51
|
Multi-Modal Regulation of Circadian Physiology by Interactive Features of Biological Clocks. BIOLOGY 2021; 11:biology11010021. [PMID: 35053019 PMCID: PMC8772734 DOI: 10.3390/biology11010021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/26/2022]
Abstract
The circadian clock is a fundamental biological timing mechanism that generates nearly 24 h rhythms of physiology and behaviors, including sleep/wake cycles, hormone secretion, and metabolism. Evolutionarily, the endogenous clock is thought to confer living organisms, including humans, with survival benefits by adapting internal rhythms to the day and night cycles of the local environment. Mirroring the evolutionary fitness bestowed by the circadian clock, daily mismatches between the internal body clock and environmental cycles, such as irregular work (e.g., night shift work) and life schedules (e.g., jet lag, mistimed eating), have been recognized to increase the risk of cardiac, metabolic, and neurological diseases. Moreover, increasing numbers of studies with cellular and animal models have detected the presence of functional circadian oscillators at multiple levels, ranging from individual neurons and fibroblasts to brain and peripheral organs. These oscillators are tightly coupled to timely modulate cellular and bodily responses to physiological and metabolic cues. In this review, we will discuss the roles of central and peripheral clocks in physiology and diseases, highlighting the dynamic regulatory interactions between circadian timing systems and multiple metabolic factors.
Collapse
|
52
|
Kim S, McMahon DG. Light sets the brain's daily clock by regional quickening and slowing of the molecular clockworks at dawn and dusk. eLife 2021; 10:e70137. [PMID: 34927581 PMCID: PMC8687663 DOI: 10.7554/elife.70137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 12/11/2021] [Indexed: 12/15/2022] Open
Abstract
How daily clocks in the brain are set by light to local environmental time and encode the seasons is not fully understood. The suprachiasmatic nucleus (SCN) is a central circadian clock in mammals that orchestrates physiology and behavior in tune with daily and seasonal light cycles. Here, we have found that optogenetically simulated light input to explanted mouse SCN changes the waveform of the molecular clockworks from sinusoids in free-running conditions to highly asymmetrical shapes with accelerated synthetic (rising) phases and extended degradative (falling) phases marking clock advances and delays at simulated dawn and dusk. Daily waveform changes arise under ex vivo entrainment to simulated winter and summer photoperiods, and to non-24 hr periods. Ex vivo SCN imaging further suggests that acute waveform shifts are greatest in the ventrolateral SCN, while period effects are greatest in the dorsomedial SCN. Thus, circadian entrainment is encoded by SCN clock gene waveform changes that arise from spatiotemporally distinct intrinsic responses within the SCN neural network.
Collapse
Affiliation(s)
- Suil Kim
- Vanderbilt Brain Institute, Vanderbilt UniversityNashvilleUnited States
| | - Douglas G McMahon
- Vanderbilt Brain Institute, Vanderbilt UniversityNashvilleUnited States
- Department of Biological Sciences, Vanderbilt UniversityNashvilleUnited States
| |
Collapse
|
53
|
Nakata M, Kumari P, Kita R, Katsui N, Takeuchi Y, Kawaguchi T, Yamazaki T, Zhang B, Shimba S, Yada T. Circadian Clock Component BMAL1 in the Paraventricular Nucleus Regulates Glucose Metabolism. Nutrients 2021; 13:4487. [PMID: 34960038 PMCID: PMC8707417 DOI: 10.3390/nu13124487] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 11/17/2022] Open
Abstract
It is suggested that clock genes link the circadian rhythm to glucose and lipid metabolism. In this study, we explored the role of the clock gene Bmal1 in the hypothalamic paraventricular nucleus (PVN) in glucose metabolism. The Sim1-Cre-mediated deletion of Bmal1 markedly reduced insulin secretion, resulting in impaired glucose tolerance. The pancreatic islets' responses to glucose, sulfonylureas (SUs) and arginine vasopressin (AVP) were well maintained. To specify the PVN neuron subpopulation targeted by Bmal1, the expression of neuropeptides was examined. In these knockout (KO) mice, the mRNA expression of Avp in the PVN was selectively decreased, and the plasma AVP concentration was also decreased. However, fasting suppressed Avp expression in both KO and Cre mice. These results demonstrate that PVN BMAL1 maintains Avp expression in the PVN and release to the circulation, possibly providing islet β-cells with more AVP. This action helps enhance insulin release and, consequently, glucose tolerance. In contrast, the circadian variation of Avp expression is regulated by feeding, but not by PVN BMAL1.
Collapse
Affiliation(s)
- Masanori Nakata
- Department of Physiology, Wakayama Medical University School of Medicine, Kimiidare 811-1, Wakayama 641-8509, Japan; (R.K.); (N.K.); (Y.T.); (T.K.); (B.Z.)
| | - Parmila Kumari
- Department of Biotechnology, University of Wroclaw, Plac Uniwersytecki 1, 50-137 Wroclaw, Poland;
| | - Rika Kita
- Department of Physiology, Wakayama Medical University School of Medicine, Kimiidare 811-1, Wakayama 641-8509, Japan; (R.K.); (N.K.); (Y.T.); (T.K.); (B.Z.)
| | - Nanako Katsui
- Department of Physiology, Wakayama Medical University School of Medicine, Kimiidare 811-1, Wakayama 641-8509, Japan; (R.K.); (N.K.); (Y.T.); (T.K.); (B.Z.)
| | - Yuriko Takeuchi
- Department of Physiology, Wakayama Medical University School of Medicine, Kimiidare 811-1, Wakayama 641-8509, Japan; (R.K.); (N.K.); (Y.T.); (T.K.); (B.Z.)
| | - Tomoki Kawaguchi
- Department of Physiology, Wakayama Medical University School of Medicine, Kimiidare 811-1, Wakayama 641-8509, Japan; (R.K.); (N.K.); (Y.T.); (T.K.); (B.Z.)
| | - Toshiya Yamazaki
- Department of Health Sciences, Kansai University of Health Sciences, Wakaba 2-11-1, Kumatoricho, Sennan-gun, Osaka 590-0482, Japan;
| | - Boyang Zhang
- Department of Physiology, Wakayama Medical University School of Medicine, Kimiidare 811-1, Wakayama 641-8509, Japan; (R.K.); (N.K.); (Y.T.); (T.K.); (B.Z.)
| | - Shigeki Shimba
- Laboratory of Health Science, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabshi 274-8555, Japan;
| | - Toshihiko Yada
- Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, 1-5-6 Minatojimaminamimachi, Chuou-ku, Kobe 650-0047, Japan;
- Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, Kusunokicho 7-5-1, Chuou-ku, Kobe 650-0017, Japan
| |
Collapse
|
54
|
Thirtamara Rajamani K, Leithead AB, Kim M, Barbier M, Peruggia M, Niblo K, Barteczko L, Lefevre A, Grinevich V, Harony-Nicolas H. Efficiency of cell-type specific and generic promoters in transducing oxytocin neurons and monitoring their neural activity during lactation. Sci Rep 2021; 11:22541. [PMID: 34795340 PMCID: PMC8602291 DOI: 10.1038/s41598-021-01818-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 11/02/2021] [Indexed: 12/31/2022] Open
Abstract
Hypothalamic oxytocin (OXT) and arginine-vasopressin (AVP) neurons have been at the center of several physiological and behavioral studies. Advances in viral vector biology and the development of transgenic rodent models have allowed for targeted gene expression to study the functions of specific cell populations and brain circuits. In this study, we compared the efficiency of various adeno-associated viral vectors in these cell populations and demonstrated that none of the widely used promoters were, on their own, effective at driving expression of a down-stream fluorescent protein in OXT or AVP neurons. As anticipated, the OXT promoter could efficiently drive gene expression in OXT neurons and this efficiency is solely attributed to the promoter and not the viral serotype. We also report that a dual virus approach using an OXT promoter driven Cre recombinase significantly improved the efficiency of viral transduction in OXT neurons. Finally, we demonstrate the utility of the OXT promoter for conducting functional studies on OXT neurons by using an OXT specific viral system to record neural activity of OXT neurons in lactating female rats across time. We conclude that extreme caution is needed when employing non-neuron-specific viral approaches/promoters to study neural populations within the paraventricular nucleus of the hypothalamus.
Collapse
Affiliation(s)
- Keerthi Thirtamara Rajamani
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda B Leithead
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michelle Kim
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marie Barbier
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Peruggia
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristi Niblo
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lara Barteczko
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Arthur Lefevre
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Hala Harony-Nicolas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
55
|
Gpr19 is a circadian clock-controlled orphan GPCR with a role in modulating free-running period and light resetting capacity of the circadian clock. Sci Rep 2021; 11:22406. [PMID: 34789778 PMCID: PMC8599615 DOI: 10.1038/s41598-021-01764-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/03/2021] [Indexed: 01/26/2023] Open
Abstract
Gpr19 encodes an evolutionarily conserved orphan G-protein-coupled receptor (GPCR) with currently no established physiological role in vivo. We characterized Gpr19 expression in the suprachiasmatic nucleus (SCN), the locus of the master circadian clock in the brain, and determined its role in the context of the circadian rhythm regulation. We found that Gpr19 is mainly expressed in the dorsal part of the SCN, with its expression fluctuating in a circadian fashion. A conserved cAMP-responsive element in the Gpr19 promoter was able to produce circadian transcription in the SCN. Gpr19−/− mice exhibited a prolonged circadian period and a delayed initiation of daily locomotor activity. Gpr19 deficiency caused the downregulation of several genes that normally peak during the night, including Bmal1 and Gpr176. In response to light exposure at night, Gpr19−/− mice had a reduced capacity for light-induced phase-delays, but not for phase-advances. This defect was accompanied by reduced response of c-Fos expression in the dorsal region of the SCN, while apparently normal in the ventral area of the SCN, in Gpr19−/− mice. Thus, our data demonstrate that Gpr19 is an SCN-enriched orphan GPCR with a distinct role in circadian regulation and may provide a potential target option for modulating the circadian clock.
