1
|
Kozlova EV, Bishay AE, Denys ME, Chinthirla BD, Valdez MC, Spurgin KA, Krum JM, Basappa KR, Currás-Collazo MC. Gene deletion of the PACAP/VIP receptor, VPAC2R, alters glycemic responses during metabolic and psychogenic stress in adult female mice. J Neuroendocrinol 2023; 35:e13354. [PMID: 37946684 DOI: 10.1111/jne.13354] [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: 05/03/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 11/12/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and the homologous peptide, vasoactive intestinal peptide (VIP), participate in glucose homeostasis using insulinotropic and counterregulatory processes. The role of VIP receptor 2 (VPAC2R) in these opposing actions needs further characterization. In this study, we examined the participation of VPAC2R on basal glycemia, fasted levels of glucoregulatory hormones and on glycemia responses during metabolic and psychogenic stress using gene-deleted (Vipr2-/- ) female mice. The mean basal glycemia was significantly greater in Vipr2-/- in the fed state and after an 8-h overnight fast as compared to wild-type (WT) mice. Insulin tolerance testing following a 5-h fast (morning fast, 0.38 U/kg insulin) indicated no effect of genotype. However, during a more intense metabolic challenge (8 h, ON fast, 0.25 U/kg insulin), Vipr2-/- females displayed significantly impaired insulin hypoglycemia. During immobilization stress, the hyperglycemic response and plasma epinephrine levels were significantly elevated above basal in Vipr2-/- , but not WT mice, in spite of similar stress levels of plasma corticosterone. Together, these results implicate participation of VPAC2R in upregulated counterregulatory processes influenced by enhanced sympathoexcitation. Moreover, the suppression of plasma GLP-1 levels in Vipr2-/- mice may have removed the inhibition on hepatic glucose production and the promotion of glucose disposal by GLP-1. qPCR analysis indicated deregulation of central gene markers of PACAP/VIP signaling in Vipr2-/- , upregulated medulla tyrosine hydroxylase (Th) and downregulated hypothalamic Vip transcripts. These results demonstrate a physiological role for VPAC2R in glucose metabolism, especially during insulin challenge and psychogenic stress, likely involving the participation of sympathoadrenal activity and/or metabolic hormones.
Collapse
Affiliation(s)
- Elena V Kozlova
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
- Neuroscience Graduate Program, University of California, Riverside, California, USA
| | - Anthony E Bishay
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Maximilian E Denys
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Bhuvaneswari D Chinthirla
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Matthew C Valdez
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
- Neuroscience Graduate Program, University of California, Riverside, California, USA
| | - Kurt A Spurgin
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
- Neuroscience Graduate Program, University of California, Riverside, California, USA
| | - Julia M Krum
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Karthik R Basappa
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | | |
Collapse
|
2
|
Yin J, Zhou J, Fang F, Yu S, Wang J, Yuan J, Zhou Z. Identification of VIPR2 rare and common variants in the Chinese Han population with schizophrenia. Front Mol Neurosci 2023; 16:1170708. [PMID: 37181653 PMCID: PMC10174236 DOI: 10.3389/fnmol.2023.1170708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/07/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction Schizophrenia is a severe and chronic psychiatric disorder with hereditary risk up to 80% as previous studies indicated. Several researches have demonstrated a significant association between schizophrenia and microduplications that overlap the vasoactive intestinal peptide receptor 2 gene (VIPR2). Methods To further investigate potential causal VIPR2 gene variants, all exons and un-translated portions of the VIPR2 gene were sequenced using amplicon targeted resequencing in 1804 Chinese Han patients with schizophrenia and 996 healthy counterparts in the present study. Results Nineteen rare non-synonymous mutations and 1 frameshift deletion was identified for schizophrenia, among which 5 variants have never been reported so far. Frequencies of rare non-synonymous mutations were significantly different between the two groups. Specifically, the non-synonymous mutation rs78564798 (Pallele = 0.006) as well as two rare variations in the VIPR2 gene's introns (rs372544903, Pallele = 0.026 and a novel mutation, chr7:159034078, GRCh38, Pallele = 0.048) were significantly associated with schizophrenia. Discussion Our findings add new evidence that the functional and probable causative variants of VIPR2 gene may play an important role in susceptibility to schizophrenia. Further studies on validations of VIPR2's function in the etiology of schizophrenia are warranted.
Collapse
Affiliation(s)
- Jiajun Yin
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
| | - Juan Zhou
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Fang Fang
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
| | - Shui Yu
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
| | - Jun Wang
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
| | - Jianmin Yuan
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
- *Correspondence: Jianmin Yuan,
| | - Zhenhe Zhou
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
- Zhenhe Zhou,
| |
Collapse
|
3
|
Abstract
Our physiology and behavior follow precise daily programs that adapt us to the alternating opportunities and challenges of day and night. Under experimental isolation, these rhythms persist with a period of approximately one day (circadian), demonstrating their control by an internal autonomous clock. Circadian time is created at the cellular level by a transcriptional/translational feedback loop (TTFL) in which the protein products of the Period and Cryptochrome genes inhibit their own transcription. Because the accumulation of protein is slow and delayed, the system oscillates spontaneously with a period of ∼24 hours. This cell-autonomous TTFL controls cycles of gene expression in all major tissues and these cycles underpin our daily metabolic programs. In turn, our innumerable cellular clocks are coordinated by a central pacemaker, the suprachiasmatic nucleus (SCN) of the hypothalamus. When isolated in slice culture, the SCN TTFL and its dependent cycles of neural activity persist indefinitely, operating as "a clock in a dish". In vivo, SCN time is synchronized to solar time by direct innervation from specialized retinal photoreceptors. In turn, the precise circadian cycle of action potential firing signals SCN-generated time to hypothalamic and brain stem targets, which co-ordinate downstream autonomic, endocrine, and behavioral (feeding) cues to synchronize and sustain the distributed cellular clock network. Circadian time therefore pervades every level of biological organization, from molecules to society. Understanding its mechanisms offers important opportunities to mitigate the consequences of circadian disruption, so prevalent in modern societies, that arise from shiftwork, aging, and neurodegenerative diseases, not least Huntington's disease.
Collapse
Affiliation(s)
- Andrew P. Patton
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | |
Collapse
|
4
|
Singla R, Mishra A, Cao R. The trilateral interactions between mammalian target of rapamycin (mTOR) signaling, the circadian clock, and psychiatric disorders: an emerging model. Transl Psychiatry 2022; 12:355. [PMID: 36045116 PMCID: PMC9433414 DOI: 10.1038/s41398-022-02120-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 02/07/2023] Open
Abstract
Circadian (~24 h) rhythms in physiology and behavior are evolutionarily conserved and found in almost all living organisms. The rhythms are endogenously driven by daily oscillatory activities of so-called "clock genes/proteins", which are widely distributed throughout the mammalian brain. Mammalian (mechanistic) target of rapamycin (mTOR) signaling is a fundamental intracellular signal transduction cascade that controls important neuronal processes including neurodevelopment, synaptic plasticity, metabolism, and aging. Dysregulation of the mTOR pathway is associated with psychiatric disorders including autism spectrum disorders (ASD) and mood disorders (MD), in which patients often exhibit disrupted daily physiological rhythms and abnormal circadian gene expression in the brain. Recent work has found that the activities of mTOR signaling are temporally controlled by the circadian clock and exhibit robust circadian oscillations in multiple systems. In the meantime, mTOR signaling regulates fundamental properties of the central and peripheral circadian clocks, including period length, entrainment, and synchronization. Whereas the underlying mechanisms remain to be fully elucidated, increasing clinical and preclinical evidence support significant crosstalk between mTOR signaling, the circadian clock, and psychiatric disorders. Here, we review recent progress in understanding the trilateral interactions and propose an "interaction triangle" model between mTOR signaling, the circadian clock, and psychiatric disorders (focusing on ASD and MD).
Collapse
Affiliation(s)
- Rubal Singla
- grid.17635.360000000419368657Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812 USA
| | - Abhishek Mishra
- grid.17635.360000000419368657Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812 USA
| | - Ruifeng Cao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA. .,Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| |
Collapse
|
5
|
Lu J, Piper SJ, Zhao P, Miller LJ, Wootten D, Sexton PM. Targeting VIP and PACAP Receptor Signaling: New Insights into Designing Drugs for the PACAP Subfamily of Receptors. Int J Mol Sci 2022; 23:8069. [PMID: 35897648 PMCID: PMC9331257 DOI: 10.3390/ijms23158069] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/16/2022] Open
Abstract
Pituitary Adenylate Cyclase-Activating Peptide (PACAP) and Vasoactive Intestinal Peptide (VIP) are neuropeptides involved in a diverse array of physiological and pathological processes through activating the PACAP subfamily of class B1 G protein-coupled receptors (GPCRs): VIP receptor 1 (VPAC1R), VIP receptor 2 (VPAC2R), and PACAP type I receptor (PAC1R). VIP and PACAP share nearly 70% amino acid sequence identity, while their receptors PAC1R, VPAC1R, and VPAC2R share 60% homology in the transmembrane regions of the receptor. PACAP binds with high affinity to all three receptors, while VIP binds with high affinity to VPAC1R and VPAC2R, and has a thousand-fold lower affinity for PAC1R compared to PACAP. Due to the wide distribution of VIP and PACAP receptors in the body, potential therapeutic applications of drugs targeting these receptors, as well as expected undesired side effects, are numerous. Designing selective therapeutics targeting these receptors remains challenging due to their structural similarities. This review discusses recent discoveries on the molecular mechanisms involved in the selectivity and signaling of the PACAP subfamily of receptors, and future considerations for therapeutic targeting.
Collapse
Affiliation(s)
- Jessica Lu
- Drug Discovery Biology, Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.L.); (S.J.P.); (P.Z.)
| | - Sarah J. Piper
- Drug Discovery Biology, Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.L.); (S.J.P.); (P.Z.)
| | - Peishen Zhao
- Drug Discovery Biology, Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.L.); (S.J.P.); (P.Z.)
| | - Laurence J. Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, USA;
| | - Denise Wootten
- Drug Discovery Biology, Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.L.); (S.J.P.); (P.Z.)
| | - Patrick M. Sexton
- Drug Discovery Biology, Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.L.); (S.J.P.); (P.Z.)
| |
Collapse
|
6
|
Vijaya Shankara J, Horsley KG, Cheng N, Rho JM, Antle MC. Circadian Responses to Light in the BTBR Mouse. J Biol Rhythms 2022; 37:498-515. [PMID: 35722987 PMCID: PMC9452857 DOI: 10.1177/07487304221102279] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Animals with altered freerunning periods are valuable in understanding properties of the circadian clock. Understanding the relationship between endogenous clock properties, entrainment, and influence of light in terms of parametric and non-parametric models can help us better understand how different populations adapt to external light cycles. Many clinical populations often show significant changes in circadian properties that in turn cause sleep and circadian problems, possibly exacerbating their underlying clinical condition. BTBR T+Itpr3tf/J (BTBR) mice are a model commonly used for the study of autism spectrum disorders (ASD). Adults and adolescents with ASD frequently exhibit profound sleep and circadian disruptions, including increased latency to sleep, insomnia, advanced and delayed sleep phase disorders, and sleep fragmentation. Here, we investigated the circadian phenotype of BTBR mice in freerunning and light-entrained conditions and found that this strain of mice showed noticeably short freerunning periods (~22.75 h). In addition, when compared to C57BL/6J controls, BTBR mice also showed higher levels of activity even though this activity was compressed into a shorter active phase. Phase delays and phase advances to light were significantly larger in BTBR mice. Despite the short freerunning period, BTBR mice exhibited normal entrainment in light-dark cycles and accelerated entrainment to both advanced and delayed light cycles. Their ability to entrain to skeleton photoperiods of 1 min suggests that this entrainment cannot be attributed to masking. Period differences were also correlated with differences in the number of vasoactive intestinal polypeptide–expressing cells in the suprachiasmatic nucleus (SCN). Overall, the BTBR model, with their unique freerunning and entrainment properties, makes an interesting model to understand the underlying circadian clock.
