Medicine and University Hospital Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Institute for Mitochondrial Diseases and Ageing, University of Cologne, Cologne, Germany
| |
Collapse
|
33
|
Circadian disruption and sleep disorders in neurodegeneration. Transl Neurodegener 2023; 12:8. [PMID: 36782262 PMCID: PMC9926748 DOI: 10.1186/s40035-023-00340-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/03/2023] [Indexed: 02/15/2023] Open
Abstract
Disruptions of circadian rhythms and sleep cycles are common among neurodegenerative diseases and can occur at multiple levels. Accumulating evidence reveals a bidirectional relationship between disruptions of circadian rhythms and sleep cycles and neurodegenerative diseases. Circadian disruption and sleep disorders aggravate neurodegeneration and neurodegenerative diseases can in turn disrupt circadian rhythms and sleep. Importantly, circadian disruption and various sleep disorders can increase the risk of neurodegenerative diseases. Thus, harnessing the circadian biology findings from preclinical and translational research in neurodegenerative diseases is of importance for reducing risk of neurodegeneration and improving symptoms and quality of life of individuals with neurodegenerative disorders via approaches that normalize circadian in the context of precision medicine. In this review, we discuss the implications of circadian disruption and sleep disorders in neurodegenerative diseases by summarizing evidence from both human and animal studies, focusing on the bidirectional links of sleep and circadian rhythms with prevalent forms of neurodegeneration. These findings provide valuable insights into the pathogenesis of neurodegenerative diseases and suggest a promising role of circadian-based interventions.
Collapse
|
34
|
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
|
35
|
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
|
36
|
Vaithianathan T, Schneider EH, Bukiya AN, Dopico AM. Cholesterol and PIP 2 Modulation of BK Ca Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:217-243. [PMID: 36988883 PMCID: PMC10683925 DOI: 10.1007/978-3-031-21547-6_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Ca2+/voltage-gated, large conductance K+ channels (BKCa) are formed by homotetrameric association of α (slo1) subunits. Their activity, however, is suited to tissue-specific physiology largely due to their association with regulatory subunits (β and γ types), chaperone proteins, localized signaling, and the channel's lipid microenvironment. PIP2 and cholesterol can modulate BKCa activity independently of downstream signaling, yet activating Ca2+i levels and regulatory subunits control ligand action. At physiological Ca2+i and voltages, cholesterol and PIP2 reduce and increase slo1 channel activity, respectively. Moreover, slo1 proteins provide sites that seem to recognize cholesterol and PIP2: seven CRAC motifs in the slo1 cytosolic tail and a string of positively charged residues (Arg329, Lys330, Lys331) immediately after S6, respectively. A model that could explain the modulation of BKCa activity by cholesterol and/or PIP2 is hypothesized. The roles of additional sites, whether in slo1 or BKCa regulatory subunits, for PIP2 and/or cholesterol to modulate BKCa function are also discussed.
Collapse
Affiliation(s)
- Thirumalini Vaithianathan
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Elizabeth H Schneider
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anna N Bukiya
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Alex M Dopico
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
37
|
Dai HR, Guo HL, Hu YH, Xu J, Ding XS, Cheng R, Chen F. Precision caffeine therapy for apnea of prematurity and circadian rhythms: New possibilities open up. Front Pharmacol 2022; 13:1053210. [PMID: 36532766 PMCID: PMC9753576 DOI: 10.3389/fphar.2022.1053210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/18/2022] [Indexed: 09/10/2024] Open
Abstract
Caffeine is the globally consumed psychoactive substance and the drug of choice for the treatment of apnea of prematurity (AOP), but its therapeutic effects are highly variable among preterm infants. Many of the molecular underpinnings of the marked individual response have remained elusive yet. Interestingly, the significant association between Clock gene polymorphisms and the response to caffeine therapy offers an opportunity to advance our understanding of potential mechanistic pathways. In this review, we delineate the functions and mechanisms of human circadian rhythms. An up-to-date advance of the formation and ontogeny of human circadian rhythms during the perinatal period are concisely discussed. Specially, we summarize and discuss the characteristics of circadian rhythms in preterm infants. Second, we discuss the role of caffeine consumption on the circadian rhythms in animal models and human, especially in neonates and preterm infants. Finally, we postulate how circadian-based therapeutic initiatives could open new possibilities to promote precision caffeine therapy for the AOP management in preterm infants.