Collapse
|
56
|
Ang G, Brown LA, Tam SKE, Davies KE, Foster RG, Harrison PJ, Sprengel R, Vyazovskiy VV, Oliver PL, Bannerman DM, Peirson SN. Deletion of AMPA receptor GluA1 subunit gene (Gria1) causes circadian rhythm disruption and aberrant responses to environmental cues. Transl Psychiatry 2021; 11:588. [PMID: 34782594 PMCID: PMC8593011 DOI: 10.1038/s41398-021-01690-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022] Open
Abstract
Dysfunction of the glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor GluA1 subunit and deficits in synaptic plasticity are implicated in schizophrenia and sleep and circadian rhythm disruption. To investigate the role of GluA1 in circadian and sleep behaviour, we used wheel-running, passive-infrared, and video-based home-cage activity monitoring to assess daily rest-activity profiles of GluA1-knockout mice (Gria1-/-). We showed that these mice displayed various circadian abnormalities, including misaligned, fragmented, and more variable rest-activity patterns. In addition, they showed heightened, but transient, behavioural arousal to light→dark and dark→light transitions, as well as attenuated nocturnal-light-induced activity suppression (negative masking). In the hypothalamic suprachiasmatic nuclei (SCN), nocturnal-light-induced cFos signals (a molecular marker of neuronal activity in the preceding ~1-2 h) were attenuated, indicating reduced light sensitivity in the SCN. However, there was no change in the neuroanatomical distribution of expression levels of two neuropeptides-vasoactive intestinal peptide (VIP) and arginine vasopressin (AVP)-differentially expressed in the core (ventromedial) vs. shell (dorsolateral) SCN subregions and both are known to be important for neuronal synchronisation within the SCN and circadian rhythmicity. In the motor cortex (area M1/M2), there was increased inter-individual variability in cFos levels during the evening period, mirroring the increased inter-individual variability in locomotor activity under nocturnal light. Finally, in the spontaneous odour recognition task GluA1 knockouts' short-term memory was impaired due to enhanced attention to the recently encountered familiar odour. These abnormalities due to altered AMPA-receptor-mediated signalling resemble and may contribute to sleep and circadian rhythm disruption and attentional deficits in different modalities in schizophrenia.
Collapse
Affiliation(s)
- Gauri Ang
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Experimental Psychology, University of Oxford, Oxford, UK
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Laurence A Brown
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- IT Services, University of Oxford, Oxford, UK
| | - Shu K E Tam
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Kay E Davies
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Russell G Foster
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Rolf Sprengel
- Research Group of the Max Planck Institute for Medical Research at the Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Vladyslav V Vyazovskiy
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Peter L Oliver
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, UK.
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK.
| | - Stuart N Peirson
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| |
Collapse
|
57
|
Sgro M, Kodila ZN, Brady RD, Reichelt AC, Mychaisuk R, Yamakawa GR. Synchronizing Our Clocks as We Age: The Influence of the Brain-Gut-Immune Axis on the Sleep-Wake Cycle Across the Lifespan. Sleep 2021; 45:6425072. [PMID: 34757429 DOI: 10.1093/sleep/zsab268] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/11/2021] [Indexed: 11/12/2022] Open
Abstract
The microbes that colonize the small and large intestines, known as the gut microbiome, play an integral role in optimal brain development and function. The gut microbiome is a vital component of the bi-directional communication pathway between the brain, immune system, and gut, also known as the brain-gut-immune axis. To date there has been minimal investigation into the implications of improper development of the gut microbiome and the brain-gut-immune axis on the sleep-wake cycle, particularly during sensitive periods of physical and neurological development, such as childhood, adolescence, and senescence. Therefore, this review will explore the current literature surrounding the overlapping developmental periods of the gut microbiome, brain, and immune system from birth through to senescence, while highlighting how the brain-gut-immune axis affects maturation and organisation of the sleep-wake cycle. We also examine how dysfunction to either the microbiome or the sleep-wake cycle negatively affects the bidirectional relationship between the brain and gut, and subsequently the overall health and functionality of this complex system. Additionally, this review integrates therapeutic studies to demonstrate when dietary manipulations, such as supplementation with probiotics and prebiotics, can modulate the gut microbiome to enhance health of the brain-gut-immune axis and optimize our sleep-wake cycle.
Collapse
Affiliation(s)
- Marissa Sgro
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Zoe N Kodila
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Amy C Reichelt
- Department of Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Richelle Mychaisuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
58
|
Morris EL, Patton AP, Chesham JE, Crisp A, Adamson A, Hastings MH. Single-cell transcriptomics of suprachiasmatic nuclei reveal a Prokineticin-driven circadian network. EMBO J 2021; 40:e108614. [PMID: 34487375 PMCID: PMC8521297 DOI: 10.15252/embj.2021108614] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 11/22/2022] Open
Abstract
Circadian rhythms in mammals are governed by the hypothalamic suprachiasmatic nucleus (SCN), in which 20,000 clock cells are connected together into a powerful time‐keeping network. In the absence of network‐level cellular interactions, the SCN fails as a clock. The topology and specific roles of its distinct cell populations (nodes) that direct network functions are, however, not understood. To characterise its component cells and network structure, we conducted single‐cell sequencing of SCN organotypic slices and identified eleven distinct neuronal sub‐populations across circadian day and night. We defined neuropeptidergic signalling axes between these nodes, and built neuropeptide‐specific network topologies. This revealed their temporal plasticity, being up‐regulated in circadian day. Through intersectional genetics and real‐time imaging, we interrogated the contribution of the Prok2‐ProkR2 neuropeptidergic axis to network‐wide time‐keeping. We showed that Prok2‐ProkR2 signalling acts as a key regulator of SCN period and rhythmicity and contributes to defining the network‐level properties that underpin robust circadian co‐ordination. These results highlight the diverse and distinct contributions of neuropeptide‐modulated communication of temporal information across the SCN.
Collapse
Affiliation(s)
- Emma L Morris
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Andrew P Patton
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Johanna E Chesham
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Alastair Crisp
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Antony Adamson
- The Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Michael H Hastings
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
59
|
Coomans C, Saaltink DJ, Deboer T, Tersteeg M, Lanooij S, Schneider AF, Mulder A, van Minnen J, Jost C, Koster AJ, Vreugdenhil E. Doublecortin-like expressing astrocytes of the suprachiasmatic nucleus are implicated in the biosynthesis of vasopressin and influences circadian rhythms. Glia 2021; 69:2752-2766. [PMID: 34343377 PMCID: PMC9291169 DOI: 10.1002/glia.24069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 11/09/2022]
Abstract
We have recently identified a novel plasticity protein, doublecortin-like (DCL), that is specifically expressed in the shell of the mouse suprachiasmatic nucleus (SCN). DCL is implicated in neuroplastic events, such as neurogenesis, that require structural rearrangements of the microtubule cytoskeleton, enabling dynamic movements of cell bodies and dendrites. We have inspected DCL expression in the SCN by confocal microscopy and found that DCL is expressed in GABA transporter-3 (GAT3)-positive astrocytes that envelope arginine vasopressin (AVP)-expressing cells. To investigate the role of these DCL-positive astrocytes in circadian rhythmicity, we have used transgenic mice expressing doxycycline-induced short-hairpin (sh) RNA's targeting DCL mRNA (DCL knockdown mice). Compared with littermate wild type (WT) controls, DCL-knockdown mice exhibit significant shorter circadian rest-activity periods in constant darkness and adjusted significantly faster to a jet-lag protocol. As DCL-positive astrocytes are closely associated with AVP-positive cells, we analyzed AVP expression in DCL-knockdown mice and in their WT littermates by 3D reconstructions and transmission electron microscopy (TEM). We found significantly higher numbers of AVP-positive cells with increased volume and more intensity in DCL-knockdown mice. We found alterations in the numbers of dense core vesicle-containing neurons at ZT8 and ZT20 suggesting that the peak and trough of neuropeptide biosynthesis is dampened in DCL-knockdown mice compared to WT littermates. Together, our data suggest an important role for the astrocytic plasticity in the regulation of circadian rhythms and point to the existence of a specific DCL+ astrocyte-AVP+ neuronal network located in the dorsal SCN implicated in AVP biosynthesis.