Collapse
Affiliation(s)
- Jhenkruthi Vijaya Shankara
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Katelyn G Horsley
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ning Cheng
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Jong M Rho
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Departments of Neurosciences and Pediatrics, University of California, San Diego and Rady Children's Hospital, San Diego, California, USA
| | - Michael C Antle
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
7
|
Sakamoto K, Chen L, Miyaoka T, Yamada M, Masutani T, Ishimoto K, Hino N, Nakagawa S, Asano S, Ago Y. Generation of KS-133 as a Novel Bicyclic Peptide with a Potent and Selective VIPR2 Antagonist Activity that Counteracts Cognitive Decline in a Mouse Model of Psychiatric Disorders. Front Pharmacol 2021; 12:751587. [PMID: 34819858 PMCID: PMC8607231 DOI: 10.3389/fphar.2021.751587] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Worldwide, more than 20 million people suffer from schizophrenia, but effective and definitive new therapeutic drugs/treatments have not been established. Vasoactive intestinal peptide receptor 2 (VIPR2) might be an attractive drug target for the treatment of schizophrenia because both preclinical and clinical studies have demonstrated a strong link between high expression/overactivation of VIPR2 and schizophrenia. Nevertheless, VIPR2-targeting drugs are not yet available. VIPR2 is a class-B G protein-coupled receptor that possesses high structural homology to its subtypes, vasoactive intestinal peptide receptor 1 (VIPR1) and pituitary adenylate cyclase-activating polypeptide type-1 receptor (PAC1). These biological and structural properties have made it difficult to discover small molecule drugs against VIPR2. In 2018, cyclic peptide VIpep-3, a VIPR2-selective antagonist, was reported. The aim of this study was to generate a VIpep-3 derivative for in vivo experiments. After amino acid substitution and structure optimization, we successfully generated KS-133 with 1) a VIPR2-selective and potent antagonistic activity, 2) at least 24 h of stability in plasma, and 3) in vivo pharmacological efficacies in a mouse model of psychiatric disorders through early postnatal activation of VIPR2. To the best of our knowledge, this is the first report of a VIPR2-selective antagonistic peptide that counteracts cognitive decline, a central feature of schizophrenia. KS-133 may contribute to studies and development of novel schizophrenia therapeutic drugs that target VIPR2.
Collapse
Affiliation(s)
- Kotaro Sakamoto
- Research and Development Department, Ichimaru Pharcos Company Limited, Gifu, Japan
| | - Lu Chen
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Tatsunori Miyaoka
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Mei Yamada
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Teruaki Masutani
- Research and Development Department, Ichimaru Pharcos Company Limited, Gifu, Japan
| | - Kenji Ishimoto
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Innovative Food Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| | - Nobumasa Hino
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Shinsaku Nakagawa
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Innovative Food Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| | - Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukio Ago
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Innovative Food Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan.,Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
8
|
Ago Y, Asano S, Hashimoto H, Waschek JA. Probing the VIPR2 Microduplication Linkage to Schizophrenia in Animal and Cellular Models. Front Neurosci 2021; 15:717490. [PMID: 34366784 PMCID: PMC8339898 DOI: 10.3389/fnins.2021.717490] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/05/2021] [Indexed: 01/30/2023] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP, gene name ADCYAP1) is a multifunctional neuropeptide involved in brain development and synaptic plasticity. With respect to PACAP function, most attention has been given to that mediated by its specific receptor PAC1 (ADCYAP1R1). However, PACAP also binds tightly to the high affinity receptors for vasoactive intestinal peptide (VIP, VIP), called VPAC1 and VPAC2 (VIPR1 and VIPR2, respectively). Depending on innervation patterns, PACAP can thus interact physiologically with any of these receptors. VPAC2 receptors, the focus of this review, are known to have a pivotal role in regulating circadian rhythms and to affect multiple other processes in the brain, including those involved in fear cognition. Accumulating evidence in human genetics indicates that microduplications at 7q36.3, containing VIPR2 gene, are linked to schizophrenia and possibly autism spectrum disorder. Although detailed molecular mechanisms have not been fully elucidated, recent studies in animal models suggest that overactivation of the VPAC2 receptor disrupts cortical circuit maturation. The VIPR2 linkage can thus be potentially explained by inappropriate control of receptor signaling at a time when neural circuits involved in cognition and social behavior are being established. Alternatively, or in addition, VPAC2 receptor overactivity may disrupt ongoing synaptic plasticity during processes of learning and memory. Finally, in vitro data indicate that PACAP and VIP have differential activities on the maturation of neurons via their distinct signaling pathways. Thus perturbations in the balance of VPAC2, VPAC1, and PAC1 receptors and their ligands may have important consequences in brain development and plasticity.
Collapse
Affiliation(s)
- Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan.,Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Japan.,Division of Bioscience, Institute for Datability Science, Osaka University, Suita, Japan.,Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - James A Waschek
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
9
|
Ruel J, Guitton MJ, Gratias P, Lenoir M, Shen S, Puel JL, Brabet P, Wang J. Endogenous Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Plays a Protective Effect Against Noise-Induced Hearing Loss. Front Cell Neurosci 2021; 15:658990. [PMID: 33828461 PMCID: PMC8019930 DOI: 10.3389/fncel.2021.658990] [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] [Received: 01/26/2021] [Accepted: 03/02/2021] [Indexed: 01/07/2023] Open
Abstract
Pituitary adenylyl cyclase-activating polypeptide (PACAP) is a member of the vasoactive intestinal polypeptide (VIP)-the secretin-glucagon family of neuropeptides. They act through two classes of receptors: PACAP type 1 (PAC1) and type 2 (VPAC1 and VPAC2). Among their pleiotropic effects throughout the body, PACAP functions as neuromodulators and neuroprotectors, rescuing neurons from apoptosis, mostly through the PAC1 receptor. To explore the potential protective effect of endogenous PACAP against Noise-induced hearing loss (NIHL), we used a knockout mouse model lacking PAC1 receptor expression (PACR1−/−) and a transgenic humanized mouse model expressing the human PAC1 receptor (TgHPAC1R). Based on complementary approaches combining electrophysiological, histochemical, and molecular biological evaluations, we show PAC1R expression in spiral ganglion neurons and in cochlear apical cells of the organ of Corti. Wild-type (WT), PAC1R−/−, and TgHPAC1R mice exhibit similar auditory thresholds. For most of the frequencies tested after acute noise damage, however, PAC1R−/− mice showed a larger elevation of the auditory threshold than did their WT counterparts. By contrast, in a transgene copy number-dependent fashion, TgHPAC1R mice showed smaller noise-induced elevations of auditory thresholds compared to their WT counterparts. Together, these findings suggest that PACAP could be a candidate for endogenous protection against noise-induced hearing loss.
Collapse
Affiliation(s)
- Jérôme Ruel
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France.,Laboratoire de Neurosciences Cognitives, UMR7291 CNRS, Aix-Marseille Université, Marseille, France
| | - Matthieu J Guitton
- CERVO Brain Research Center, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Paul Gratias
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France
| | - Marc Lenoir
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France
| | - Sanbing Shen
- Regenerative Medicine Institute, National University of Ireland (NUI), Galway, Ireland
| | - Jean-Luc Puel
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France
| | - Philippe Brabet
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France
| | - Jing Wang
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France
| |
Collapse
|
10
|
Manoogian ENC, Kumar A, Obed D, Bergan J, Bittman EL. Suprachiasmatic function in a circadian period mutant: Duper alters light-induced activation of vasoactive intestinal peptide cells and PERIOD1 immunostaining. Eur J Neurosci 2019; 48:3319-3334. [PMID: 30346078 DOI: 10.1111/ejn.14214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 09/07/2018] [Accepted: 09/18/2018] [Indexed: 11/28/2022]
Abstract
Mammalian circadian rhythms are entrained by photic stimuli that are relayed by retinal projections to the core of the suprachiasmatic nucleus (SCN). Neuronal activation, as demonstrated by expression of the immediate early gene c-fos, leads to transcription of the core clock gene per1. The duper mutation in hamsters shortens circadian period and amplifies light-induced phase shifts. We performed two experiments to compare the number of c-FOS immunoreactive (ir) and PER1-ir cells, and the intensity of staining, in the SCN of wild-type (WT) and duper hamsters at various intervals after presentation of a 15-min light pulse in the early subjective night. Light-induced c-FOS-ir within 1 hr in the dorsocaudal SCN of duper, but not WT hamsters. In cells that express vasoactive intestinal peptide (VIP), which plays a critical role in synchronization of SCN cellular oscillators, light-induced c-FOS-ir was greater in duper than WT hamsters. After the light pulse, PER1-ir cells were found in more medial portions of the SCN than FOS-ir, and appeared with a longer latency and over a longer time course, in VIP cells of duper than wild-type hamsters. Our results indicate that the duper allele alters SCN function in ways that may contribute to changes in free running period and phase resetting.