Collapse
Affiliation(s)
- Hao-Ran Dai
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hong-Li Guo
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Ya-Hui Hu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Xu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan-Sheng Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rui Cheng
- Neonatal Intensive Care Unit, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Chen
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
38
|
Wang ZZ, Sun Z, Zhang ML, Xiong K, Zhou F. Relationship between shift work, night work, and subsequent dementia: A systematic evaluation and meta-analysis. Front Neurol 2022; 13:997181. [DOI: 10.3389/fneur.2022.997181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022] Open
Abstract
BackgroundThe association between shift work, night work, and the risk of dementia are conflicting in the literature.ObjectivesThis study was performed to obtain evidence on the potential relationship between shift work, night work, and dementia.MethodsTo investigate the link between shift work, night work, and dementia, we systematically searched PubMed, Embase, and Web of Science from inception to January 1, 2022. The eligibility of the retrieved records and extracted data were independently reviewed by two researchers. The Preferred Reporting Items for Systematic Evaluations and Meta-Analyses (PRISMA) statement was followed. STATA 16.0 was used to conduct the meta-analysis.ResultsA total of four studies involving 103,104 participants were included in this meta-analysis. Pooled results indicated that night shift workers had a 12% increased risk of dementia compared to subjects without night work (HR = 1.12, 95% CI 1.03–1.23, P = 0.094). Shift work was not significantly associated with dementia risk (HR: 1.09, 95% CI: 0.83–1.43, P = 0.546), but the effect of shift work on dementia risk appeared to increase with age, with a correlation observed among workers older than 50 years (HR = 1.31; 95% CI: 1.03–1.68, P = 0.030).ConclusionThe data presented in our study suggest that night work may be a risk factor for dementia. More prospective studies with objective exposure measurements are required to further confirm this result.Systematic review registrationhttps://doi.org/10.37766/inplasy2022.6.0079, identifier: INPLASY202260079.
Collapse
|
39
|
Slayden AV, Dyer CL, Ma D, Li W, Bukiya AN, Parrill AL, Dopico AM. Discovery of agonist-antagonist pairs for the modulation of Ca [2]+ and voltage-gated K + channels of large conductance that contain beta1 subunits. Bioorg Med Chem 2022; 68:116876. [PMID: 35716586 PMCID: PMC10464842 DOI: 10.1016/j.bmc.2022.116876] [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: 04/08/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 11/02/2022]
Abstract
Large conductance, calcium/voltage-gated potassium channels (BK) regulate critical body processes, including neuronal, secretory and smooth muscle (SM) function. While BK-forming alpha subunits are ubiquitous, accessory beta1 subunits are highly expressed in SM. This makes beta1 an attractive target for pharmaceutical development to treat SM disorders, such as hypertension or cerebrovascular spasm. Compounds activating BK via beta1 have been identified, yet they exhibit low potency and off-target effects while antagonists that limit agonist activity via beta 1 remain unexplored. Beta1-dependent BK ligand-based pharmacophore modeling and ZINC database searches identified 15 commercially available hits. Concentration-response curves on BK alpha + beta1 subunit-mediated currents were obtained in CHO cells. One potent (EC50 = 20 nM) and highly efficacious activator (maximal activation = ×10.3 of control) was identified along with a potent antagonist (KB = 3.02 nM), both of which were dependent on beta1. Our study provides the first proof-of-principle that an agonist/antagonist pair can be used to control beta1-containing BK activity.
Collapse
Affiliation(s)
- Alexandria V Slayden
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis TN, 38103, USA
| | - Christy L Dyer
- Department of Chemistry, The University of Memphis, Memphis TN, 38152, USA
| | - Dejian Ma
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis TN, 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis TN, 38163, USA
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis TN, 38103, USA
| | - Abby L Parrill
- Department of Chemistry, The University of Memphis, Memphis TN, 38152, USA
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis TN, 38103, USA.