Collapse
Affiliation(s)
- Claudia Coomans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk-Jan Saaltink
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom Deboer
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mayke Tersteeg
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne Lanooij
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne Fleur Schneider
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Aat Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan van Minnen
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carolina Jost
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Abraham J Koster
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erno Vreugdenhil
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
60
|
Whylings J, Rigney N, de Vries GJ, Petrulis A. Reduction in vasopressin cells in the suprachiasmatic nucleus in mice increases anxiety and alters fluid intake. Horm Behav 2021; 133:104997. [PMID: 34062279 PMCID: PMC8529700 DOI: 10.1016/j.yhbeh.2021.104997] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/21/2021] [Accepted: 05/10/2021] [Indexed: 11/19/2022]
Abstract
Central vasopressin (AVP) has been implicated in the control of multiple behaviors, including social behavior, anxiety-like behavior, and sickness behavior. The extent to which the different AVP-producing cell groups contribute to regulating these behaviors has not been extensively investigated. Here we test the role of AVP cells in the suprachiasmatic nucleus (SCN) in these behaviors by ablating these cells using viral-mediated, Cre-dependent caspase in male and female AVP-Cre + mice and Cre-controls. We compared anxiety and social behaviors, as well as sickness behaviors (lethargy, anhedonia (indexed by sucrose consumption), and changes in anxiety-like- and social behavior) induced via injection of bacterial lipopolysaccharide (LPS). We found that SCN AVP cell ablation increased anxiety-like behavior and sucrose consumption in both sexes, as well as increased urine marking by males in a non-social context, but did not alter behavioral responses to sickness. Our data suggest that SCN AVP does not strongly affect LPS-induced behavioral changes, but may contribute to anxiety-like behavior, and may play a role in ingestive reward/motivation and fluid intake.
Collapse
Affiliation(s)
- Jack Whylings
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| | - Nicole Rigney
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Geert J de Vries
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA; Department of Biology, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Aras Petrulis
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| |
Collapse
|
61
|
Hubbard J, Kobayashi Frisk M, Ruppert E, Tsai JW, Fuchs F, Robin-Choteau L, Husse J, Calvel L, Eichele G, Franken P, Bourgin P. Dissecting and modeling photic and melanopsin effects to predict sleep disturbances induced by irregular light exposure in mice. Proc Natl Acad Sci U S A 2021; 118:e2017364118. [PMID: 34155139 PMCID: PMC8237663 DOI: 10.1073/pnas.2017364118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Artificial lighting, day-length changes, shift work, and transmeridian travel all lead to sleep-wake disturbances. The nychthemeral sleep-wake cycle (SWc) is known to be controlled by output from the central circadian clock in the suprachiasmatic nuclei (SCN), which is entrained to the light-dark cycle. Additionally, via intrinsically photosensitive retinal ganglion cells containing the photopigment melanopsin (Opn4), short-term light-dark alternations exert direct and acute influences on sleep and waking. However, the extent to which longer exposures typically experienced across the 24-h day exert such an effect has never been clarified or quantified, as disentangling sustained direct light effects (SDLE) from circadian effects is difficult. Recording sleep in mice lacking a circadian pacemaker, either through transgenesis (Syt10cre/creBmal1fl/- ) or SCN lesioning and/or melanopsin-based phototransduction (Opn4-/- ), we uncovered, contrary to prevailing assumptions, that the contribution of SDLE is as important as circadian-driven input in determining SWc amplitude. Specifically, SDLE were primarily mediated (>80%) through melanopsin, of which half were then relayed through the SCN, revealing an ancillary purpose for this structure, independent of its clock function in organizing SWc. Based on these findings, we designed a model to estimate the effect of atypical light-dark cycles on SWc. This model predicted SWc amplitude in mice exposed to simulated transequatorial or transmeridian paradigms. Taken together, we demonstrate this SDLE is a crucial mechanism influencing behavior on par with the circadian system. In a broader context, these findings mandate considering SDLE, in addition to circadian drive, for coping with health consequences of atypical light exposure in our society.
Collapse
Affiliation(s)
- Jeffrey Hubbard
- CNRS-Unité Propre de Recherche (UPR) 3212, Institute of Cellular and Integrative Neurosciences, 67084 Strasbourg, France
- International Research Center for ChronoSomnology, Translational Medicine Federation Strasbourg, Sleep Disorders Center, Strasbourg University Hospital, University of Strasbourg, 67000 Strasbourg, France
| | - Mio Kobayashi Frisk
- CNRS-Unité Propre de Recherche (UPR) 3212, Institute of Cellular and Integrative Neurosciences, 67084 Strasbourg, France
- International Research Center for ChronoSomnology, Translational Medicine Federation Strasbourg, Sleep Disorders Center, Strasbourg University Hospital, University of Strasbourg, 67000 Strasbourg, France
| | - Elisabeth Ruppert
- CNRS-Unité Propre de Recherche (UPR) 3212, Institute of Cellular and Integrative Neurosciences, 67084 Strasbourg, France
- International Research Center for ChronoSomnology, Translational Medicine Federation Strasbourg, Sleep Disorders Center, Strasbourg University Hospital, University of Strasbourg, 67000 Strasbourg, France
| | - Jessica W Tsai
- Department of Biology, Stanford University, Stanford, CA 94305
| | - Fanny Fuchs
- CNRS-Unité Propre de Recherche (UPR) 3212, Institute of Cellular and Integrative Neurosciences, 67084 Strasbourg, France
- International Research Center for ChronoSomnology, Translational Medicine Federation Strasbourg, Sleep Disorders Center, Strasbourg University Hospital, University of Strasbourg, 67000 Strasbourg, France
| | - Ludivine Robin-Choteau
- CNRS-Unité Propre de Recherche (UPR) 3212, Institute of Cellular and Integrative Neurosciences, 67084 Strasbourg, France
- European Center for Diabetes Studies, 67200 Strasbourg, France
| | - Jana Husse
- Department of Genes and Behavior, Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany
| | - Laurent Calvel
- CNRS-Unité Propre de Recherche (UPR) 3212, Institute of Cellular and Integrative Neurosciences, 67084 Strasbourg, France
- International Research Center for ChronoSomnology, Translational Medicine Federation Strasbourg, Sleep Disorders Center, Strasbourg University Hospital, University of Strasbourg, 67000 Strasbourg, France
| | - Gregor Eichele
- Department of Genes and Behavior, Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany
| | - Paul Franken
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Patrice Bourgin
- CNRS-Unité Propre de Recherche (UPR) 3212, Institute of Cellular and Integrative Neurosciences, 67084 Strasbourg, France;
- International Research Center for ChronoSomnology, Translational Medicine Federation Strasbourg, Sleep Disorders Center, Strasbourg University Hospital, University of Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
62
|
Hughes ATL, Samuels RE, Baño-Otálora B, Belle MDC, Wegner S, Guilding C, Northeast RC, Loudon ASI, Gigg J, Piggins HD. Timed daily exercise remodels circadian rhythms in mice. Commun Biol 2021; 4:761. [PMID: 34145388 PMCID: PMC8213798 DOI: 10.1038/s42003-021-02239-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/18/2021] [Indexed: 01/26/2023] Open
Abstract
Regular exercise is important for physical and mental health. An underexplored and intriguing property of exercise is its actions on the body’s 24 h or circadian rhythms. Molecular clock cells in the brain’s suprachiasmatic nuclei (SCN) use electrical and chemical signals to orchestrate their activity and convey time of day information to the rest of the brain and body. To date, the long-lasting effects of regular physical exercise on SCN clock cell coordination and communication remain unresolved. Utilizing mouse models in which SCN intercellular neuropeptide signaling is impaired as well as those with intact SCN neurochemical signaling, we examined how daily scheduled voluntary exercise (SVE) influenced behavioral rhythms and SCN molecular and neuronal activities. We show that in mice with disrupted neuropeptide signaling, SVE promotes SCN clock cell synchrony and robust 24 h rhythms in behavior. Interestingly, in both intact and neuropeptide signaling deficient animals, SVE reduces SCN neural activity and alters GABAergic signaling. These findings illustrate the potential utility of regular exercise as a long-lasting and effective non-invasive intervention in the elderly or mentally ill where circadian rhythms can be blunted and poorly aligned to the external world. Using mice with disrupted neuropeptide signaling, Hughes et al. show that daily scheduled voluntary exercise (SVE) promotes suprachiasmatic nuclei (SCN) clock cell synchrony and robust 24 h rhythms in behavior. This study suggests the potential utility of regular exercise as a non-invasive intervention for the elderly or mentally ill, where circadian rhythms can be poorly aligned to the external world.