Collapse
Affiliation(s)
- Emily N C Manoogian
- Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts
| | - Ajay Kumar
- Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts
| | - Doha Obed
- Department of Biology, University of Massachusetts, Amherst, Massachusetts
| | - Joseph Bergan
- Psychological and Brain Sciences and Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts
| | - Eric L Bittman
- Department of Biology, Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts
| |
Collapse
|
11
|
Lindberg PT, Mitchell JW, Burgoon PW, Beaulé C, Weihe E, Schäfer MKH, Eiden LE, Jiang SZ, Gillette MU. Pituitary Adenylate Cyclase-Activating Peptide (PACAP)-Glutamate Co-transmission Drives Circadian Phase-Advancing Responses to Intrinsically Photosensitive Retinal Ganglion Cell Projections by Suprachiasmatic Nucleus. Front Neurosci 2019; 13:1281. [PMID: 31866806 PMCID: PMC6909886 DOI: 10.3389/fnins.2019.01281] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 11/11/2019] [Indexed: 12/24/2022] Open
Abstract
Results from a variety of sources indicate a role for pituitary adenylate cyclase-activating polypeptide (PACAP) in light/glutamate-induced phase resetting of the circadian clock mediated by the retinohypothalamic tract (RHT). Attempts to block or remove PACAP’s contribution to clock-resetting have generated phenotypes that differ in their responses to light or glutamate. For example, previous studies of circadian behaviors found that period-maintenance and early-night phase delays are intact in PACAP-null mice, yet there is a consistent deficit in behavioral phase-resetting to light stimulation in the late night. Here we report rodent stimulus–response characteristics of PACAP release from the RHT, and map these to responses of the suprachiasmatic nucleus (SCN) in intact and PACAP-deficient mouse hypothalamus with regard to phase-resetting. SCN of PACAP-null mice exhibit normal circadian rhythms in neuronal activity, but are “blind” to glutamate stimulating phase-advance responses in late night, although not in early night, consistent with previously reported selective lack of late-night light behavioral responsiveness of these mice. Induction of CREB phosphorylation, a hallmark of the light/glutamate response of the SCN, also is absent in SCN-containing ex vivo slices from PACAP-deficient mouse hypothalamus. PACAP replacement to the SCN of PACAP-null mice restored wild-type phase-shifting of firing-rate patterns in response to glutamate applied to the SCN in late night. Likewise, ex vivo SCN of wild-type mice post-orbital enucleation are unresponsive to glutamate unless PACAP also is restored. Furthermore, we demonstrate that the period of efficacy of PACAP at SCN nerve terminals corresponds to waxing of PACAP mRNA expression in ipRGCs during the night, and waning during the day. These results validate the use of PACAP-deficient mice in defining the role and specificity of PACAP as a co-transmitter with glutamate in ipRGC-RHT projections to SCN in phase advancing the SCN circadian rhythm in late night.
Collapse
Affiliation(s)
- Peder T Lindberg
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Jennifer W Mitchell
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Penny W Burgoon
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Christian Beaulé
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Eberhard Weihe
- Institute of Anatomy and Cell Biology and Center of Mind, Brain and Behaviour, University of Marburg, Marburg, Germany
| | - Martin K-H Schäfer
- Institute of Anatomy and Cell Biology and Center of Mind, Brain and Behaviour, University of Marburg, Marburg, Germany
| | - Lee E Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, Bethesda, MD, United States
| | - Sunny Z Jiang
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, Bethesda, MD, United States
| | - Martha U Gillette
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
12
|
Slow shift of dead zone after an abrupt shift of the light-dark cycle. Brain Res 2019; 1714:73-80. [DOI: 10.1016/j.brainres.2019.02.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/24/2019] [Accepted: 02/12/2019] [Indexed: 12/27/2022]
|
13
|
Xie Y, Tang Q, Chen G, Xie M, Yu S, Zhao J, Chen L. New Insights Into the Circadian Rhythm and Its Related Diseases. Front Physiol 2019; 10:682. [PMID: 31293431 PMCID: PMC6603140 DOI: 10.3389/fphys.2019.00682] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/13/2019] [Indexed: 12/18/2022] Open
Abstract
Circadian rhythms (CR) are a series of endogenous autonomous oscillators generated by the molecular circadian clock which acting on coordinating internal time with the external environment in a 24-h daily cycle. The circadian clock system is a major regulatory factor for nearly all physiological activities and its disorder has severe consequences on human health. CR disruption is a common issue in modern society, and researches about people with jet lag or shift works have revealed that CR disruption can cause cognitive impairment, psychiatric illness, metabolic syndrome, dysplasia, and cancer. In this review, we summarized the synchronizers and the synchronization methods used in experimental research, and introduced CR monitoring and detection methods. Moreover, we evaluated conventional CR databases, and analyzed experiments that characterized the underlying causes of CR disorder. Finally, we further discussed the latest developments in understanding of CR disruption, and how it may be relevant to health and disease. Briefly, this review aimed to synthesize previous studies to aid in future studies of CR and CR-related diseases.
Collapse
Affiliation(s)
- Yanling Xie
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingming Tang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangjin Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengru Xie
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoling Yu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiajia Zhao
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Bastos MAV, Oliveira Bastos PRHD, Portella RB, Soares LFG, Conde RB, Rodrigues PMF, Lucchetti G. Pineal gland and schizophrenia: A systematic review and meta-analysis. Psychoneuroendocrinology 2019; 104:100-114. [PMID: 30831343 DOI: 10.1016/j.psyneuen.2019.02.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 01/17/2019] [Accepted: 02/24/2019] [Indexed: 12/29/2022]
Abstract
Melatonin (MLT), the main hormone of the pineal gland (PG), is assumed to support initiation and maintenance of sleep, and a stable sleep-wake cycle, exerting antioxidative and neuroprotective actions. Evidence demonstrates that sleep and circadian rhythm abnormalities are very common in schizophrenia patients. Some imaging studies suggest structural abnormalities of the PG in these patients as well. We aimed to critically appraise the literature on PG imaging and melatonin secretion in schizophrenia patients, in comparison to matched healthy controls, and to review placebo-controlled trials of add-on exogenous MLT treatment in schizophrenia patients. In this systematic review, twenty-nine studies were included. Meta-analytical evaluation of data was possible only for MLT secretion finding that midnight plasma levels were significantly reduced in individuals with schizophrenia as compared to healthy controls (Hedge`s g = 1.32, p < 0.01). Imaging studies demonstrated greater prevalence of enlarged calcifications (>1 cm) of the PG (2 out of 2 computed tomography studies) and smaller PG volume (2 out of 3 magnetic resonance studies) compared with healthy controls. Anatomic and functional abnormalities of the PG were not associated with duration of illness or with treatment factors, maybe suggesting them to be primary characteristics of the disease and genetically based. Add-on MLT treatment leads to a modest improvement of objective and subjective sleep quality, of metabolic adverse effects of antipsychotics, and of tardive dyskinesia symptoms in schizophrenia patients. It remains to be established whether MLT treatment in prodromal phases of the disease could prevent neurostructural abnormalities.
Collapse
Affiliation(s)
- Marco Aurélio Vinhosa Bastos
- Federal University of Mato Grosso do Sul, Postgraduate Program in Health and Development, Av. Senador Filinto Muller, s/n - Cidade Universitária, Campo Grande, MS, 79070-900, Brazil.
| | - Paulo Roberto Haidamus de Oliveira Bastos
- Federal University of Mato Grosso do Sul, Postgraduate Program in Health and Development, Av. Senador Filinto Muller, s/n - Cidade Universitária, Campo Grande, MS, 79070-900, Brazil
| | - Renata Boschi Portella
- Federal University of Mato Grosso do Sul, Faculty of Medicine, Av. Senador Filinto Muller, s/n - Cidade Universitária, Campo Grande, MS, 79070-900, Brazil
| | - Leonardo Fabrício Gomes Soares
- Federal University of Mato Grosso do Sul, Postgraduate Program in Health and Development, Av. Senador Filinto Muller, s/n - Cidade Universitária, Campo Grande, MS, 79070-900, Brazil
| | - Ricardo Brilhante Conde
- Proexames Imaging Clinic, Av. Mato Grosso, 1772 - Centro, Campo Grande, MS, 79020-201, Brazil
| | | | - Giancarlo Lucchetti
- Federal University of Juiz de Fora, School of Medicine, Av. Eugênio do Nascimento, s/n - Dom Bosco, Juiz de Fora, MG, 36036-330, Brazil
| |
Collapse
|
15
|
Ago Y, Hayata A, Hashimoto H. [Pathophysiological implication of the VPAC2 receptor in psychiatric disorders]. Nihon Yakurigaku Zasshi 2019; 151:249-253. [PMID: 29887574 DOI: 10.1254/fpj.151.249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
The advent of the genomic era has led to the discovery of linkages of several genes and pathways to schizophrenia and autism spectrum disorder (ASD) that may serve as new biomarkers or therapeutic targets for these diseases. Two large-scale genetic studies published early in 2011 provided evidence that functional microduplications at 7q36.3, containing VIPR2, are a risk factor for schizophrenia. 7q36.3 microduplications were also reported to be significantly increased in ASD. VIPR2 encodes VPAC2, a seven transmembrane heterotrimeric G protein-coupled receptor that binds two homologous neuropeptides with high affinity, PACAP and VIP. These clinical studies demonstrate a VIPR2 genetic linkage to schizophrenia and ASD and should lead to novel insights into the etiology of these mental health disorders. However, the mechanism by which overactive VPAC2 signaling may lead to schizophrenia and ASD is unknown. In the present review, we will describe recent advances in the genetics of schizophrenia and attempt to discuss the pathophysiological role of altered VPAC2 signaling in psychiatric disorders.
Collapse
Affiliation(s)
- Yukio Ago
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Atsuko Hayata
- Center for Child Mental Development, United Graduate School of Child Development, Osaka University
| | - Hitoshi Hashimoto
- Center for Child Mental Development, United Graduate School of Child Development, Osaka University.,Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University.,Division of Bioscience, Institute for Datability Science, Osaka University
| |
Collapse
|
16
|
Yilmaz A, Kalsbeek A, Buijs RM. Functional changes of the SCN in spontaneous hypertension but not after the induction of hypertension. Chronobiol Int 2018; 35:1221-1235. [DOI: 10.1080/07420528.2018.1469035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ajda Yilmaz
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), Amsterdam The Netherlands
| | - Ruud M Buijs
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Cell Biology and Physiology, Institute for Biomedical Research, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| |
Collapse
|
17
|
Firouzabadi SG, Kariminejad R, Vameghi R, Darvish H, Ghaedi H, Banihashemi S, Firouzkouhi Moghaddam M, Jamali P, Mofidi Tehrani HF, Dehghani H, Narooie-Nejad M, Jamshidi J, Tafakhori A, Sadabadi S, Najmabadi H, Behjati F. Copy Number Variants in Patients with Autism and Additional Clinical Features: Report of VIPR2 Duplication and a Novel Microduplication Syndrome. Mol Neurobiol 2016; 54:7019-7027. [PMID: 27796743 DOI: 10.1007/s12035-016-0202-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 10/11/2016] [Indexed: 10/20/2022]
Abstract
Autism is a common neurodevelopmental disorder estimated to affect 1 in 68 children. Many studies have shown the role of copy number variants (CNVs) as a major contributor in the etiology of autism with the overall detection rate of about 10-15 % and over 20 % when syndromic forms of autism exist. In this study, we used array CGH to identify CNVs in 15 Iranian patients with autism. To elevate our diagnostic yield, we selected the sporadic patients who had additional clinical features including intellectual disability (ID), craniofacial anomaly, and seizure. Six out of 15 patients showed clinically relevant CNVs including pathogenic and likely pathogenic copy number gains or losses. We report a novel gene duplication syndrome (10q21.2q21.3 microduplication) and present a new evidence for VIPR2 duplication, as a candidate gene for autism. Furthermore, we describe the first manifesting carrier female with deletion of SLC6A8 and BCAP31 genes on Xq28. Our findings suggest that there might be a higher prevalence of clinically significant CNVs in patients with autism and additional clinical manifestations. The CNV analysis in such patients could lead to the discovery of novel syndromes as well as unraveling the etiology of autism.