| |
Collapse
|
40
|
Loss of Ca V1.3 RNA editing enhances mouse hippocampal plasticity, learning, and memory. Proc Natl Acad Sci U S A 2022; 119:e2203883119. [PMID: 35914168 PMCID: PMC9371748 DOI: 10.1073/pnas.2203883119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
L-type CaV1.3 calcium channels are expressed on the dendrites and soma of neurons, and there is a paucity of information about its role in hippocampal plasticity. Here, by genetic targeting to ablate CaV1.3 RNA editing, we demonstrate that unedited CaV1.3ΔECS mice exhibited improved learning and enhanced long-term memory, supporting a functional role of RNA editing in behavior. Significantly, the editing paradox that functional recoding of CaV1.3 RNA editing sites slows Ca2+-dependent inactivation to increase Ca2+ influx but reduces channel open probability to decrease Ca2+ influx was resolved. Mechanistically, using hippocampal slice recordings, we provide evidence that unedited CaV1.3 channels permitted larger Ca2+ influx into the hippocampal pyramidal neurons to bolster neuronal excitability, synaptic transmission, late long-term potentiation, and increased dendritic arborization. Of note, RNA editing of the CaV1.3 IQ-domain was found to be evolutionarily conserved in mammals, which lends support to the importance of the functional recoding of the CaV1.3 channel in brain function.
Collapse
|
41
|
Fagiani F, Baronchelli E, Pittaluga A, Pedrini E, Scacchi C, Govoni S, Lanni C. The Circadian Molecular Machinery in CNS Cells: A Fine Tuner of Neuronal and Glial Activity With Space/Time Resolution. Front Mol Neurosci 2022; 15:937174. [PMID: 35845604 PMCID: PMC9283971 DOI: 10.3389/fnmol.2022.937174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/07/2022] [Indexed: 11/24/2022] Open
Abstract
The circadian molecular machinery is a fine timekeeper with the capacity to harmonize physiological and behavioral processes with the external environment. This tight-knit regulation is coordinated by multiple cellular clocks across the body. In this review, we focus our attention on the molecular mechanisms regulated by the clock in different brain areas and within different cells of the central nervous system. Further, we discuss evidence regarding the role of circadian rhythms in the regulation of neuronal activity and neurotransmitter systems. Not only neurons, but also astrocytes and microglia actively participate in the maintenance of timekeeping within the brain, and the diffusion of circadian information among these cells is fine-tuned by neurotransmitters (e.g., dopamine, serotonin, and γ-aminobutyric acid), thus impacting on the core clock machinery. The bidirectional interplay between neurotransmitters and the circadian clockwork is fundamental in maintaining accuracy and precision in daily timekeeping throughout different brain areas. Deepening the knowledge of these correlations allows us to define the basis of drug interventions to restore circadian rhythms, as well as to predict the onset of drug treatment/side effects that might promote daily desynchronization. Furthermore, it may lead to a deeper understanding of the potential impacts of modulations in rhythmic activities on the pace of aging and provide an insight in to the pathogenesis of psychiatric diseases and neurodegenerative disorders.
Collapse
Affiliation(s)
- Francesca Fagiani
- Institute of Experimental Neurology, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Eva Baronchelli
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Anna Pittaluga
- Department of Pharmacy (DiFar), School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, 3Rs Center, University of Genoa, Genoa, Italy
| | - Edoardo Pedrini
- Institute of Experimental Neurology, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Chiara Scacchi
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Stefano Govoni
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Cristina Lanni
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
- Centro 3R (Inter-University Center for the Promotion of the 3Rs Principles in Teaching and Research), Italy
- *Correspondence: Cristina Lanni
| |
Collapse
|
42
|
Ma Z, Eaton M, Liu Y, Zhang J, Chen X, Tu X, Shi Y, Que Z, Wettschurack K, Zhang Z, Shi R, Chen Y, Kimbrough A, Lanman NA, Schust L, Huang Z, Yang Y. Deficiency of autism-related Scn2a gene in mice disrupts sleep patterns and circadian rhythms. Neurobiol Dis 2022; 168:105690. [PMID: 35301122 PMCID: PMC9018617 DOI: 10.1016/j.nbd.2022.105690] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/21/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) affects ~2% of the population in the US, and monogenic forms of ASD often result in the most severe manifestation of the disorder. Recently, SCN2A has emerged as a leading gene associated with ASD, of which abnormal sleep pattern is a common comorbidity. SCN2A encodes the voltage-gated sodium channel NaV1.2. Predominantly expressed in the brain, NaV1.2 mediates the action