Collapse
Affiliation(s)
- Alun Thomas Lloyd Hughes
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,School of Biological and Environmental Sciences, Liverpool John Moores University, Liverpool, UK
| | - Rayna Eve Samuels
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Beatriz Baño-Otálora
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Mino David Charles Belle
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,University of Exeter Medical School, Exeter, UK
| | - Sven Wegner
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Clare Guilding
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,School of Medical Education, Newcastle University, Newcastle, UK
| | | | | | - John Gigg
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Hugh David Piggins
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK. .,School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, UK.
| |
Collapse
|
63
|
Wei HH, Yuan XS, Chen ZK, Chen PP, Xiang Z, Qu WM, Li RX, Zhou GM, Huang ZL. Presynaptic inputs to vasopressin neurons in the hypothalamic supraoptic nucleus and paraventricular nucleus in mice. Exp Neurol 2021; 343:113784. [PMID: 34139240 DOI: 10.1016/j.expneurol.2021.113784] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 05/29/2021] [Accepted: 06/13/2021] [Indexed: 11/29/2022]
Abstract
Arginine vasopressin (AVP) neurons in the hypothalamic supraoptic nucleus (SON) and paraventricular nucleus (PVN) are involved in important physiological behaviors, such as controling osmotic stability and thermoregulation. However, the presynaptic input patterns governing AVP neurons have remained poorly understood due to their heterogeneity, as well as intermingling of AVP neurons with other neurons both in the SON and PVN. In the present study, we employed a retrograde modified rabies-virus system to reveal the brain areas that provide specific inputs to AVP neurons in the SON and PVN. We found that AVP neurons of the SON and PVN received similar input patterns from multiple areas of the brain, particularly massive afferent inputs from the diencephalon and other brain regions of the limbic system; however, PVNAVP neurons received relatively broader and denser inputs compared to SONAVP neurons. Additionally, SONAVP neurons received more projections from the median preoptic nucleus and organum vasculosum of the lamina terminalis (a circumventricular organ), compared to PVNAVP neurons, while PVNAVP neurons received more afferent inputs from the bed nucleus of stria terminalis and dorsomedial nucleus of the hypothalamus, both of which are thermoregulatory nuclei, compared to those of SONAVP neurons. In addition, both SONAVP and PVNAVP neurons received direct afferent projections from the bilateral suprachiasmatic nucleus, which is the master regulator of circadian rhythms and is concomitantly responsible for fluctuations in AVP levels. Taken together, our present results provide a comprehensive understanding of the specific afferent framework of AVP neurons both in the SON and PVN, and lay the foundation for further dissecting the diverse roles of SONAVP and PVNAVP neurons.
Collapse
Affiliation(s)
- Hao-Hua Wei
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Xiang-Shan Yuan
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Ze-Ka Chen
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Pei-Pei Chen
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zhe Xiang
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Rui-Xi Li
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Guo-Min Zhou
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
64
|
Hosono T, Ono M, Daikoku T, Mieda M, Nomura S, Kagami K, Iizuka T, Nakata R, Fujiwara T, Fujiwara H, Ando H. Time-Restricted Feeding Regulates Circadian Rhythm of Murine Uterine Clock. Curr Dev Nutr 2021; 5:nzab064. [PMID: 33981944 PMCID: PMC8099714 DOI: 10.1093/cdn/nzab064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/23/2021] [Accepted: 04/07/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Skipping breakfast is associated with dysmenorrhea in young women. This suggests that the delay of food intake in the active phase impairs uterine functions by interfering with circadian rhythms. OBJECTIVES To examine the relation between the delay of feeding and uterine circadian rhythms, we investigated the effects of the first meal occasion in the active phase on the uterine clock. METHODS Zeitgeber time (ZT) was defined as ZT0 (08:45) with lights on and ZT12 (20:45) with lights off. Young female mice (8 wk of age) were divided into 3 groups: group I (ad libitum consumption), group II (time-restricted feeding during ZT12-16, initial 4 h of the active period), and group III (time-restricted feeding during ZT20-24, last 4 h of the active period, a breakfast-skipping model). After 2 wk of dietary restriction, mice in each group were killed at 4-h intervals and the expression profiles of uterine clock genes, Bmal1 (brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1), Per1 (period circadian clock 1), Per2, and Cry1 (cryptochrome 1), were examined. RESULTS qPCR and western blot analyses demonstrated synchronized circadian clock gene expression within the uterus. Immunohistochemical analysis confirmed that BMAL1 protein expression was synchronized among the endometrium and myometrium. In groups I and II, mRNA expression of Bmal1 was elevated after ZT12 at the start of the active phase. In contrast, Bmal1 expression was elevated just after ZT20 in group III, showing that the uterine clock rhythm had shifted 8 h backward. The changes in BMAL1 protein expression were confirmed by western blot analysis. CONCLUSIONS This study is the first to indicate that time-restricted feeding regulates a circadian rhythm of the uterine clock that is synchronized throughout the uterine body. These findings suggest that the uterine clock system is a new candidate to explain the etiology of breakfast skipping-induced uterine dysfunction.
Collapse
Affiliation(s)
- Takashi Hosono
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Takiko Daikoku
- Institute for Experimental Animals, Advanced Science Research Center, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Satoshi Nomura
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Kyosuke Kagami
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Takashi Iizuka
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Rieko Nakata
- Department of Food Science and Nutrition, Nara Women's University, Nara, Japan
| | - Tomoko Fujiwara
- Department of Social Work and Life Design, Kyoto Notre Dame University, Kyoto, Japan
| | - Hiroshi Fujiwara
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Hitoshi Ando
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
65
|
Hagiwara D, Tochiya M, Azuma Y, Tsumura T, Hodai Y, Kawaguchi Y, Miyata T, Kobayashi T, Sugiyama M, Onoue T, Takagi H, Ito Y, Iwama S, Suga H, Banno R, Arima H. Arginine vasopressin-Venus reporter mice as a tool for studying magnocellular arginine vasopressin neurons. Peptides 2021; 139:170517. [PMID: 33647312 DOI: 10.1016/j.peptides.2021.170517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 11/20/2022]
Abstract
Arginine vasopressin (AVP) synthesized in the magnocellular neurons of the hypothalamus is transported through their axons and released from the posterior pituitary into the systemic circulation to act as an antidiuretic hormone. AVP synthesis and release are precisely regulated by changes in plasma osmolality. Magnocellular AVP neurons receive innervation from osmosensory and sodium-sensing neurons, but previous studies showed that AVP neurons per se are osmosensitive as well. In the current study, we made AVP-Venus reporter mice and showed that Venus was expressed exclusively in AVP neurons and was upregulated under water deprivation. In hypothalamic organotypic cultures from the AVP-Venus mice, Venus-labeled AVP neurons in the supraoptic and paraventricular nuclei survived for 1 month, and Venus expression was upregulated by forskolin. Furthermore, in dissociated Venus-labeled magnocellular neurons, treatment with NaCl, but not with mannitol, decreased Venus fluorescence in the soma of the AVP neurons. Thus, Venus expression in AVP-Venus transgenic mice, as well as in primary cultures, faithfully showed the properties of intrinsic AVP expression. These findings indicate that AVP-Venus mice as well as the primary hypothalamic cultures could be useful for studying magnocellular AVP neurons.
Collapse
Affiliation(s)
- Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| | - Masayoshi Tochiya
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Yoshinori Azuma
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Tetsuro Tsumura
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Yuichi Hodai
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Yohei Kawaguchi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Takashi Miyata
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Tomoko Kobayashi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Hiroshi Takagi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Yoshihiro Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan; Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya, 464-8601, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| |
Collapse
|
66
|
Nesan D, Feighan KM, Antle MC, Kurrasch DM. Gestational low-dose BPA exposure impacts suprachiasmatic nucleus neurogenesis and circadian activity with transgenerational effects. SCIENCE ADVANCES 2021; 7:eabd1159. [PMID: 34049886 PMCID: PMC8163075 DOI: 10.1126/sciadv.abd1159] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 04/09/2021] [Indexed: 05/30/2023]
Abstract
Critical physiological processes such as sleep and stress that underscore health are regulated by an intimate interplay between the endocrine and nervous systems. Here, we asked how fetal exposure to the endocrine disruptor found in common plastics, bisphenol A (BPA), causes lasting effects on adult animal behaviors. Adult mice exposed to low-dose BPA during gestation displayed notable disruption in circadian activity, social interactions, and associated neural hyperactivity, with some phenotypes maintained transgenerationally. Gestational BPA exposure increased vasopressin+ neurons in the suprachiasmatic nucleus (SCN), the region that regulates circadian rhythms, of F1 and F3 generations. Mechanistically, BPA increased proliferation of hypothalamic neural progenitors ex vivo and caused precocious neurogenesis in vivo. Co-antagonism of both estrogen and androgen receptors was necessary to block BPA's effects on hypothalamic neural progenitors, illustrating a dual role for these endocrine targets. Together, gestational BPA exposure affects development of circadian centers, with lasting consequences across generations.
Collapse
Affiliation(s)
- Dinushan Nesan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Kira M Feighan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Michael C Antle
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Psychology, Faculty of Arts, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
67
|
Arginine Vasopressin-Containing Neurons of the Suprachiasmatic Nucleus Project to CSF. eNeuro 2021; 8:ENEURO.0363-20.2021. [PMID: 33472866 PMCID: PMC8174031 DOI: 10.1523/eneuro.0363-20.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 02/01/2023] Open
Abstract
While it is well established that there are robust circadian rhythms of arginine vasopressin (AVP) in the cerebrospinal fluid (CSF), the route whereby the peptide reaches the CSF is not clear. A, AVP neurons constitute the largest fraction of the SCN neuronal population. Here, we show that processes of AVP-expressing SCN neurons cross the epithelium of the 3rd ventricular wall to reach the CSF (black arrows). Additionally, we report rostro-caudal differences in AVP neuron size and demonstrate that the localization of cells expressing the clock protein PER2 extend beyond the AVP population, thereby indicating that the size of this nucleus is somewhat larger than previously understood. B, Following lateral ventricle (LV) injection of cholera toxin β subunit (CTβ ; magenta) the retrograde tracer is seen in AVP neurons of the SCN, supporting the anatomical evidence that AVP neuronal processes directly contact the CSF. Arginine vasopressin (AVP) expressing neurons form the major population in the brain’s circadian clock located in the hypothalamic suprachiasmatic nucleus (SCN). They participate in inter-neuronal coupling and provide an output signal for synchronizing daily rhythms. AVP is present at high concentrations in the cerebrospinal fluid (CSF) and fluctuates on a circadian timescale. While it is assumed that rhythms in CSF AVP are of SCN origin, a route of communication between these compartments has not been delineated. Using immunochemistry (ICC) and cell filling techniques, we determine the morphology and location of AVP neurons in mouse and delineate their axonal and dendritic processes. Cholera toxin β subunit (CTβ) tracer injected into the lateral ventricle tests whether AVP neurons communicate with CSF. Most importantly, the results indicate that AVP neurons lie in close proximity to the third ventricle, and their processes cross the ventricular wall into the CSF. We also report that contrary to widely held assumptions, AVP neurons do not fully delineate the SCN borders as PER2 expression extends beyond the AVP region. Also, AVP neurons form a rostral prong originating in the SCN medial-most and ventral-most aspect. AVP is lacking in the mid-dorsal shell but does occur at the base of the SCN just above the optic tract. Finally, neurons of the rostral SCN are smaller than those lying caudally. These findings extend our understanding of AVP signaling potential, demonstrate the heterogeneity of AVP neurons, and highlight limits in using this peptide to delineate the mouse SCN.