Collapse
Affiliation(s)
| | | | - Roshanak Vameghi
- Pediatric Neurorehabilitation Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Hossein Darvish
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ghaedi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Susan Banihashemi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Mahboubeh Firouzkouhi Moghaddam
- Child and Adolescent Psychiatry Department, Zahedan University of Medical Sciences, Zahedan, Iran.,Research Center for Children and Adolescents Health, Zahedan University of Medical Sciences, Zahedan, Iran
| | | | | | - Hossein Dehghani
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Mehrnaz Narooie-Nejad
- Genetics of Non-communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Javad Jamshidi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Abbas Tafakhori
- Department of Neurology, School of Medicine, Imam Khomeini Hospital and Iranian Center of Neurological Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeid Sadabadi
- Bahar Education and Rehabilitation Center for the handicapped, Tehran, Iran
| | - Hossein Najmabadi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.,Kariminejad-Najmabadi Pathology and Genetics Center, Tehran, Iran
| | - Farkhondeh Behjati
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Evans JA. Collective timekeeping among cells of the master circadian clock. J Endocrinol 2016; 230:R27-49. [PMID: 27154335 PMCID: PMC4938744 DOI: 10.1530/joe-16-0054] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/06/2016] [Indexed: 01/09/2023]
Abstract
The suprachiasmatic nucleus (SCN) of the anterior hypothalamus is the master circadian clock that coordinates daily rhythms in behavior and physiology in mammals. Like other hypothalamic nuclei, the SCN displays an impressive array of distinct cell types characterized by differences in neurotransmitter and neuropeptide expression. Individual SCN neurons and glia are able to display self-sustained circadian rhythms in cellular function that are regulated at the molecular level by a 24h transcriptional-translational feedback loop. Remarkably, SCN cells are able to harmonize with one another to sustain coherent rhythms at the tissue level. Mechanisms of cellular communication in the SCN network are not completely understood, but recent progress has provided insight into the functional roles of several SCN signaling factors. This review discusses SCN organization, how intercellular communication is critical for maintaining network function, and the signaling mechanisms that play a role in this process. Despite recent progress, our understanding of SCN circuitry and coupling is far from complete. Further work is needed to map SCN circuitry fully and define the signaling mechanisms that allow for collective timekeeping in the SCN network.
Collapse
Affiliation(s)
- Jennifer A Evans
- Department of Biomedical SciencesMarquette University, Milwaukee, WI, USA
| |
Collapse
|
19
|
Kingsbury NJ, Taylor SR, Henson MA. Inhibitory and excitatory networks balance cell coupling in the suprachiasmatic nucleus: A modeling approach. J Theor Biol 2016; 397:135-44. [PMID: 26972478 DOI: 10.1016/j.jtbi.2016.02.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 01/07/2016] [Accepted: 02/01/2016] [Indexed: 01/13/2023]
Abstract
Neuronal coupling contributes to circadian rhythms formation in the suprachiasmatic nucleus (SCN). While the neurotransmitter vasoactive intestinal polypeptide (VIP) is considered essential for synchronizing the oscillations of individual neurons, γ-aminobutyric acid (GABA) does not have a clear functional role despite being highly concentrated in the SCN. While most studies have examined the role of either GABA or VIP, our mathematical modeling approach explored their interplay on networks of SCN neurons. Tuning the parameters that control the release of GABA and VIP enabled us to optimize network synchrony, which was achieved at a peak firing rate during the subjective day of about 7Hz. Furthermore, VIP and GABA modulation could adjust network rhythm amplitude and period without sacrificing synchrony. We also performed simulations of SCN networks to phase shifts during 12h:12h light-dark cycles and showed that GABA networks reduced the average time for the SCN model to re-synchronize. We hypothesized that VIP and GABA balance cell coupling in the SCN to promote synchronization of heterogeneous oscillators while allowing flexibility for adjustment to environmental changes.
Collapse
Affiliation(s)
- Nathaniel J Kingsbury
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA 01007, United States
| | - Stephanie R Taylor
- Department of Computer Science, Colby College, Waterville, ME 04901, United States
| | - Michael A Henson
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA 01007, United States.
| |
Collapse
|
20
|
Evans JA, Gorman MR. In synch but not in step: Circadian clock circuits regulating plasticity in daily rhythms. Neuroscience 2016; 320:259-80. [PMID: 26861419 DOI: 10.1016/j.neuroscience.2016.01.072] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/26/2016] [Accepted: 01/27/2016] [Indexed: 11/16/2022]
Abstract
The suprachiasmatic nucleus (SCN) is a network of neural oscillators that program daily rhythms in mammalian behavior and physiology. Over the last decade much has been learned about how SCN clock neurons coordinate together in time and space to form a cohesive population. Despite this insight, much remains unknown about how SCN neurons communicate with one another to produce emergent properties of the network. Here we review the current understanding of communication among SCN clock cells and highlight a collection of formal assays where changes in SCN interactions provide for plasticity in the waveform of circadian rhythms in behavior. Future studies that pair analytical behavioral assays with modern neuroscience techniques have the potential to provide deeper insight into SCN circuit mechanisms.
Collapse
Affiliation(s)
- J A Evans
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA.
| | - M R Gorman
- Department of Psychology, University of San Diego, La Jolla, CA, USA
| |
Collapse
|
21
|
Ganea D, Hooper KM, Kong W. The neuropeptide vasoactive intestinal peptide: direct effects on immune cells and involvement in inflammatory and autoimmune diseases. Acta Physiol (Oxf) 2015; 213:442-52. [PMID: 25422088 DOI: 10.1111/apha.12427] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 10/13/2014] [Accepted: 11/21/2014] [Indexed: 12/13/2022]
Abstract
Neuropeptides represent an important category of endogenous contributors to the establishment and maintenance of immune deviation in the immune-privileged organs such as the CNS and in the control of acute inflammation in the peripheral immune organs. Vasoactive intestinal peptide (VIP) is a major immunoregulatory neuropeptide widely distributed in the central and peripheral nervous system. In addition to neurones, VIP is synthesized by immune cells which also express VIP receptors. Here, we review the current information on VIP production and VIP-receptor-mediated effects in the immune system, the role of endogenous and exogenous VIP in inflammatory and autoimmune disorders and the present and future VIP therapeutic approaches.
Collapse
Affiliation(s)
- D. Ganea
- Department of Microbiology and Immunology; Temple University School of Medicine; Philadelphia PA USA
| | - K. M. Hooper
- Department of Microbiology and Immunology; Temple University School of Medicine; Philadelphia PA USA
| | - W. Kong
- Department of Microbiology and Immunology; Temple University School of Medicine; Philadelphia PA USA
| |
Collapse
|
22
|
Brown R, Hynes-Allen A, Swan AJ, Dissanayake KN, Gillingwater TH, Ribchester RR. Activity-dependent degeneration of axotomized neuromuscular synapses in Wld S mice. Neuroscience 2015; 290:300-20. [PMID: 25617654 PMCID: PMC4362769 DOI: 10.1016/j.neuroscience.2015.01.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/10/2015] [Accepted: 01/12/2015] [Indexed: 12/12/2022]
Abstract
Use and disuse may influence synaptic maintenance but so far evidence for this has been indirect. We tested whether stimulation or disuse of neuromuscular junctions in adult WldS mice altered vulnerability to axotomy. Moderate activity optimized resistance to axotomy while disuse or stimulation increased the rate of synaptic degeneration.
Activity and disuse of synapses are thought to influence progression of several neurodegenerative diseases in which synaptic degeneration is an early sign. Here we tested whether stimulation or disuse renders neuromuscular synapses more or less vulnerable to degeneration, using axotomy as a robust trigger. We took advantage of the slow synaptic degeneration phenotype of axotomized neuromuscular junctions in flexor digitorum brevis (FDB) and deep lumbrical (DL) muscles of Wallerian degeneration-Slow (WldS) mutant mice. First, we maintained ex vivo FDB and DL nerve-muscle explants at 32 °C for up to 48 h. About 90% of fibers from WldS mice remained innervated, compared with about 36% in wild-type muscles at the 24-h checkpoint. Periodic high-frequency nerve stimulation (100 Hz: 1 s/100 s) reduced synaptic protection in WldS preparations by about 50%. This effect was abolished in reduced Ca2+ solutions. Next, we assayed FDB and DL innervation after 7 days of complete tetrodotoxin (TTX)-block of sciatic nerve conduction in vivo, followed by tibial nerve axotomy. Five days later, only about 9% of motor endplates remained innervated in the paralyzed muscles, compared with about 50% in 5 day-axotomized muscles from saline-control-treated WldS mice with no conditioning nerve block. Finally, we gave mice access to running wheels for up to 4 weeks prior to axotomy. Surprisingly, exercising WldS mice ad libitum for 4 weeks increased about twofold the amount of subsequent axotomy-induced synaptic degeneration. Together, the data suggest that vulnerability of mature neuromuscular synapses to axotomy, a potent neurodegenerative trigger, may be enhanced bimodally, either by disuse or by hyperactivity.
Collapse
Affiliation(s)
- R Brown
- Euan MacDonald Centre for Motor Neurone Disease Research, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - A Hynes-Allen
- Euan MacDonald Centre for Motor Neurone Disease Research, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - A J Swan
- Euan MacDonald Centre for Motor Neurone Disease Research, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - K N Dissanayake
- Euan MacDonald Centre for Motor Neurone Disease Research, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - T H Gillingwater
- Euan MacDonald Centre for Motor Neurone Disease Research, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - R R Ribchester
- Euan MacDonald Centre for Motor Neurone Disease Research, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK.
| |
Collapse
|
23
|
Kunst M, Tso MCF, Ghosh DD, Herzog ED, Nitabach MN. Rhythmic control of activity and sleep by class B1 GPCRs. Crit Rev Biochem Mol Biol 2014; 50:18-30. [PMID: 25410535 DOI: 10.3109/10409238.2014.985815] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Members of the class B1 family of G-protein coupled receptors (GPCRs) whose ligands are neuropeptides have been implicated in regulation of circadian rhythms and sleep in diverse metazoan clades. This review discusses the cellular and molecular mechanisms by which class B1 GPCRs, especially the mammalian VPAC2 receptor and its functional homologue PDFR in Drosophila and C. elegans, regulate arousal and daily rhythms of sleep and wake. There are remarkable parallels in the cellular and molecular roles played by class B1 intercellular signaling pathways in coordinating arousal and circadian timekeeping across multiple cells and tissues in these very different genetic model organisms.