potential firing of neurons. Clinical studies found that a large portion of children with SCN2A deficiency have sleep disorders, which severely impact the quality of life of affected individuals and their caregivers. The underlying mechanism of sleep disturbances related to NaV1.2 deficiency, however, is not known. Using a gene-trap Scn2a-deficient mouse model (Scn2atrap), we found that Scn2a deficiency results in increased wakefulness and reduced non-rapid-eye-movement (NREM) sleep. Brain region-specific Scn2a deficiency in the suprachiasmatic nucleus (SCN) containing region, which is involved in circadian rhythms, partially recapitulates the sleep disturbance phenotypes. At the cellular level, we found that Scn2a deficiency disrupted the firing pattern of spontaneously firing neurons in the SCN region. At the molecular level, RNA-sequencing analysis revealed differentially expressed genes in the circadian entrainment pathway including core clock genes Per1 and Per2. Performing a transcriptome-based compound discovery, we identified dexanabinol (HU-211), a putative glutamate receptor modulator, that can partially reverse the sleep disturbance in mice. Overall, our study reveals possible molecular and cellular mechanisms underlying Scn2a deficiency-related sleep disturbances, which may inform the development of potential pharmacogenetic interventions for the affected individuals.
Collapse
Affiliation(s)
- Zhixiong Ma
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China; Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Muriel Eaton
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Yushuang Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Jingliang Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Xiaoling Chen
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Xinyu Tu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yiqiang Shi
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zhefu Que
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Kyle Wettschurack
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Zaiyang Zhang
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Yueyi Chen
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Adam Kimbrough
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Nadia A Lanman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Leah Schust
- FamilieSCN2A Foundation, P.O. Box 82, East Longmeadow, MA 01028, USA
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA.
| |
Collapse
|
43
|
Hoyt KR, Obrietan K. Circadian clocks, cognition, and Alzheimer's disease: synaptic mechanisms, signaling effectors, and chronotherapeutics. Mol Neurodegener 2022; 17:35. [PMID: 35525980 PMCID: PMC9078023 DOI: 10.1186/s13024-022-00537-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/08/2022] [Indexed: 12/20/2022] Open
Abstract
Modulation of basic biochemical and physiological processes by the circadian timing system is now recognized as a fundamental feature of all mammalian organ systems. Within the central nervous system, these clock-modulating effects are reflected in some of the most complex behavioral states including learning, memory, and mood. How the clock shapes these behavioral processes is only now beginning to be realized. In this review we describe recent findings regarding the complex set of cellular signaling events, including kinase pathways, gene networks, and synaptic circuits that are under the influence of the clock timing system and how this, in turn, shapes cognitive capacity over the circadian cycle. Further, we discuss the functional roles of the master circadian clock located in the suprachiasmatic nucleus, and peripheral oscillator populations within cortical and limbic circuits, in the gating of synaptic plasticity and memory over the circadian cycle. These findings are then used as the basis to discuss the connection between clock dysregulation and cognitive impairments resulting from Alzheimer's disease (AD). In addition, we discuss the conceptually novel idea that in AD, there is a selective disruption of circadian timing within cortical and limbic circuits, and that it is the disruption/desynchronization of these regions from the phase-entraining effects of the SCN that underlies aspects of the early- and mid-stage cognitive deficits in AD. Further, we discuss the prospect that the disruption of circadian timing in AD could produce a self-reinforcing feedback loop, where disruption of timing accelerates AD pathogenesis (e.g., amyloid deposition, oxidative stress and cell death) that in turn leads to a further disruption of the circadian timing system. Lastly, we address potential therapeutic approaches that could be used to strengthen cellular timing networks and, in turn, how these approaches could be used to improve cognitive capacity in Alzheimer's patients.
Collapse
Affiliation(s)
- Kari R Hoyt
- Division of Pharmaceutics and Pharmacology, Ohio State University, 412 Riffe Building, 12th Ave, Columbus, OH, 43210, USA.