Collapse
|
68
|
Ono D, Honma KI, Honma S. Roles of Neuropeptides, VIP and AVP, in the Mammalian Central Circadian Clock. Front Neurosci 2021; 15:650154. [PMID: 33935635 PMCID: PMC8081951 DOI: 10.3389/fnins.2021.650154] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
In mammals, the central circadian clock is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Individual SCN cells exhibit intrinsic oscillations, and their circadian period and robustness are different cell by cell in the absence of cellular coupling, indicating that cellular coupling is important for coherent circadian rhythms in the SCN. Several neuropeptides such as arginine vasopressin (AVP) and vasoactive intestinal polypeptide (VIP) are expressed in the SCN, where these neuropeptides function as synchronizers and are important for entrainment to environmental light and for determining the circadian period. These neuropeptides are also related to developmental changes of the circadian system of the SCN. Transcription factors are required for the formation of neuropeptide-related neuronal networks. Although VIP is critical for synchrony of circadian rhythms in the neonatal SCN, it is not required for synchrony in the embryonic SCN. During postnatal development, the clock genes cryptochrome (Cry)1 and Cry2 are involved in the maturation of cellular networks, and AVP is involved in SCN networks. This mini-review focuses on the functional roles of neuropeptides in the SCN based on recent findings in the literature.
Collapse
Affiliation(s)
- Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ken-Ichi Honma
- Research and Education Center for Brain Science, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Sato Honma
- Research and Education Center for Brain Science, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
69
|
Parnell AA, De Nobrega AK, Lyons LC. Translating around the clock: Multi-level regulation of post-transcriptional processes by the circadian clock. Cell Signal 2021; 80:109904. [PMID: 33370580 PMCID: PMC8054296 DOI: 10.1016/j.cellsig.2020.109904] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022]
Abstract
The endogenous circadian clock functions to maintain optimal physiological health through the tissue specific coordination of gene expression and synchronization between tissues of metabolic processes throughout the 24 hour day. Individuals face numerous challenges to circadian function on a daily basis resulting in significant incidences of circadian disorders in the United States and worldwide. Dysfunction of the circadian clock has been implicated in numerous diseases including cancer, diabetes, obesity, cardiovascular and hepatic abnormalities, mood disorders and neurodegenerative diseases. The circadian clock regulates molecular, metabolic and physiological processes through rhythmic gene expression via transcriptional and post-transcriptional processes. Mounting evidence indicates that post-transcriptional regulation by the circadian clock plays a crucial role in maintaining tissue specific biological rhythms. Circadian regulation affecting RNA stability and localization through RNA processing, mRNA degradation, and RNA availability for translation can result in rhythmic protein synthesis, even when the mRNA transcripts themselves do not exhibit rhythms in abundance. The circadian clock also targets the initiation and elongation steps of translation through multiple pathways. In this review, the influence of the circadian clock across the levels of post-transcriptional, translation, and post-translational modifications are examined using examples from humans to cyanobacteria demonstrating the phylogenetic conservation of circadian regulation. Lastly, we briefly discuss chronotherapies and pharmacological treatments that target circadian function. Understanding the complexity and levels through which the circadian clock regulates molecular and physiological processes is important for future advancement of therapeutic outcomes.
Collapse
Affiliation(s)
- Amber A Parnell
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Aliza K De Nobrega
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Lisa C Lyons
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA.
| |
Collapse
|
70
|
Mendoza J. Nighttime Light Hurts Mammalian Physiology: What Diurnal Rodent Models Are Telling Us. Clocks Sleep 2021; 3:236-250. [PMID: 33915800 PMCID: PMC8167723 DOI: 10.3390/clockssleep3020014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/16/2021] [Accepted: 03/15/2021] [Indexed: 01/24/2023] Open
Abstract
Natural sunlight permits organisms to synchronize their physiology to the external world. However, in current times, natural sunlight has been replaced by artificial light in both day and nighttime. While in the daytime, indoor artificial light is of lower intensity than natural sunlight, leading to a weak entrainment signal for our internal biological clock, at night the exposure to artificial light perturbs the body clock and sleep. Although electric light at night allows us "to live in darkness", our current lifestyle facilitates nighttime exposure to light by the use, or abuse, of electronic devices (e.g., smartphones). The chronic exposure to light at nighttime has been correlated to mood alterations, metabolic dysfunctions, and poor cognition. To decipher the brain mechanisms underlying these alterations, fundamental research has been conducted using animal models, principally of nocturnal nature (e.g., mice). Nevertheless, because of the diurnal nature of human physiology, it is also important to find and propose diurnal animal models for the study of the light effects in circadian biology. The present review provides an overview of the effects of light at nighttime on physiology and behavior in diurnal mammals, including humans. Knowing how the brain reacts to artificial light exposure, using diurnal rodent models, is fundamental for the development of new strategies in human health based in circadian biology.
Collapse
Affiliation(s)
- Jorge Mendoza
- Institute of Cellular and Integrative Neuroscience CNRS UPR3212, University of Strasburg, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| |
Collapse
|
71
|
Effect of acupuncture on the daytime function in patients with post-stroke sleep-wake disturbance: A randomized controlled trial. WORLD JOURNAL OF ACUPUNCTURE-MOXIBUSTION 2021. [DOI: 10.1016/j.wjam.2021.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
72
|
Cheng AH, Cheng HYM. Genesis of the Master Circadian Pacemaker in Mice. Front Neurosci 2021; 15:659974. [PMID: 33833665 PMCID: PMC8021851 DOI: 10.3389/fnins.2021.659974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is the central circadian clock of mammals. It is responsible for communicating temporal information to peripheral oscillators via humoral and endocrine signaling, ultimately controlling overt rhythms such as sleep-wake cycles, body temperature, and locomotor activity. Given the heterogeneity and complexity of the SCN, its genesis is tightly regulated by countless intrinsic and extrinsic factors. Here, we provide a brief overview of the development of the SCN, with special emphasis on the murine system.
Collapse
Affiliation(s)
- Arthur H. Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
73
|
Maywood ES, Chesham JE, Winsky-Sommerer R, Smyllie NJ, Hastings MH. Circadian Chimeric Mice Reveal an Interplay Between the Suprachiasmatic Nucleus and Local Brain Clocks in the Control of Sleep and Memory. Front Neurosci 2021; 15:639281. [PMID: 33679317 PMCID: PMC7935531 DOI: 10.3389/fnins.2021.639281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
Sleep is regulated by circadian and homeostatic processes. Whereas the suprachiasmatic nucleus (SCN) is viewed as the principal mediator of circadian control, the contributions of sub-ordinate local circadian clocks distributed across the brain are unknown. To test whether the SCN and local brain clocks interact to regulate sleep, we used intersectional genetics to create temporally chimeric CK1ε Tau mice, in which dopamine 1a receptor (Drd1a)-expressing cells, a powerful pacemaking sub-population of the SCN, had a cell-autonomous circadian period of 24 h whereas the rest of the SCN and the brain had intrinsic periods of 20 h. We compared these mice with non-chimeric 24 h wild-types (WT) and 20 h CK1ε Tau mutants. The periods of the SCN ex vivo and the in vivo circadian behavior of chimeric mice were 24 h, as with WT, whereas other tissues in the chimeras had ex vivo periods of 20 h, as did all tissues from Tau mice. Nevertheless, the chimeric SCN imposed its 24 h period on the circadian patterning of sleep. When compared to 24 h WT and 20 h Tau mice, however, the sleep/wake cycle of chimeric mice under free-running conditions was disrupted, with more fragmented sleep and an increased number of short NREMS and REMS episodes. Even though the chimeras could entrain to 20 h light:dark cycles, the onset of activity and wakefulness was delayed, suggesting that SCN Drd1a-Cre cells regulate the sleep/wake transition. Chimeric mice also displayed a blunted homeostatic response to 6 h sleep deprivation (SD) with an impaired ability to recover lost sleep. Furthermore, sleep-dependent memory was compromised in chimeras, which performed significantly worse than 24 h WT and 20 h Tau mice. These results demonstrate a central role for the circadian clocks of SCN Drd1a cells in circadian sleep regulation, but they also indicate a role for extra-SCN clocks. In circumstances where the SCN and sub-ordinate local clocks are temporally mis-aligned, the SCN can maintain overall circadian control, but sleep consolidation and recovery from SD are compromised. The importance of temporal alignment between SCN and extra-SCN clocks for maintaining vigilance state, restorative sleep and memory may have relevance to circadian misalignment in humans, with environmental (e.g., shift work) causes.