Collapse
Affiliation(s)
- Michael Kunst
- Department of Cellular and Molecular Physiology, Yale University School of Medicine , New Haven, CT , USA and
| | | | | | | | | |
Collapse
|
24
|
Gu C, Ramkisoensing A, Liu Z, Meijer JH, Rohling JHT. The proportion of light-responsive neurons determines the limit cycle properties of the suprachiasmatic nucleus. J Biol Rhythms 2014; 29:16-27. [PMID: 24492879 DOI: 10.1177/0748730413516752] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In mammals, the central clock in the suprachiasmatic nucleus (SCN) controls physiological and behavioral circadian rhythms and is entrained to the external light-dark cycle. The ability of the SCN to entrain can be measured by exposing the animal to a light-dark cycle with a duration that deviates from 24 h (T-cycles); a wider entrainment range reflects a higher ability to entrain. The neurons of the SCN are either light responsive or light unresponsive and are mutually synchronized. The coupling and synchronization between individual SCN neurons and between groups of neurons within the SCN influence the SCN's ability to entrain. Some studies suggest that enhanced coupling decreases the entrainment range, whereas others suggest that enhanced coupling increases the entrainment range. The latter results are surprising, as they are not consistent with the prevalent assumption that the SCN is a limit cycle oscillator that has larger phase shifts when the amplitude is smaller. Here, we used the Poincaré and Goodwin models to test entrainment properties using various proportions of neurons that are responsive to an external stimulus. If all neurons receive external input, the SCN shows limit cycle behavior in all conditions. If all neurons do not receive light input, we found that the entrainment range of the SCN was positively related to coupling strength when coupling was weak. When coupling strength was stronger and above a critical value, the entrainment range was negatively correlated with coupling strength. The results obtained from our simulations were confirmed by analytical studies. Thus, the limit cycle behavior of the SCN appears to be critically dependent on the coupling strength among the neurons and the proportion of neurons that respond to the entraining stimulus.
Collapse
Affiliation(s)
- Changgui Gu
- Laboratory for Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | |
Collapse
|
25
|
Ananthasubramaniam B, Herzog ED, Herzel H. Timing of neuropeptide coupling determines synchrony and entrainment in the mammalian circadian clock. PLoS Comput Biol 2014; 10:e1003565. [PMID: 24743470 PMCID: PMC3990482 DOI: 10.1371/journal.pcbi.1003565] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 02/25/2014] [Indexed: 11/18/2022] Open
Abstract
Robust synchronization is a critical feature of several systems including the mammalian circadian clock. The master circadian clock in mammals consists of about 20000 ‘sloppy’ neuronal oscillators within the hypothalamus that keep robust time by synchronization driven by inter-neuronal coupling. The complete understanding of this synchronization in the mammalian circadian clock and the mechanisms underlying it remain an open question. Experiments and computational studies have shown that coupling individual oscillators can achieve robust synchrony, despite heterogeneity and different network topologies. But, much less is known regarding the mechanisms and circuits involved in achieving this coupling, due to both system complexity and experimental limitations. Here, we computationally study the coupling mediated by the primary coupling neuropeptide, vasoactive intestinal peptide (VIP) and its canonical receptor, VPAC2R, using the transcriptional elements and generic mode of VIP-VPAC2R signaling. We find that synchrony is only possible if VIP (an inducer of Per expression) is released in-phase with activators of Per expression. Moreover, anti-phasic VIP release suppresses coherent rhythms by moving the network into a desynchronous state. Importantly, experimentally observed rhythms in VPAC2R have little effect on network synchronization, but can improve the amplitude of the SCN network rhythms while narrowing the network entrainment range. We further show that these findings are valid across several computational network models. Thus, we identified a general design principle to achieve robust synchronization: An activating coupling agent, such as VIP, must act in-phase with the activity of core-clock promoters. More generally, the phase of coupling is as critical as the strength of coupling from the viewpoint of synchrony and entrainment. Synchronization among multiple oscillators is a common theme in many biological and engineered systems. Here, we look at its use by the mammalian biological clock to keep accurate time. Through biochemical interactions among a network of inaccurate neuron clocks, a strong precise clock is produced. Although we are gradually learning more about these biochemical interactions, the details still remain largely unclear. Studies, both computational and experimental, have shown that the strength of the rhythmic interaction critically decides if a system can synchronize, i.e., the interactions must be strong enough. In this work, we show that the rhythmic interaction between these neuronal clocks must be timed correctly (in the right phase) in addition to being strong enough to synchronize the network. Activating (repressing) interactions must coincide with other activators (repressors) in each neuron to achieve synchrony. Since this principle imposes certain properties on synchronizing interactions, these properties can be used to identify and understand novel interaction mechanisms. Further, these principles are applicable to interactions between cellular oscillators in other tissues and organisms.
Collapse
Affiliation(s)
| | - Erik D. Herzog
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Hanspeter Herzel
- Institute for Theoretical Biology, Charité and Humboldt University Berlin, Berlin, Germany
| |
Collapse
|
26
|
Soetedjo L, Jin H. Agonist-induced GPCR shedding from the ciliary surface is dependent on ESCRT-III and VPS4. Curr Biol 2014; 24:509-18. [PMID: 24530064 DOI: 10.1016/j.cub.2014.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/18/2013] [Accepted: 01/03/2014] [Indexed: 01/22/2023]
Abstract
BACKGROUND Membrane trafficking of G protein-coupled receptors (GPCRs) is crucial for temporal and spatial control of cell-surface GPCR signaling. Receptor internalization is a well-documented method cells use for regulating a wide variety of GPCRs following their exposure to agonists. RESULTS We report that, upon agonist stimulation, a GPCR called vasoactive intestinal peptide receptor 2 (VPAC2) is shed, rather than being internalized, in vitro and in vivo, from the membrane of primary cilia--solitary hair-like organelles that project from the cell surface. VPAC2 is released into the extracellular milieu in the form of ciliary ectosomes that are devoid of exosome markers. The agonist-induced VPAC2 shedding is selective, as shown by the fact that other ciliary membrane proteins including two ciliary GPCRs are not shed with VPAC2. VPAC2 ectosome shedding is dependent on several components of endosomal sorting complexes required for transport (ESCRT), including a subset of ESCRT-III, VPS4, and LIP5. Agonist-stimulated VPAC2 is important for ciliary-ectosome generation because it allows VPS4 and LIP5 to transiently accumulate in primary cilia. Shedding of VPAC2 from the ciliary surface results in termination of intracellular VPAC2 signaling. CONCLUSIONS Agonist-induced GPCR shedding from the ciliary surface may represent an additional mode of GPCR trafficking and signal regulation.
Collapse
Affiliation(s)
- Livana Soetedjo
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Hua Jin
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
27
|
Shen S, Gehlert DR, Collier DA. PACAP and PAC1 receptor in brain development and behavior. Neuropeptides 2013; 47:421-30. [PMID: 24220567 DOI: 10.1016/j.npep.2013.10.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 10/12/2013] [Accepted: 10/13/2013] [Indexed: 10/26/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) act through three class B G-protein coupled receptors, PAC1, VPAC1 and VPAC2, initiating multiple signaling pathways. In addition to natural peptides ligands, a number of synthetic peptides and a small molecular antagonist have been generated. Genetically modified animals have been produced for the neuropeptides and receptors. Neuroanatomical, electrophysiological, behavioral and pharmacological characterization of the mutants and transgenic mice uncovered diverse roles of PACAP-PAC1-VAPC2 signaling in peripheral tissues and in the central nervous system. Human genetic studies suggest that the PACAP-PAC1-VPAC2 signaling can be associated with psychiatric illness via mechanisms of not only loss-of-function, but also gain-of-function. For example, a duplication of chromosome 7q36.3 (encoding the VPAC2 receptor) was shown to be associated with schizophrenia, and high levels of PACAP-PAC1 signaling are associated with posttraumatic stress disorder. Whereas knockout animals are appropriate to address loss-of-function of human genetics, transgenic mice overexpressing human transgenes in native environment using artificial chromosomes are particularly valuable and essential to address the consequences of gain-of-function. This review focuses on role of PACAP and PAC1 receptor in brain development, behavior of animals and potential implication in human neurodevelopmental disorders. It also encourages keeping an open mind that alterations of VIP/PACAP signaling may associate with psychiatric illness without overt neuroanatomic changes, and that tuning of VIP/PACAP signaling may represent a novel avenue for the treatment of the psychiatric illness.
Collapse
Affiliation(s)
- Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, National University of Ireland (NUI) Galway, Galway, Ireland.
| | | | | |
Collapse
|
28
|
Quinn JP, Warburton A, Myers P, Savage AL, Bubb VJ. Polymorphic variation as a driver of differential neuropeptide gene expression. Neuropeptides 2013; 47:395-400. [PMID: 24210140 DOI: 10.1016/j.npep.2013.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 10/09/2013] [Accepted: 10/10/2013] [Indexed: 11/15/2022]
Abstract
The regulation of neuropeptide gene expression and their receptors in a tissue specific and stimulus inducible manner will determine in part behaviour and physiology. This can be a dynamic process resulting from short term changes in response to the environment or long term modulation imposed by epigenetically determined mechanisms established during life experiences. The latter underpins what is termed 'nature and nurture, or 'gene×environment interactions'. Dynamic gene expression of neuropeptides or their receptors is a key component of signalling in the CNS and their inappropriate regulation is therefore a predicted target underpinning psychiatric disorders and neuropathological processes. Finding the regulatory domains within our genome which have the potential to direct gene expression is a difficult challenge as 98% of our genome is non-coding and, with the exception of proximal promoter regions, such elements can be quite distant from the gene that they regulate. This review will deal with how we can find such domains by addressing both the most conserved non-exonic regions in the genome using comparative genomics and the most recent or constantly evolving DNA such as repetitive DNA or retrotransposons. We shall also explore how polymorphic changes in such domains can be associated with CNS disorders by altering the appropriate gene expression patterns which maintain normal physiology.
Collapse
Affiliation(s)
- John P Quinn
- Neurogenetics in Wellbeing and Disease Section, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, UK.
| | | | | | | | | |
Collapse
|
29
|
Cao R, Robinson B, Xu H, Gkogkas C, Khoutorsky A, Alain T, Yanagiya A, Nevarko T, Liu AC, Amir S, Sonenberg N. Translational control of entrainment and synchrony of the suprachiasmatic circadian clock by mTOR/4E-BP1 signaling. Neuron 2013; 79:712-24. [PMID: 23972597 DOI: 10.1016/j.neuron.2013.06.026] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2013] [Indexed: 11/28/2022]
Abstract
Protein synthesis is critical for circadian clock function, but little is known of how translational regulation controls the master pacemaker in mammals, the suprachiasmatic nucleus (SCN). Here we demonstrate that the pivotal translational repressor, the eukaryotic translational initiation factor 4E binding protein 1 (4E-BP1), is rhythmically regulated via the mechanistic target of rapamycin (mTOR) signaling in the SCN and preferentially represses vasoactive intestinal peptide (Vip) mRNA translation. Knockout (KO) of Eif4ebp1 (gene encoding 4E-BP1) leads to upregulation of VIP and higher amplitude of molecular rhythms in the SCN. Consequently, the 4E-BP1 null mice exhibit accelerated re-entrainment to a shifted light/dark cycle and are more resistant to the rhythm-disruptive effects of constant light. Conversely, in Mtor(+/-) mice VIP expression is decreased and susceptibility to the effects of constant light is increased. These results reveal a key role for mTOR/4E-BP1-mediated translational control in regulating entrainment and synchrony of the master clock.