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Graves Hall, 333 W. 10th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
44
|
Schroder EA, Ono M, Johnson SR, Rozmus ER, Burgess DE, Esser KA, Delisle BP. The role of the cardiomyocyte circadian clocks in ion channel regulation and cardiac electrophysiology. J Physiol 2022; 600:2037-2048. [PMID: 35301719 PMCID: PMC9980729 DOI: 10.1113/jp282402] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/04/2022] [Indexed: 11/08/2022] Open
Abstract
Daily variations in cardiac electrophysiology and the incidence for different types of arrhythmias reflect ≈24 h changes in the environment, behaviour and internal circadian rhythms. This article focuses on studies that use animal models to separate the impact that circadian rhythms, as well as changes in the environment and behaviour, have on 24 h rhythms in heart rate and ventricular repolarization. Circadian rhythms are initiated at the cellular level by circadian clocks, transcription-translation feedback loops that cycle with a periodicity of 24 h. Several studies now show that the circadian clock in cardiomyocytes regulates the expression of cardiac ion channels by multiple mechanisms; underlies time-of-day changes in sinoatrial node excitability/intrinsic heart rate; and limits the duration of the ventricular action potential waveform. However, the 24 h rhythms in heart rate and ventricular repolarization are primarily driven by autonomic signalling. A functional role for the cardiomyocyte circadian clock appears to buffer the heart against perturbations. For example, the cardiomyocyte circadian clock limits QT-interval prolongation (especially at slower heart rates), and it may facilitate the realignment of the 24 h rhythm in heart rate to abrupt changes in the light cycle. Additional studies show that modifying rhythmic behaviours (including feeding behaviour) can dramatically impact the 24 h rhythms in heart rate and ventricular repolarization. If these mechanisms are conserved, these studies suggest that targeting endogenous circadian mechanisms in the heart, as well as modifying the timing of certain rhythmic behaviours, could emerge as therapeutic strategies to support heart function against perturbations and regulate 24 h rhythms in cardiac electrophysiology.
Collapse
Affiliation(s)
- Elizabeth A. Schroder
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Kentucky, 740 S. Limestone Street, L543, Lexington, KY 40536-0284
| | - Makoto Ono
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
| | - Sidney R. Johnson
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
| | - Ezekiel R. Rozmus
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
| | - Don E. Burgess
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
| | - Karyn A. Esser
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Brian P. Delisle
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
| |
Collapse
|
45
|
Schellinger JN, Sun Q, Pleinis JM, An SW, Hu J, Mercenne G, Titos I, Huang CL, Rothenfluh A, Rodan AR. Chloride oscillation in pacemaker neurons regulates circadian rhythms through a chloride-sensing WNK kinase signaling cascade. Curr Biol 2022; 32:1429-1438.e6. [PMID: 35303418 PMCID: PMC8972083 DOI: 10.1016/j.cub.2022.03.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/02/2021] [Accepted: 03/04/2022] [Indexed: 12/21/2022]
Abstract
Central pacemaker neurons regulate circadian rhythms and undergo diurnal variation in electrical activity in mammals and flies.1,2 Circadian variation in the intracellular chloride concentration of mammalian pacemaker neurons has been proposed to influence the response to GABAergic neurotransmission through GABAA receptor chloride channels.3 However, results have been contradictory,4-9 and a recent study demonstrated circadian variation in pacemaker neuron chloride without an effect on GABA response.10 Therefore, whether and how intracellular chloride regulates circadian rhythms remains controversial. Here, we demonstrate a signaling role for intracellular chloride in the Drosophila small ventral lateral (sLNv) pacemaker neurons. In control flies, intracellular chloride increases in sLNvs over the course of the morning. Chloride transport through sodium-potassium-2-chloride (NKCC) and potassium-chloride (KCC) cotransporters is a major determinant of intracellular chloride concentrations.11Drosophila melanogaster with loss-of-function mutations in the NKCC encoded by Ncc69 have abnormally low intracellular chloride 6 h after lights on, loss of morning anticipation, and a prolonged circadian period. Loss of kcc, which is expected to increase intracellular chloride, suppresses the long-period phenotype of Ncc69 mutant flies. Activation of a chloride-inhibited kinase cascade, consisting of WNK (with no lysine [K]) kinase and its downstream substrate, Fray, is necessary and sufficient to prolong period length. Fray activation of an inwardly rectifying potassium channel, Irk1, is also required for the long-period phenotype. These results indicate that the NKCC-dependent rise in intracellular chloride in Drosophila sLNv pacemakers restrains WNK-Fray signaling and overactivation of an inwardly rectifying potassium channel to maintain normal circadian period length.