Collapse
Affiliation(s)
| | | | - Raphaelle Winsky-Sommerer
- Surrey Sleep Research Centre, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Nicola Jane Smyllie
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | |
Collapse
|
74
|
Maejima T, Tsuno Y, Miyazaki S, Tsuneoka Y, Hasegawa E, Islam MT, Enoki R, Nakamura TJ, Mieda M. GABA from vasopressin neurons regulates the time at which suprachiasmatic nucleus molecular clocks enable circadian behavior. Proc Natl Acad Sci U S A 2021; 118:e2010168118. [PMID: 33526663 PMCID: PMC8017960 DOI: 10.1073/pnas.2010168118] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The suprachiasmatic nucleus (SCN), the central circadian pacemaker in mammals, is a network structure composed of multiple types of γ-aminobutyric acid (GABA)-ergic neurons and glial cells. However, the roles of GABA-mediated signaling in the SCN network remain controversial. Here, we report noticeable impairment of the circadian rhythm in mice with a specific deletion of the vesicular GABA transporter in arginine vasopressin (AVP)-producing neurons. These mice showed disturbed diurnal rhythms of GABAA receptor-mediated synaptic transmission in SCN neurons and marked lengthening of the activity time in circadian behavioral rhythms due to the extended interval between morning and evening locomotor activities. Synchrony of molecular circadian oscillations among SCN neurons did not significantly change, whereas the phase relationships between SCN molecular clocks and circadian morning/evening locomotor activities were altered significantly, as revealed by PER2::LUC imaging of SCN explants and in vivo recording of intracellular Ca2+ in SCN AVP neurons. In contrast, daily neuronal activity in SCN neurons in vivo clearly showed a bimodal pattern that correlated with dissociated morning/evening locomotor activities. Therefore, GABAergic transmission from AVP neurons regulates the timing of SCN neuronal firing to temporally restrict circadian behavior to appropriate time windows in SCN molecular clocks.
Collapse
Affiliation(s)
- Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 920-8640 Ishikawa, Japan
| | - Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 920-8640 Ishikawa, Japan
| | - Shota Miyazaki
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, 214-8571 Kanagawa, Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Faculty of Medicine, Toho University, 143-8540 Tokyo, Japan
| | - Emi Hasegawa
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 920-8640 Ishikawa, Japan
| | - Md Tarikul Islam
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 920-8640 Ishikawa, Japan
| | - Ryosuke Enoki
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 444-8787 Okazaki, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 444-8787 Okazaki, Japan
| | - Takahiro J Nakamura
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, 214-8571 Kanagawa, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 920-8640 Ishikawa, Japan;
| |
Collapse
|
75
|
Lee SB, Park J, Kwak Y, Park YU, Nhung TTM, Suh BK, Woo Y, Suh Y, Cho E, Cho S, Park SK. Disrupted-in-schizophrenia 1 enhances the quality of circadian rhythm by stabilizing BMAL1. Transl Psychiatry 2021; 11:110. [PMID: 33542182 PMCID: PMC7862247 DOI: 10.1038/s41398-021-01212-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/15/2020] [Accepted: 01/07/2021] [Indexed: 11/27/2022] Open
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is a scaffold protein that has been implicated in multiple mental disorders. DISC1 is known to regulate neuronal proliferation, signaling, and intracellular calcium homeostasis, as well as neurodevelopment. Although DISC1 was linked to sleep-associated behaviors, whether DISC1 functions in the circadian rhythm has not been determined yet. In this work, we revealed that Disc1 expression exhibits daily oscillating pattern and is regulated by binding of circadian locomotor output cycles kaput (CLOCK) and Brain and muscle Arnt-like protein-1 (BMAL1) heterodimer to E-box sequences in its promoter. Interestingly, Disc1 deficiency increases the ubiquitination of BMAL1 and de-stabilizes it, thereby reducing its protein levels. DISC1 inhibits the activity of GSK3β, which promotes BMAL1 ubiquitination, suggesting that DISC1 regulates BMAL1 stability by inhibiting its ubiquitination. Moreover, Disc1-deficient cells and mice show reduced expression of other circadian genes. Finally, Disc1-LI (Disc1 knockout) mice exhibit damped circadian physiology and behaviors. Collectively, these findings demonstrate that the oscillation of DISC1 expression is under the control of CLOCK and BMAL1, and that DISC1 contributes to the core circadian system by regulating BMAL1 stability.
Collapse
Affiliation(s)
- Su Been Lee
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jihyun Park
- grid.289247.20000 0001 2171 7818Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Yongdo Kwak
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea ,grid.507563.2Present Address: SK biopharmaceuticals Ltd, Seongnam-Si, Republic of Korea
| | - Young-Un Park
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea ,grid.49606.3d0000 0001 1364 9317Present Address: Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| | - Truong Thi My Nhung
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Bo Kyoung Suh
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Youngsik Woo
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Yeongjun Suh
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Eunbyul Cho
- grid.49100.3c0000 0001 0742 4007Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Sehyung Cho
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea.
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
76
|
Hamnett R, Chesham JE, Maywood ES, Hastings MH. The Cell-Autonomous Clock of VIP Receptor VPAC2 Cells Regulates Period and Coherence of Circadian Behavior. J Neurosci 2021; 41:502-512. [PMID: 33234609 PMCID: PMC7821861 DOI: 10.1523/jneurosci.2015-20.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/28/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023] Open
Abstract
Circadian (approximately daily) rhythms pervade mammalian behavior. They are generated by cell-autonomous, transcriptional/translational feedback loops (TTFLs), active in all tissues. This distributed clock network is coordinated by the principal circadian pacemaker, the hypothalamic suprachiasmatic nucleus (SCN). Its robust and accurate time-keeping arises from circuit-level interactions that bind its individual cellular clocks into a coherent time-keeper. Cells that express the neuropeptide vasoactive intestinal peptide (VIP) mediate retinal entrainment of the SCN; and in the absence of VIP, or its cognate receptor VPAC2, circadian behavior is compromised because SCN cells cannot synchronize. The contributions to pace-making of other cell types, including VPAC2-expressing target cells of VIP, are, however, not understood. We therefore used intersectional genetics to manipulate the cell-autonomous TTFLs of VPAC2-expressing cells. Measuring circadian behavioral and SCN rhythmicity in these temporally chimeric male mice thus enabled us to determine the contribution of VPAC2-expressing cells (∼35% of SCN cells) to SCN time-keeping. Lengthening of the intrinsic TTFL period of VPAC2 cells by deletion of the CK1εTau allele concomitantly lengthened the period of circadian behavioral rhythms. It also increased the variability of the circadian period of bioluminescent TTFL rhythms in SCN slices recorded ex vivo Abrogation of circadian competence in VPAC2 cells by deletion of Bmal1 severely disrupted circadian behavioral rhythms and compromised TTFL time-keeping in the corresponding SCN slices. Thus, VPAC2-expressing cells are a distinct, functionally powerful subset of the SCN circuit, contributing to computation of ensemble period and maintenance of circadian robustness. These findings extend our understanding of SCN circuit topology.
Collapse
Affiliation(s)
- Ryan Hamnett
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH, United Kingdom
| | - Johanna E Chesham
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH, United Kingdom
| | - Elizabeth S Maywood
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH, United Kingdom
| | - Michael H Hastings
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH, United Kingdom
| |
Collapse
|
77
|
Modulation of single cell circadian response to NMDA by diacylglycerol lipase inhibition reveals a role of endocannabinoids in light entrainment of the suprachiasmatic nucleus. Neuropharmacology 2021; 185:108455. [PMID: 33444638 DOI: 10.1016/j.neuropharm.2021.108455] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/23/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022]
Abstract
Suprachiasmatic nucleus (SCN) of the hypothalamus is the master clock that drives circadian rhythms in physiology and behavior and adjusts their timing to external cues. Neurotransmitter glutamate and glutamatergic receptors sensitive to N-methyl-d-aspartate (NMDA) play a dual role in the SCN by coupling astrocytic and neuronal single cell oscillators and by resetting their phase in response to light. Recent reports suggested that signaling by endogenous cannabinoids (ECs) participates in both of these functions. We have previously shown that ECs, such as 2-arachidonoylglycerol (2-AG), act via CB1 receptors to affect the SCN response to light-mimicking NMDA stimulus in a time-dependent manner. We hypothesized that this ability is linked to the circadian regulation of EC signaling. We demonstrate that circadian clock in the rat SCN regulates expression of 2-AG transport, synthesis and degradation enzymes as well as its receptors. Inhibition of the major 2-AG synthesis enzyme, diacylglycerol lipase, enhanced the phase delay and lowered the amplitude of explanted SCN rhythm in response to NMDAR activation. Using microscopic PER2 bioluminescence imaging, we visualized how individual single cell oscillators in different parts of the SCN respond to the DAGL inhibition/NMDAR activation and shape response of the whole pacemaker. Additionally, we present strong evidence that the zero amplitude behavior of the SCN in response to single NMDA stimulus in the middle of subjective night is the result of a loss of rhythm in individual SCN cells. The paper provides new insights into the modulatory role of endocannabinoid signaling during the light entrainment of the SCN.