Collapse
Affiliation(s)
- Ruifeng Cao
- Department of Biochemistry and Goodman Cancer Research Center, McGill University, Montreal, QC H3A 1A3, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Yiu WC, Yap MKH, Fung WY, Ng PW, Yip SP. Genetic susceptibility to refractive error: association of vasoactive intestinal peptide receptor 2 (VIPR2) with high myopia in Chinese. PLoS One 2013; 8:e61805. [PMID: 23637909 PMCID: PMC3630195 DOI: 10.1371/journal.pone.0061805] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 03/14/2013] [Indexed: 11/26/2022] Open
Abstract
Myopia is the most common ocular disease worldwide. We investigated the association of high myopia with the common single nucleotide polymorphisms (SNPs) of five candidate genes – early growth response 1 (EGR1), v-fos FBJ murine osteosarcoma viral oncogene homolog (FOS), jun oncogene (JUN), vasoactive intestinal peptide (VIP), and vasoactive intestinal peptide receptor 2 (VIPR2). We recruited 1200 unrelated Chinese subjects with 600 cases (spherical equivalent ≤−8.00 diopters) and 600 controls (spherical equivalent within ±1.00 diopter). A discovery sample set was formed from 300 cases and 300 controls, and a replication sample set from the remaining samples. Tag SNPs were genotyped for the discovery sample set, and the most significant haplotypes and their constituent SNPs were followed up with the replication sample set. The allele and haplotype frequencies in cases and controls were compared by logistic regression adjusted for sex and age to give Pa values, and multiple comparisons were corrected by permutation test to give Paemp values. Odd ratios (OR) were calculated accordingly. In the discovery phase, EGR1, JUN and VIP did not show any significant association while FOS and VIPR2 demonstrated significant haplotype association with high myopia. In the replication phase, the haplotype association for VIPR2 was successfully replicated, but not FOS. In analysis combining both sample sets, the most significant association signals of VIPR2 were the single marker rs2071625 (Pa = 0.0008, Paemp = 0.0046 and OR = 0.75) and the 4-SNP haplotype window rs2071623-rs2071625-rs2730220-rs885863 (omnibus test, Pa = 9.10e-10 and Paemp = 0.0001) with one protective haplotype (GGGG: Paemp = 0.0002 and OR = 0.52) and one high-risk haplotype (GAGA: Paemp = 0.0027 and OR = 4.68). This 4-SNP haplotype window was the most significant in all sample sets examined. This is the first study to suggest a role of VIPR2 in the genetic susceptibility to high myopia. EGR1, JUN, FOS and VIP are unlikely to be important in predisposing humans to high myopia.
Collapse
Affiliation(s)
- Wai Chi Yiu
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | | | | | | | | |
Collapse
|
31
|
An S, Tsai C, Ronecker J, Bayly A, Herzog ED. Spatiotemporal distribution of vasoactive intestinal polypeptide receptor 2 in mouse suprachiasmatic nucleus. J Comp Neurol 2013; 520:2730-41. [PMID: 22684939 DOI: 10.1002/cne.23078] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Vasoactive intestinal polypeptide (VIP) signaling is critical for circadian rhythms. For example, the expression of VIP and its main receptor, VPAC2R, is necessary for maintaining synchronous daily rhythms among neurons in the suprachiasmatic nucleus (SCN), a master circadian pacemaker in animals. Where and when VPAC2R protein is expressed in the SCN and other brain areas has not been examined. Using immunohistochemistry, we characterized a new antibody and found that VPAC2R was highly enriched in the SCN and detectable at low levels in many brain areas. Within the SCN, VPAC2R was circadian, peaking in the subjective morning, and abundantly expressed from the rostral to caudal margins with more in the dorsomedial than ventrolateral area. VPAC2R was found in nearly all SCN cells including neurons expressing either VIP or vasopressin (AVP). SCN neurons mainly expressed VPAC2R in their somata and dendrites, not axons. Finally, constant light increased VIP and AVP expression, but not VPAC2R. We conclude that the circadian clock, not the ambient light level, regulates VPAC2R protein localization. These results are consistent with VPAC2R playing a role in VIP signaling at all times of day, broadly throughout the brain and in all SCN cells.
Collapse
Affiliation(s)
- Sungwon An
- Department of Biology, Washington University, Saint Louis, Missouri 63130, USA
| | | | | | | | | |
Collapse
|
32
|
Abstract
In mammals, the suprachiasmatic nucleus (SCN) of the hypothalamus is considered as the master circadian pacemaker. Each cell in the SCN contains an autonomous molecular clock, and the SCN is composed of multiple single-cell circadian oscillators. The fundamental question is how the individual cellular oscillators, expressing a wide range of periods, interact and assemble to achieve phase synchronization. In natural conditions, the interaction between the SCN neurons is non-negligible and coupling between cells in the SCN is achieved partly by neurotransmitters. Though there have been a lot of works about the synchronization of circadian oscillators coupled by neurotransmitter, most of the works are based on numerical simulation with little theoretical analysis. In this paper, from the theoretical analysis, we prove that the clocks can be synchronized by the neurotransmitter with mathematical knowledge. Our results also show that the coupling among neurons can synchronize circadian oscillators but the synchronized oscillators are not with period of 24 h, which indicates that environmental cues are necessary to entrain oscillators with period of 24 h. This work can offer theoretical basis to study circadian synchronization in a numerical or experimental way.
Collapse
Affiliation(s)
- YING LI
- College of Information Technology, Shanghai Ocean University, 999 Hucheng Huan Road, Shanghai 201306, China
| | - ZENGRONG LIU
- Institute of Systems Biology, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| |
Collapse
|
33
|
|
34
|
Vasalou C, Herzog E, Henson M. Multicellular model for intercellular synchronization in circadian neural networks. Biophys J 2011; 101:12-20. [PMID: 21723810 PMCID: PMC3127187 DOI: 10.1016/j.bpj.2011.04.051] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 03/22/2011] [Accepted: 04/18/2011] [Indexed: 12/22/2022] Open
Abstract
We developed a multicellular model characterized by a high degree of heterogeneity to investigate possible mechanisms that underlie circadian network synchronization and rhythmicity in the suprachiasmatic nucleus (SCN). We populated a two-dimensional grid with 400 model neurons coupled via γ-aminobutyric acid (GABA) and vasoactive intestinal polypeptide (VIP) neurotransmitters through a putative Ca(2+) mediated signaling cascade to investigate their roles in gene expression and electrical firing activity of cell populations. As observed experimentally, our model predicted that GABA would affect the amplitude of circadian oscillations but not synchrony among individual oscillators. Our model recapitulated experimental findings of decreased synchrony and average periods, loss of rhythmicity, and reduced circadian amplitudes as VIP signaling was eliminated. In addition, simulated increases of VIP reduced periodicity and synchrony. We therefore postulated a physiological range of VIP within which the system is able to produce sustained and synchronized oscillations. Our model recapitulated experimental findings of diminished amplitudes and periodicity with decreasing intracellular Ca(2+) concentrations, suggesting that such behavior could be due to simultaneous decrease of individual oscillation amplitudes and population synchrony. Simulated increases in Cl(-) levels resulted in increased Cl(-) influx into the cytosol, a decrease of inhibitory postsynaptic currents, and ultimately a shift of GABA-elicited responses from inhibitory to excitatory. The simultaneous reduction of IPSCs and increase in membrane resting potential produced GABA dose-dependent increases in firing rates across the population, as has been observed experimentally. By integrating circadian gene regulation and electrophysiology with intracellular and intercellular signaling, we were able to develop the first (to our knowledge) multicellular model that allows the effects of clock genes, electrical firing, Ca(2+), GABA, and VIP on circadian system behavior to be predicted.
Collapse
Affiliation(s)
- Christina Vasalou
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts
| | - Erik D. Herzog
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri
| | - Michael A. Henson
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts
| |
Collapse
|
35
|
Eban-Rothschild A, Belluci S, Bloch G. Maternity-related plasticity in circadian rhythms of bumble-bee queens. Proc Biol Sci 2011; 278:3510-6. [PMID: 21508036 DOI: 10.1098/rspb.2011.0579] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Unlike most animals studied so far in which the activity with no circadian rhythms is pathological or linked to deteriorating performance, worker bees and ants naturally care for their sibling brood around the clock with no apparent ill effects. Here, we tested whether bumble-bee queens that care alone for their first batch of offspring are also capable of a similar chronobiological plasticity. We monitored locomotor activity of Bombus terrestris queens at various life cycle stages, and queens for which we manipulated the presence of brood or removed the ovaries. We found that gynes typically emerged from the pupae with no circadian rhythms, but after several days showed robust rhythms that were not affected by mating or diapauses. Colony-founding queens with brood showed attenuated circadian rhythms, irrespective of the presence of ovaries. By contrast, queens that lost their brood switched again to activity with strong circadian rhythms. The discovery that circadian rhythms in bumble-bee queens are regulated by the life cycle and the presence of brood suggests that plasticity in the circadian clock of bees is ancient and related to maternal behaviour or physiology, and is not a derived trait that evolved with the evolution of the worker caste.
Collapse
Affiliation(s)
- Ada Eban-Rothschild
- Department of Ecology, Evolution and Behavior, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | | | | |
Collapse
|
36
|
Nishimoto M, Miyakawa H, Wada K, Furuta A. Activation of the VIP/VPAC2 system induces reactive astrocytosis associated with increased expression of glutamate transporters. Brain Res 2011; 1383:43-53. [DOI: 10.1016/j.brainres.2011.01.082] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 01/21/2011] [Accepted: 01/21/2011] [Indexed: 11/28/2022]
|
37
|
Hannibal J, Hsiung HM, Fahrenkrug J. Temporal phasing of locomotor activity, heart rate rhythmicity, and core body temperature is disrupted in VIP receptor 2-deficient mice. Am J Physiol Regul Integr Comp Physiol 2011; 300:R519-30. [DOI: 10.1152/ajpregu.00599.2010] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Neurons of the brain's biological clock located in the hypothalamic suprachiasmatic nucleus (SCN) generate circadian rhythms of physiology (core body temperature, hormone secretion, locomotor activity, sleep/wake, and heart rate) with distinct temporal phasing when entrained by the light/dark (LD) cycle. The neuropeptide vasoactive intestinal polypetide (VIP) and its receptor (VPAC2) are highly expressed in the SCN. Recent studies indicate that VIPergic signaling plays an essential role in the maintenance of ongoing circadian rhythmicity by synchronizing SCN cells and by maintaining rhythmicity within individual neurons. To further increase the understanding of the role of VPAC2 signaling in circadian regulation, we implanted telemetric devices and simultaneously measured core body temperature, spontaneous activity, and heart rate in a strain of VPAC2-deficient mice and compared these observations with observations made from mice examined by wheel-running activity. The study demonstrates that VPAC2 signaling is necessary for a functional circadian clock driving locomotor activity, core body temperature, and heart rate rhythmicity, since VPAC2-deficient mice lose the rhythms in all three parameters when placed under constant conditions (of either light or darkness). Furthermore, although 24-h rhythms for three parameters are retained in VPAC2-deficient mice during the LD cycle, the temperature rhythm displays markedly altered time course and profile, rising earlier and peaking ∼4–6 h prior to that of wild-type mice. The use of telemetric devices to measure circadian locomotor activity, temperature, and heart rate, together with the classical determination of circadian rhythms of wheel-running activity, raises questions about how representative wheel-running activity may be of other behavioral parameters, especially when animals have altered circadian phenotype.