Collapse
Affiliation(s)
- Jeffrey N Schellinger
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Qifei Sun
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern, Dallas, TX 75390, USA
| | - John M Pleinis
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Sung-Wan An
- Department of Internal Medicine, Division of Nephrology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Jianrui Hu
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Gaëlle Mercenne
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Iris Titos
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Chou-Long Huang
- Department of Internal Medicine, Division of Nephrology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Adrian Rothenfluh
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; Department of Psychiatry, Huntsman Mental Health Institute, University of Utah, Salt Lake City, UT 84108, USA; Department of Neurobiology, University of Utah, Salt Lake City, UT 84112, USA; Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA; Department of Internal Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, UT 84132, USA; Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT 84148, USA.
| |
Collapse
|
46
|
Sun Z, Yuan W, Li L, Cai H, Mao X, Zhang L, Zang G, Wang Z. Macrophage CD36 and TLR4 Cooperation Promotes Foam Cell Formation and VSMC Migration and Proliferation Under Circadian Oscillations. J Cardiovasc Transl Res 2022; 15:985-997. [PMID: 35257279 DOI: 10.1007/s12265-022-10225-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/21/2022] [Indexed: 11/29/2022]
Abstract
Circadian rhythm disorders can accelerate atherosclerosis. This study aimed to determine the role of circadian disordered macrophages in atherosclerotic development. Mice were divided into NC group (normal circadian rhythm), L24 group (constant light), D12L12 group (weekly shift light/dark cycle), and D24 group (constant dark). Atherosclerotic progression was significantly accelerated in L24, D12L12, and D24 groups. Peritoneal macrophages from circadian disruption groups exhibited enhanced cytokine secretion and foam cell formation. Migration and proliferation of vascular smooth muscle cells (VSMCs) were increased under the conditioned medium of circadian disordered macrophages. The blockade of CD36 markedly inhibited foam cell formation. Compared with blocking CD36 or TLR4 alone, the co-inhibition of CD36 and TLR4 in macrophages further reduced cytokine secretion and more effectively inhibited VSMC migration and proliferation. In conclusion, the activation of CD36 and TLR4 in circadian disordered macrophages promotes foam cell formation and cytokine secretion and enhances VSMC migration and proliferation. Circadian rhythm disorders promote lipid uptake and cytokine secretion of macrophages by regulating CD36 and TLR4, and enhance VSMC migration and proliferation through the paracrine effect of macrophages.
Collapse
Affiliation(s)
- Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Honghua Cai
- Department of Burn Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xiang Mao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Guangyao Zang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
47
|
Ion Channel Partnerships: Odd and Not-So-Odd Couples Controlling Neuronal Ion Channel Function. Int J Mol Sci 2022; 23:ijms23041953. [PMID: 35216068 PMCID: PMC8878034 DOI: 10.3390/ijms23041953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
The concerted function of the large number of ion channels expressed in excitable cells, including brain neurons, shapes diverse signaling events by controlling the electrical properties of membranes. It has long been recognized that specific groups of ion channels are functionally coupled in mediating ionic fluxes that impact membrane potential, and that these changes in membrane potential impact ion channel gating. Recent studies have identified distinct sets of ion channels that can also physically and functionally associate to regulate the function of either ion channel partner beyond that afforded by changes in membrane potential alone. Here, we review canonical examples of such ion channel partnerships, in which a Ca2+ channel is partnered with a Ca2+-activated K+ channel to provide a dedicated route for efficient coupling of Ca2+ influx to K+ channel activation. We also highlight examples of non-canonical ion channel partnerships between Ca2+ channels and voltage-gated K+ channels that are not intrinsically Ca2+ sensitive, but whose partnership nonetheless yields enhanced regulation of one or the other ion channel partner. We also discuss how these ion channel partnerships can be shaped by the subcellular compartments in which they are found and provide perspectives on how recent advances in techniques to identify proteins in close proximity to one another in native cells may lead to an expanded knowledge of other ion channel partnerships.