Collapse
|
78
|
Dannerfjord AA, Brown LA, Foster RG, Peirson SN. Light Input to the Mammalian Circadian Clock. Methods Mol Biol 2021; 2130:233-247. [PMID: 33284449 DOI: 10.1007/978-1-0716-0381-9_18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Circadian rhythms are 24-h cycles in physiology and behavior that occur in virtually all organisms. These processes are not simply driven by changes in the external environment as they persist under constant conditions, providing evidence for an internal biological clock. In mammals, this clock is located in the hypothalamic suprachiasmatic nuclei (SCN) and is based upon an intracellular mechanism composed of a transcriptional-translational feedback loop composed of a number of core clock genes. However, a clock is of no use unless it can be set to the correct time. The primary time cue for the molecular clock in the SCN is light detected by the eye. The photoreceptors involved in this process include the rods and cones that mediate vision, as well as the recently identified melanopsin-expressing photosensitive retinal ganglion cells (pRGCs). Light information is conveyed to the SCN via the retinohypothalamic tract, resulting in an intracellular signaling cascade which converges on cAMP-response elements in the promoters of several key clock genes. Over the last two decades a number of studies have investigated the transcriptional response of the SCN to light stimuli with the aim of further understanding these molecular signaling pathways. Here we provide an overview of these studies and provide protocols for studying the molecular responses to light in the SCN clock.
Collapse
Affiliation(s)
- Adam A Dannerfjord
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute (SCNi), Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, Oxford, UK
| | - Laurence A Brown
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Russell G Foster
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute (SCNi), Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, Oxford, UK
| | - Stuart N Peirson
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK. .,Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute (SCNi), Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, Oxford, UK.
| |
Collapse
|
79
|
Rohr KE, Telega A, Savaglio A, Evans JA. Vasopressin regulates daily rhythms and circadian clock circuits in a manner influenced by sex. Horm Behav 2021; 127:104888. [PMID: 33202247 PMCID: PMC7855892 DOI: 10.1016/j.yhbeh.2020.104888] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/30/2020] [Accepted: 11/04/2020] [Indexed: 12/17/2022]
Abstract
Arginine vasopressin (AVP) is a neurohormone that alters cellular physiology through both endocrine and synaptic signaling. Circadian rhythms in AVP release and other biological processes are driven by the suprachiasmatic nucleus (SCN) of the anterior hypothalamus. Loss of vasopressin signaling alters circadian behavior, but the basis of these effects remains unclear. Here we investigate the role of AVP signaling in circadian timekeeping by analyzing behavior and SCN function in a novel AVP-deficient mouse model. Consistent with previous work, loss of AVP signaling increases water consumption and accelerates recovery to simulated jetlag. We expand on these results to show that loss of AVP increases period, imprecision and plasticity of behavioral rhythms under constant darkness. Interestingly, the effect of AVP deficiency on circadian period was influenced by sex, with loss of AVP lengthening period in females but not males. Examining SCN function directly with ex vivo bioluminescence imaging of clock protein expression, we demonstrate that loss of AVP signaling modulates the period, precision, and phase relationships of SCN neurons in both sexes. This pattern of results suggests that there are likely sex differences in downstream targets of the SCN. Collectively, this work indicates that AVP signaling modulates circadian circuits in a manner influenced by sex, which provides new insight into sexual dimorphisms in the regulation of daily rhythms.
Collapse
Affiliation(s)
- Kayla E Rohr
- Marquette University, Department of Biomedical Sciences, United States of America
| | - Adam Telega
- Marquette University, Department of Biomedical Sciences, United States of America
| | - Alexandra Savaglio
- Marquette University, Department of Biomedical Sciences, United States of America
| | - Jennifer A Evans
- Marquette University, Department of Biomedical Sciences, United States of America.
| |
Collapse
|
80
|
Optogenetic Methods for the Study of Circadian Rhythms. Methods Mol Biol 2020. [PMID: 33284455 DOI: 10.1007/978-1-0716-0381-9_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
A fundamental feature of circadian clock neurons across species is that they express circadian rhythms in spontaneous spike frequency. Spike frequency rhythms serve as both output timing signals of clock neurons as well as resonant elements of rhythms generation. Importantly, optogenetics, as applied to clock neurons, can enable investigation of the roles of clock neuron electrical activity in circadian timing. Here we describe protocols for using both in vitro and in vivo optogenetics directed to mammalian clock neurons in the suprachiasmatic nucleus to study circadian physiology and behavior. Optogenetic stimulation via channelrhodopsin, or inhibition via halorhodopsin, allows for the precise manipulation of neuronal firing rates across the SCN, and within specific neuronal subpopulations thereof, and can be combined with actigraphy and gene expression analysis.
Collapse
|
81
|
Vasopressin in circadian function of SCN. J Biosci 2020. [DOI: 10.1007/s12038-020-00109-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
82
|
Shan Y, Abel JH, Li Y, Izumo M, Cox KH, Jeong B, Yoo SH, Olson DP, Doyle FJ, Takahashi JS. Dual-Color Single-Cell Imaging of the Suprachiasmatic Nucleus Reveals a Circadian Role in Network Synchrony. Neuron 2020; 108:164-179.e7. [PMID: 32768389 PMCID: PMC8265161 DOI: 10.1016/j.neuron.2020.07.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/17/2020] [Accepted: 07/10/2020] [Indexed: 01/08/2023]
Abstract
The suprachiasmatic nucleus (SCN) acts as a master pacemaker driving circadian behavior and physiology. Although the SCN is small, it is composed of many cell types, making it difficult to study the roles of particular cells. Here we develop bioluminescent circadian reporter mice that are Cre dependent, allowing the circadian properties of genetically defined populations of cells to be studied in real time. Using a Color-Switch PER2::LUCIFERASE reporter that switches from red PER2::LUCIFERASE to green PER2::LUCIFERASE upon Cre recombination, we assess circadian rhythms in two of the major classes of peptidergic neurons in the SCN: AVP (arginine vasopressin) and VIP (vasoactive intestinal polypeptide). Surprisingly, we find that circadian function in AVP neurons, not VIP neurons, is essential for autonomous network synchrony of the SCN and stability of circadian rhythmicity.
Collapse
Affiliation(s)
- Yongli Shan
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - John H Abel
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yan Li
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Mariko Izumo
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Kimberly H Cox
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Byeongha Jeong
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Seung-Hee Yoo
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - David P Olson
- Department of Pediatrics, Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francis J Doyle
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| |
Collapse
|
83
|
Suprachiasmatic VIP neurons are required for normal circadian rhythmicity and comprised of molecularly distinct subpopulations. Nat Commun 2020; 11:4410. [PMID: 32879310 PMCID: PMC7468160 DOI: 10.1038/s41467-020-17197-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 06/12/2020] [Indexed: 12/02/2022] Open
Abstract
The hypothalamic suprachiasmatic (SCN) clock contains several neurochemically defined cell groups that contribute to the genesis of circadian rhythms. Using cell-specific and genetically targeted approaches we have confirmed an indispensable role for vasoactive intestinal polypeptide-expressing SCN (SCNVIP) neurons, including their molecular clock, in generating the mammalian locomotor activity (LMA) circadian rhythm. Optogenetic-assisted circuit mapping revealed functional, di-synaptic connectivity between SCNVIP neurons and dorsomedial hypothalamic neurons, providing a circuit substrate by which SCNVIP neurons may regulate LMA rhythms. In vivo photometry revealed that while SCNVIP neurons are acutely responsive to light, their activity is otherwise behavioral state invariant. Single-nuclei RNA-sequencing revealed that SCNVIP neurons comprise two transcriptionally distinct subtypes, including putative pacemaker and non-pacemaker populations. Altogether, our work establishes necessity of SCNVIP neurons for the LMA circadian rhythm, elucidates organization of circadian outflow from and modulatory input to SCNVIP cells, and demonstrates a subpopulation-level molecular heterogeneity that suggests distinct functions for specific SCNVIP subtypes. Cell groups in the hypothalamic suprachiasmatic clock contribute to the genesis of circadian rhythms. The authors identified two populations of vasoactive intestinal polypeptide-expressing neurons in the suprachiasmatic nucleus which regulate locomotor circadian rhythm in mice.
Collapse
|
84
|
Goto Y, Nakamura TJ, Ogawa K, Hattori A, Tsujimoto M. Reciprocal Expression Patterns of Placental Leucine Aminopeptidase/Insulin-Regulated Aminopeptidase and Vasopressin in the Murine Brain. Front Mol Biosci 2020; 7:168. [PMID: 32793633 PMCID: PMC7393517 DOI: 10.3389/fmolb.2020.00168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/01/2020] [Indexed: 01/23/2023] Open
Abstract
Placental leucine aminopeptidase/insulin-regulated aminopeptidase (P-LAP/IRAP) regulates vasopressin and oxytocin levels in the brain and peripheral tissues by controlled degradation of these peptides. In this study, we determined the relationship between P-LAP/IRAP and vasopressin levels in subregions of the murine brain. P-LAP/IRAP expression was observed in almost all brain regions. The expression patterns of P-LAP/IRAP and vasopressin indicated that cells expressing one of these protein/peptide were distinct from those expressing the other, although there was significant overlap between the expression regions. In addition, we found reciprocal diurnal rhythm patterns in P-LAP/IRAP and arginine vasopressin (AVP) expression in the hippocampus and pituitary gland. Further, synchronously cultured PC12 cells on treatment with nerve growth factor (NGF) showed circadian expression patterns of P-LAP/IRAP and enzymatic activity during 24 h of incubation. Considering that vasopressin is one of the most efficient peptide substrates of P-LAP/IRAP, these results suggest a possible feedback loop between P-LAP/IRAP and vasopressin expression, that regulates the function of these substrate peptides of the enzyme via translocation of P-LAP/IRAP from intracellular vesicles to the plasma membrane in brain cells. These findings provide novel insights into the functions of P-LAP/IRAP in the brain and suggest the involvement of these peptides in modulation of brain AVP functions in hyperosmolality, memory, learning, and circadian rhythm.