Collapse
Affiliation(s)
- Jens Hannibal
- Department of Clinical Biochemistry, Bispebjerg Hospital, University of Copenhagen; and
| | - Hansen M. Hsiung
- Division of Endocrine Research, Eli Lilly and Co., Indianapolis, Indiana
| | - Jan Fahrenkrug
- Department of Clinical Biochemistry, Bispebjerg Hospital, University of Copenhagen; and
| |
Collapse
|
38
|
Komin N, Murza AC, Hernández-García E, Toral R. Synchronization and entrainment of coupled circadian oscillators. Interface Focus 2011; 1:167-76. [PMID: 22419982 PMCID: PMC3262239 DOI: 10.1098/rsfs.2010.0327] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 09/20/2010] [Indexed: 11/12/2022] Open
Abstract
Circadian rhythms in mammals are controlled by the neurons located in the suprachiasmatic nucleus of the hypothalamus. In physiological conditions, the system of neurons is very efficiently entrained by the 24 h light-dark cycle. Most of the studies carried out so far emphasize the crucial role of the periodicity imposed by the light-dark cycle in neuronal synchronization. Nevertheless, heterogeneity as a natural and permanent ingredient of these cellular interactions seemingly plays a major role in these biochemical processes. In this paper, we use a model that considers the neurons of the suprachiasmatic nucleus as chemically coupled modified Goodwin oscillators, and introduce non-negligible heterogeneity in the periods of all neurons in the form of quenched noise. The system response to the light-dark cycle periodicity is studied as a function of the interneuronal coupling strength, external forcing amplitude and neuronal heterogeneity. Our results indicate that the right amount of heterogeneity helps the extended system to respond globally in a more coherent way to the external forcing. Our proposed mechanism for neuronal synchronization under external periodic forcing is based on heterogeneity-induced oscillator death, damped oscillators being more entrainable by the external forcing than the self-oscillating neurons with different periods.
Collapse
Affiliation(s)
| | | | | | - R. Toral
- IFISC (Instituto de Física Interdisciplinar y Sistemas Complejos), CSIC-UIB, Campus UIB, 07122 Palma de Mallorca, Spain
| |
Collapse
|
39
|
Abstract
Mammalian circadian rhythms are controlled by endogenous biological oscillators, including a master clock located in the hypothalamic suprachiasmatic nuclei (SCN). Since the period of this oscillation is of approximately 24 h, to keep synchrony with the environment, circadian rhythms need to be entrained daily by means of Zeitgeber ("time giver") signals, such as the light-dark cycle. Recent advances in the neurophysiology and molecular biology of circadian rhythmicity allow a better understanding of synchronization. In this review we cover several aspects of the mechanisms for photic entrainment of mammalian circadian rhythms, including retinal sensitivity to light by means of novel photopigments as well as circadian variations in the retina that contribute to the regulation of retinal physiology. Downstream from the retina, we examine retinohypothalamic communication through neurotransmitter (glutamate, aspartate, pituitary adenylate cyclase-activating polypeptide) interaction with SCN receptors and the resulting signal transduction pathways in suprachiasmatic neurons, as well as putative neuron-glia interactions. Finally, we describe and analyze clock gene expression and its importance in entrainment mechanisms, as well as circadian disorders or retinal diseases related to entrainment deficits, including experimental and clinical treatments.
Collapse
Affiliation(s)
- Diego A Golombek
- Laboratory of Chronobiology, Department of Science and Technology, University of Quilmes/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Quilmes, Argentina.
| | | |
Collapse
|
40
|
Ruggiero L, Allen CN, Brown RL, Robinson DW. Mice with early retinal degeneration show differences in neuropeptide expression in the suprachiasmatic nucleus. Behav Brain Funct 2010; 6:36. [PMID: 20604961 PMCID: PMC2912232 DOI: 10.1186/1744-9081-6-36] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 07/06/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In mammals, the brain clock responsible for generating circadian rhythms is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Light entrainment of the clock occurs through intrinsically photosensitive retinal ganglion cells (ipRGCs) whose axons project to the SCN via the retinohypothalamic tract. Although ipRGCs are sufficient for photoentrainment, rod and cone photoreceptors also contribute. Adult CBA/J mice, which exhibit loss of rod and cone photoreceptors during early postnatal development, have greater numbers of ipRGCs compared to CBA/N control mice. A greater number of photosensitive cells might argue for enhanced light responses, however, these mice exhibit attenuated phase shifting behaviors. To reconcile these findings, we looked for potential differences in SCN neurons of CBA/J mice that might underly the altered circadian behaviors. We hypothesized that CBA/J mice have differences in the expression of neuropeptides in the SCN, where ipRGCs synapse. The neuropeptides vasoactive intestinal peptide (VIP) and vasopressin (VP) are expressed by many SCN neurons and play an important role in the generation of circadian rhythms and photic entrainment. METHODS Using immunohistochemistry, we looked for differences in the expression of VIP and VP in the SCN of CBA/J mice, and using a light-induced FOS assay, we also examined the degree of retinal innervation of the SCN by ipRGCs. RESULTS Our data demonstrate greater numbers of VIP-and VP-positive cells in the SCN of CBA/J mice and a greater degree of light-induced FOS expression. CONCLUSIONS These results implicate changes in neuropeptide expression in the SCN which may underlie the altered circadian responses to light in these animals.
Collapse
Affiliation(s)
- Linda Ruggiero
- Center for Research on Occupational and Environmental Toxicology, Oregon Health & Science University, Portland 97239, USA
| | | | | | | |
Collapse
|
41
|
Pantazopoulos H, Dolatshad H, Davis FC. Chronic stimulation of the hypothalamic vasoactive intestinal peptide receptor lengthens circadian period in mice and hamsters. Am J Physiol Regul Integr Comp Physiol 2010; 299:R379-85. [PMID: 20463182 DOI: 10.1152/ajpregu.00176.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Evidence suggests that circadian rhythms are regulated through diffusible signals generated by the suprachiasmatic nucleus (SCN). Vasoactive intestinal peptide (VIP) is located in SCN neurons positioned to receive photic input from the retinohypothalamic tract and transmit information to other SCN cells and adjacent hypothalamic areas. Studies using knockout mice indicate that VIP is essential for synchrony among SCN cells and for the expression of normal circadian rhythms. To test the hypothesis that VIP is also an SCN output signal, we recorded wheel-running activity rhythms in hamsters and continuously infused the VIP receptor agonist BAY 55-9837 in the third ventricle for 28 days. Unlike other candidate output signals, infusion of BAY 55-9837 did not affect activity levels. Instead, BAY 55-9837 lengthened the circadian period by 0.69 +/- 0.04 h (P < 0.0002 compared with controls). Period returned to baseline after infusions. We analyzed the effect of BAY 55-9837 on cultured SCN from PER2::LUC mice to determine if lengthening of the period by BAY 55-9837 is a direct effect on the SCN. Application of 10 muM BAY 55-9837 to SCN in culture lengthened the period of PER2 luciferase expression (24.73 +/- 0.24 h) compared with control SCN (23.57 +/- 0.26, P = 0.01). In addition, rhythm amplitude was significantly increased, consistent with increased synchronization of SCN neurons. The effect of BAY 55-9837 in vivo on period is similar to the effect of constant light. The present results suggest that VIP-VPAC2 signaling in the SCN may play two roles, synchronizing SCN neurons and setting the period of the SCN as a whole.
Collapse
Affiliation(s)
- Harry Pantazopoulos
- Department of Biology, Northeastern University, Boston, Massachusetts 02478, USA
| | | | | |
Collapse
|
42
|
Abstract
The suprachiasmatic nucleus (SCN) is the primary circadian pacemaker in mammals. Individual SCN neurons in dispersed culture can generate independent circadian oscillations of clock gene expression and neuronal firing. However, SCN rhythmicity depends on sufficient membrane depolarization and levels of intracellular calcium and cAMP. In the intact SCN, cellular oscillations are synchronized and reinforced by rhythmic synaptic input from other cells, resulting in a reproducible topographic pattern of distinct phases and amplitudes specified by SCN circuit organization. The SCN network synchronizes its component cellular oscillators, reinforces their oscillations, responds to light input by altering their phase distribution, increases their robustness to genetic perturbations, and enhances their precision. Thus, even though individual SCN neurons can be cell-autonomous circadian oscillators, neuronal network properties are integral to normal function of the SCN.
Collapse
Affiliation(s)
- David K Welsh
- Department of Psychiatry, University of California-San Diego, La Jolla, CA 92093, USA.
| | | | | |
Collapse
|
43
|
Trbovic SM. Schizophrenia as a possible dysfunction of the suprachiasmatic nucleus. Med Hypotheses 2010; 74:127-31. [DOI: 10.1016/j.mehy.2009.07.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 07/06/2009] [Accepted: 07/06/2009] [Indexed: 10/20/2022]
|
44
|
Ospeck MC, Coffey B, Freeman D. Light-dark cycle memory in the mammalian suprachiasmatic nucleus. Biophys J 2009; 97:1513-24. [PMID: 19751655 DOI: 10.1016/j.bpj.2009.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 05/18/2009] [Accepted: 06/08/2009] [Indexed: 11/15/2022] Open
Abstract
The mammalian circadian oscillator, or suprachiasmatic nucleus (SCN), contains several thousand clock neurons in its ventrolateral division, many of which are spontaneous oscillators with period lengths that range from 22 to 28 h. In complete darkness, this network synchronizes through the exchange of action potentials that release vasoactive intestinal polypeptide, striking a compromise, free-running period close to 24 h long. We entrained Siberian hamsters to various light-dark cycles and then tracked their activity into constant darkness to show that they retain a memory of the previous light-dark cycle before returning to their own free-running period. Employing Leloup-Goldbeter mammalian clock neurons we model the ventrolateral SCN network and show that light acting weakly upon a strongly rhythmic vasoactive intestinal polypeptide oscillation can explain the observed light-dark cycle memory. In addition, light is known to initiate a mitogen-activated protein kinase signaling cascade that induces transcription of both per and mkp1 phosphatase. We show that the ensuing phosphatase-kinase interaction can account for the dead zone in the mammalian phase response curve and hypothesize that the SCN behaves like a lock-in amplifier to entrain to the light edges of the circadian day.