Collapse
|
48
|
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
|
49
|
Mei L, Zheng Y, Ma T, Xia B, Gao X, Hao Y, Luo Z, Huang J. (-)-Epigallocatechin-3-gallate Ameliorates Intervertebral Disc Degeneration Through Reprogramming of the Circadian Clock. Front Pharmacol 2021; 12:753548. [PMID: 34803694 PMCID: PMC8599576 DOI: 10.3389/fphar.2021.753548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/13/2021] [Indexed: 12/15/2022] Open
Abstract
The circadian clock is vital in the management of our daily physiological as well as metabolic processes. Disturbances of the clock can cause degenerative and age-related diseases. Increasing evidence has indicated that the intervertebral discs contain an internal biological clock related to degeneration. However, to date, no bioactive compounds have been found that can ameliorate intervertebral disc degeneration (IDD) by restoring the circadian clock. (-)-Epigallocatechin-3-gallate (EGCG) is a nutritious food with powerful antioxidant properties, as well as entraining biological clock to improve health. The purpose of this study was to determine whether the protective effects of EGCG on nucleus pulposus (NPCs) under oxidative stress is related to the circadian clock. First, we found that EGCG attenuated H2O2-induced extracellular matrix degradation in NPCs and inhibited H2O2-induced NPC apoptosis. Our in vivo experiments also confirmed this finding. Furthermore, EGCG attenuated H2O2-triggered dampening of phase shifts and daily oscillations in circadian clock gene transcription as well as protein expression levels. Intriguingly, core clock gene (Bmal1) knockdown notably blocked the protective effects of EGCG. To our knowledge, this study provides the first convincing evidence that EGCG prevents IDD in a Bmal1-dependent manner. In general, EGCG supplementation can be used as a nutritional prevention strategy for the rehabilitation of degenerative diseases related to the circadian clock.
Collapse
Affiliation(s)
- Liangwei Mei
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| | - Yi Zheng
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| | - Teng Ma
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| | - Bing Xia
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| | - Xue Gao
- Faculty of Life Sciences, Northwest University, Shaanxi, China
| | - Yiming Hao
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| | - Zhuojing Luo
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| |
Collapse
|
50
|
Li B, Li D, Ni H, Liu C, Xiong J, Liu H, Gao R, Zhang L, Chen G. The circadian clock regulator Bmal1 affects traumatic brain injury in rats through the p38 MAPK signalling pathway. Brain Res Bull 2021; 178:17-28. [PMID: 34774994 DOI: 10.1016/j.brainresbull.2021.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 11/02/2022]
Abstract
Traumatic brain injury (TBI) is still one of the main causes of death and disability worldwide. Bmal1 (brain and muscle Arnt-like protein-1) is the most central factor of the circadian rhythms that control life and cells. Studies have shown that Bmal1 is involved in inflammation, oxidative stress, vasodilation, glucose and lipid metabolism. This study explored the effect of Bmal1 on secondary brain injury after TBI in rats and the possible mechanism. We established a rat model of TBI induced by the free fall of a weight in rats. The Western blotting and immunofluorescence results showed that the Bmal1 levels decreased in the cerebral cortex after TBI, especially at 48 h. The effects of Bmal1 levels on rats after TBI were evaluated by brain oedema measurement, adhesive removal tests, behavioural tests, and TUNEL and FJC staining. We found that the recombinant Bmal1 protein increased Bmal1 levels after TBI and reduced brain oedema, neurobehavioural injury, somatosensory disturbances, and nerve cell necrosis and apoptosis. The ELISA results showed that Bmal1 overexpression could reduce the inflammatory factors IL-4 and TNF-α after TBI. In contrast, inhibiting Bmal1 expression had the opposite effect. The changes in Bmal1 levels were closely related to the phosphorylation of p38 MAPK after TBI. In conclusion, a decrease in Bmal1 after TBI may exacerbate pathological symptoms in vivo by activating p38 MAPK phosphorylation.
Collapse
Affiliation(s)
- Bing Li
- Department of Neurosurgery, the Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Di Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Haibo Ni
- Department of Neurosurgery, the Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Chenglin Liu
- Department of Neurosurgery, the Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Jian Xiong
- Department of Rehabilitation, the Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Huixiang Liu
- Department of Neurosurgery, the Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Rong Gao
- Department of Neurosurgery, the Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China.
| | - Li Zhang
- Department of Neurosurgery, the Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, the First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|