Collapse
Affiliation(s)
- Mark C Ospeck
- Physics Department, University of Memphis, Memphis, Tennessee, USA.
| | | | | |
Collapse
|
45
|
Vaudry D, Falluel-Morel A, Bourgault S, Basille M, Burel D, Wurtz O, Fournier A, Chow BKC, Hashimoto H, Galas L, Vaudry H. Pituitary Adenylate Cyclase-Activating Polypeptide and Its Receptors: 20 Years after the Discovery. Pharmacol Rev 2009; 61:283-357. [DOI: 10.1124/pr.109.001370] [Citation(s) in RCA: 829] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
46
|
Dickson L, Finlayson K. VPAC and PAC receptors: From ligands to function. Pharmacol Ther 2008; 121:294-316. [PMID: 19109992 DOI: 10.1016/j.pharmthera.2008.11.006] [Citation(s) in RCA: 278] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Accepted: 11/18/2008] [Indexed: 02/03/2023]
Abstract
Vasoactive intestinal peptide (VIP) and the pituitary adenylate cyclase activating polypeptides (PACAPs) share 68% identity at the amino acid level and belong to the secretin peptide family. Following the initial discovery of VIP almost four decades ago a substantial amount of knowledge has been presented describing the mechanisms of action, distribution and pleiotropic functions of these related peptides. It is now known that the physiological actions of these widely distributed peptides are produced through activation of three common G-protein coupled receptors (VPAC(1), VPAC(2) and PAC(1)R) which preferentially stimulate adenylate cyclase and increase intracellular cAMP, although stimulation of other intracellular messengers, including calcium and phospholipase D, has been reported. Using a range of in vitro and in vivo approaches, including cell-based functional assays, transgenic animals and rodent models of disease, VPAC/PAC receptor activation has been associated with numerous physiological processes (e.g. control of circadian rhythms) and clinical conditions (e.g. pulmonary hypertension), which underlies on-going research efforts and makes these peptides and their cognate receptors attractive targets for the pharmaceutical industry. However, despite the considerable interest in VPAC/PAC receptors and the processes which they mediate, there is still a paucity of selective and available, non-peptide ligands, which has hindered further advances in this field both at the basic research and clinical level. This review summarises the current knowledge of VIP/PACAP and the VPAC/PAC receptors with regard to their distribution, pharmacology, signalling pathways, splice variants and finally, the utility of animal models in exploring their physiological roles.
Collapse
Affiliation(s)
- Louise Dickson
- Centre for Integrative Physiology, University of Edinburgh, EH8 9XD, UK
| | | |
Collapse
|
47
|
Vosko AM, Schroeder A, Loh DH, Colwell CS. Vasoactive intestinal peptide and the mammalian circadian system. Gen Comp Endocrinol 2007; 152:165-75. [PMID: 17572414 PMCID: PMC1994114 DOI: 10.1016/j.ygcen.2007.04.018] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 04/17/2007] [Accepted: 04/19/2007] [Indexed: 11/25/2022]
Abstract
In mammals, the circadian oscillators that drive daily behavioral and endocrine rhythms are located in the hypothalamic suprachiasmatic nucleus (SCN). While the SCN is anatomically well-situated to receive and transmit temporal cues to the rest of the brain and periphery, there are many holes in our understanding of how this temporal regulation occurs. Unanswered questions include how cell autonomous circadian oscillations within the SCN remain synchronized to each other as well as communicate temporal information to downstream targets. In recent years, it has become clear that neuropeptides are critically involved in circadian timekeeping. One such neuropeptide, vasoactive intestinal peptide (VIP), defines a cell population within the SCN and is likely used as a signaling molecule by these neurons. Converging lines of evidence suggest that the loss of VIP or its receptor has a major influence on the ability of the SCN neurons to generate circadian oscillations as well as synchronize these cellular oscillations. VIP, acting through the VPAC(2) receptor, exerts these effects in the SCN by activating intracellular signaling pathways and, consequently, modulating synaptic transmission and intrinsic membrane currents. Anatomical evidence suggests that these VIP expressing neurons connect both directly and indirectly to endocrine and other output targets. Striking similarities exist between the role of VIP in mammals and the role of Pigment Dispersing Factor (PDF), a functionally related neuropeptide, in the Drosophila circadian system. Work in both mammals and insects suggests that further research into neuropeptide function is necessary to understand how circadian oscillators work as a coordinated system to impose a temporal structure on physiological processes within the organism.
Collapse
Affiliation(s)
- Andrew M Vosko
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience, University of California--Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90024-1759, USA
| | | | | | | |
Collapse
|
48
|
To TL, Henson MA, Herzog ED, Doyle FJ. A molecular model for intercellular synchronization in the mammalian circadian clock. Biophys J 2007; 92:3792-803. [PMID: 17369417 PMCID: PMC1868999 DOI: 10.1529/biophysj.106.094086] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Accepted: 01/22/2007] [Indexed: 11/18/2022] Open
Abstract
The mechanisms and consequences of synchrony among heterogeneous oscillators are poorly understood in biological systems. We present a multicellular, molecular model of the mammalian circadian clock that incorporates recent data implicating the neurotransmitter vasoactive intestinal polypeptide (VIP) as the key synchronizing agent. The model postulates that synchrony arises among circadian neurons because they release VIP rhythmically on a daily basis and in response to ambient light. Two basic cell types, intrinsically rhythmic pacemakers and damped oscillators, are assumed to arise from a distribution of Period gene transcription rates. Postsynaptic neurons show time-of-day dependent responses to VIP binding through a signaling cascade that activates Period mRNA transcription. The heterogeneous cell ensemble model self-synchronizes, entrains to ambient light-dark cycles, and desynchronizes in constant bright light or upon removal of VIP signaling. The degree of synchronicity observed depends on cell-specific features (e.g., mean and variability of parameters within the rhythm-generating loop), in addition to the more commonly studied effect of intercellular coupling strength. These simulations closely replicate experimental data and predict that heterogeneous oscillations (e.g., sustained, damped, and arrhythmic) arise from small differences in the molecular parameters between cells, that damped oscillators participate in entrainment and synchrony of the ensemble of cells, and that constant light desynchronizes oscillators by maximizing VIP release.
Collapse
Affiliation(s)
- Tsz-Leung To
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | | | | |
Collapse
|
49
|
Hao YH, Yong HY, Murphy CN, Wax D, Samuel M, Rieke A, Lai L, Liu Z, Durtschi DC, Welbern VR, Price EM, McAllister RM, Turk JR, Laughlin MH, Prather RS, Rucker EB. Production of endothelial nitric oxide synthase (eNOS) over-expressing piglets. Transgenic Res 2006; 15:739-50. [PMID: 17080303 DOI: 10.1007/s11248-006-9020-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2006] [Accepted: 06/20/2006] [Indexed: 10/24/2022]
Abstract
Vascular function, vascular structure, and homeostasis are thought to be regulated in part by nitric oxide (NO) released by endothelial cell nitric oxide synthase (eNOS), and NO released by eNOS plays an important role in modulating metabolism of skeletal and cardiac muscle in health and disease. The pig is an optimal model for human diseases because of the large number of important similarities between the genomic, metabolic and cardiovascular systems of pigs and humans. To gain a better understanding of cardiovascular regulation by eNOS we produced pigs carrying an endogenous eNOS gene driven by a Tie-2 promoter and tagged with a V5 His tag. Nuclear transfer was conducted to create these animals and the effects of two different oocyte activation treatments and two different culture systems were examined. Donor cells were electrically fused to the recipient oocytes. Electrical fusion/activation (1 mM calcium in mannitol: Treatment 1) and electrical fusion (0.1 mM calcium in mannitol)/chemical activation (200 microM Thimerosal for 10 min followed by 8 mM DTT for 30 min: Treatment 2) were used. Embryos were surgically transferred to the oviducts of gilts that exhibited estrus on the day of fusion or the day of transfer. Two cloned transgenic piglets were born from Treatment 1 and low oxygen, and another two from Treatment 2 and normal oxygen. PCR, RT-PCR, Western blotting and immunohistochemistry confirmed that the pigs were transgenic, made message, made the fusion protein and that the fusion protein localized to the endothelial cells of placental vasculature from the conceptuses as did the endogenous eNOS. Thus both activation conditions and culture systems are compatible with development to term. These pigs will serve as the founders for a colony of miniature pigs that will help to elucidate the function of eNOS in regulating muscle metabolism and the cardiorespiratory system.
Collapse
Affiliation(s)
- Y H Hao
- Division of Animal Sciences, University of Missouri-Columbia, Columbia, MO, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lang B, Song B, Davidson W, MacKenzie A, Smith N, McCaig CD, Harmar AJ, Shen S. Expression of the human PAC1 receptor leads to dose-dependent hydrocephalus-related abnormalities in mice. J Clin Invest 2006; 116:1924-34. [PMID: 16823490 PMCID: PMC1483148 DOI: 10.1172/jci27597] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Accepted: 04/25/2006] [Indexed: 01/16/2023] Open
Abstract
Hydrocephalus is a common and potentially devastating birth defect affecting the CNS, and its relationship with G protein-coupled receptors (GPCRs) is unknown. We have expressed 2, 4, or 6 copies of a GPCR--the human PAC1 receptor with a 130-kb transgene in the mouse nervous system in a pattern closely resembling that of the endogenous gene. Consistent with PAC1 actions, PKA and PKC activity were elevated in the brains of Tg mice. Remarkably, Tg mice developed dose-dependent hydrocephalus-like characteristics, including enlarged third and lateral ventricles and reduced cerebral cortex, corpus callosum, and subcommissural organ (SCO). Neuronal proliferation and apoptosis were implicated in hydrocephalus, and we observed significantly reduced neuronal proliferation and massively increased neuronal apoptosis in the developing cortex and SCO of Tg embryos, while neurite outgrowth and neuronal migration in vitro remain uncompromised. Ventricular ependymal cilia are crucial for directing cerebrospinal fluid flow, and ependyma of Tg mice exhibited disrupted cilia with increased phospho-CREB immunoreactivity. These data demonstrate that altered neuronal proliferation/apoptosis and disrupted ependymal cilia are the main factors contributing to hydrocephalus in PAC1-overexpressing mice. This is the first report to our knowledge demonstrating that misregulation of GPCRs can be involved in hydrocephalus-related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Bing Lang
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.
Ultrasound Department, Aberdeen Maternity Hospital, Grampian University Hospitals NHS Trust, Aberdeen, United Kingdom.
Centre for Neuroscience Research, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Bing Song
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.
Ultrasound Department, Aberdeen Maternity Hospital, Grampian University Hospitals NHS Trust, Aberdeen, United Kingdom.
Centre for Neuroscience Research, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Wendy Davidson
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.
Ultrasound Department, Aberdeen Maternity Hospital, Grampian University Hospitals NHS Trust, Aberdeen, United Kingdom.
Centre for Neuroscience Research, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Alastair MacKenzie
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.
Ultrasound Department, Aberdeen Maternity Hospital, Grampian University Hospitals NHS Trust, Aberdeen, United Kingdom.
Centre for Neuroscience Research, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Norman Smith
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.
Ultrasound Department, Aberdeen Maternity Hospital, Grampian University Hospitals NHS Trust, Aberdeen, United Kingdom.
Centre for Neuroscience Research, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Colin D. McCaig
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.
Ultrasound Department, Aberdeen Maternity Hospital, Grampian University Hospitals NHS Trust, Aberdeen, United Kingdom.
Centre for Neuroscience Research, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Anthony J. Harmar
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.
Ultrasound Department, Aberdeen Maternity Hospital, Grampian University Hospitals NHS Trust, Aberdeen, United Kingdom.
Centre for Neuroscience Research, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Sanbing Shen
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.
Ultrasound Department, Aberdeen Maternity Hospital, Grampian University Hospitals NHS Trust, Aberdeen, United Kingdom.
Centre for Neuroscience Research, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|