Collapse
Affiliation(s)
- Yoshikuni Goto
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Nakano, Japan
| | - Takahiro J Nakamura
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Kenji Ogawa
- Laboratory of Veterinary Epizootiology, Department of Veterinary Medicine, Nihon University, Fujisawa, Japan
| | - Akira Hattori
- Department of System Chemotherapy and Molecular Sciences, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Masafumi Tsujimoto
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Nakano, Japan
| |
Collapse
|
85
|
Sun H, Li C, Zhang Y, Jiang M, Dong Q, Wang Z. Light-resetting impact on behavior and the central circadian clock in two vole species (genus: Lasiopodomys). Comp Biochem Physiol B Biochem Mol Biol 2020; 248-249:110478. [PMID: 32687979 DOI: 10.1016/j.cbpb.2020.110478] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/03/2020] [Accepted: 07/13/2020] [Indexed: 10/23/2022]
Abstract
The behavioral circadian rhythms of subterranean rodents show intra- and interspecies diversity in terms of adaptation to dark underground environments, but the endogenous molecular mechanism of rhythm regulation in the suprachiasmatic nuclei (SCN) is stable to many species. In this study, we sought to determine the rhythms of behavior and central molecular regulatory mechanisms in the SCN of the subterranean Mandarin voles (Lasiopodomys mandarinus) compared with a related aboveground species, Brandt's voles (Lasiopodomys brandtii). Both species were reared under a 12 L:12 D cycle or in continuous darkness for 4 weeks. The pattern of wheel-running activity was similar in both species and had a periodicity of almost 24 h regardless of rearing conditions. However, the intensity of daily activity in Brandt's voles decreased markedly in darkness, while there was no significant difference in activity intensity in mandarin voles under different light regimes. In both vole species, all tested genes in the SCN showed significant time-dependent expression regardless of rearing conditions, and the expression levels of most genes did not differ significantly between different species and conditions. However, the peak phase shift in gene expression differed between the two species. In conclusion, behavioral patterns in mandarin and Brandt's voles were regulated by a stable molecular endogenous biological clock. The observed differences in activity intensity and phase shift suggest that different mechanisms regulate circadian rhythms in different living environments.
Collapse
Affiliation(s)
- Hong Sun
- College of Physical Education (main Campus), Zhengzhou University, Zhengzhou, Henan Province, China; School of Life Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chuyi Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yifeng Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Mengwan Jiang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Qianqian Dong
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zhenlong Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan Province, China.
| |
Collapse
|
86
|
Time-Restricted G-Protein Signaling Pathways via GPR176, G z, and RGS16 Set the Pace of the Master Circadian Clock in the Suprachiasmatic Nucleus. Int J Mol Sci 2020; 21:ijms21145055. [PMID: 32709014 PMCID: PMC7404074 DOI: 10.3390/ijms21145055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 11/24/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) are an important source of drug targets with diverse therapeutic applications. However, there are still more than one hundred orphan GPCRs, whose ligands and functions remain unidentified. The suprachiasmatic nucleus (SCN) is the central circadian clock of the brain, directing daily rhythms in activity–rest behavior and physiology. Malfunction of the circadian clock has been linked to a wide variety of diseases, including sleep–wake disorders, obesity, diabetes, cancer, and hypertension, making the circadian clock an intriguing target for drug development. The orphan receptor GPR176 is an SCN-enriched orphan GPCR that sets the pace of the circadian clock. GPR176 undergoes asparagine (N)-linked glycosylation, a post-translational modification required for its proper cell-surface expression. Although its ligand remains unknown, this orphan receptor shows agonist-independent basal activity. GPR176 couples to the unique G-protein subclass Gz (or Gx) and participates in reducing cAMP production during the night. The regulator of G-protein signaling 16 (RGS16) is equally important for the regulation of circadian cAMP synthesis in the SCN. Genome-wide association studies, employing questionnaire-based evaluations of individual chronotypes, revealed loci near clock genes and in the regions containing RGS16 and ALG10B, a gene encoding an enzyme involved in protein N-glycosylation. Therefore, increasing evidence suggests that N-glycosylation of GPR176 and its downstream G-protein signal regulation may be involved in pathways characterizing human chronotypes. This review argues for the potential impact of focusing on GPCR signaling in the SCN for the purpose of fine-tuning the entire body clock.
Collapse
|
87
|
Breakfast Skipping in Female College Students Is a Potential and Preventable Predictor of Gynecologic Disorders at Health Service Centers. Diagnostics (Basel) 2020; 10:diagnostics10070476. [PMID: 32668795 PMCID: PMC7400274 DOI: 10.3390/diagnostics10070476] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 11/23/2022] Open
Abstract
Inadequate dietary habits in youth are known to increase the risk of onset of various diseases in adulthood. Previously, we found that female college students who skipped breakfast had higher incidences of dysmenorrhea, suggesting that breakfast skipping interferes with ovarian and uterine functions. Since dietary habits can be managed by education, it is preferable to establish a convenient screening system for meal skipping that is associated with dysmenorrhea as part of routine services of health service centers. In this study, we recruited 3172 female students aged from 18 to 25 at Kanazawa University and carried out an annual survey of the status of students’ health and lifestyle in 2019, by a questionnaire. We obtained complete responses from 3110 students and analyzed the relationship between dietary habits, such as meal skipping and history of dieting, and menstrual disorders, such as troubles or worries with menstruation, menstrual irregularity, menstrual pain, and use of oral contraceptives. The incidence of troubles or worries with menstruation was significantly higher in those with breakfast skipping (p < 0.05) and a history of dieting (p < 0.001). This survey successfully confirmed the positive relationship between breakfast skipping and menstrual pain (p < 0.001), indicating that this simple screening test is suitable for picking up breakfast skippers who are more prone to gynecologic disorders. In conclusions, since dysmenorrhea is one of the important clinical signs, breakfast skipping may become an effective marker to predict the subsequent onset of gynecological diseases at health service centers. Considering educational correction of meal skipping, breakfast skipping is a potential and preventable predictor that will contribute to managing menstrual disorders from a preventive standpoint in the future.
Collapse
|
88
|
Patton AP, Edwards MD, Smyllie NJ, Hamnett R, Chesham JE, Brancaccio M, Maywood ES, Hastings MH. The VIP-VPAC2 neuropeptidergic axis is a cellular pacemaking hub of the suprachiasmatic nucleus circadian circuit. Nat Commun 2020; 11:3394. [PMID: 32636383 PMCID: PMC7341843 DOI: 10.1038/s41467-020-17110-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 06/05/2020] [Indexed: 12/01/2022] Open
Abstract
The hypothalamic suprachiasmatic nuclei (SCN) are the principal mammalian circadian timekeeper, co-ordinating organism-wide daily and seasonal rhythms. To achieve this, cell-autonomous circadian timing by the ~20,000 SCN cells is welded into a tight circuit-wide ensemble oscillation. This creates essential, network-level emergent properties of precise, high-amplitude oscillation with tightly defined ensemble period and phase. Although synchronised, regional cell groups exhibit differentially phased activity, creating stereotypical spatiotemporal circadian waves of cellular activation across the circuit. The cellular circuit pacemaking components that generate these critical emergent properties are unknown. Using intersectional genetics and real-time imaging, we show that SCN cells expressing vasoactive intestinal polypeptide (VIP) or its cognate receptor, VPAC2, are neurochemically and electrophysiologically distinct, but together they control de novo rhythmicity, setting ensemble period and phase with circuit-level spatiotemporal complexity. The VIP/VPAC2 cellular axis is therefore a neurochemically and topologically specific pacemaker hub that determines the emergent properties of the SCN timekeeper. Circadian activity modulation in the suprachiasmatic nucleus (SCN) is a network-level emergent property that requires neuropeptide VIP signaling, yet the precise cellular mechanisms are unknown. Patton et al. show that cells expressing VIP or its receptor VPAC2 together determine these emergent properties of the SCN.
Collapse
Affiliation(s)
- Andrew P Patton
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Mathew D Edwards
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.,UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, London, UK
| | - Nicola J Smyllie
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Ryan Hamnett
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.,Department of Neurosurgery, Stanford University, Stanford, USA
| | - Johanna E Chesham
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Marco Brancaccio
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.,Department of Brain Sciences, UK Dementia Research Institute, Imperial College London, London, UK
| | - Elizabeth S Maywood
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Michael H Hastings
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.
| |
Collapse
|
89
|
Ono D, Honma KI, Honma S. GABAergic mechanisms in the suprachiasmatic nucleus that influence circadian rhythm. J Neurochem 2020; 157:31-41. [PMID: 32198942 DOI: 10.1111/jnc.15012] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 01/23/2023]
Abstract
The mammalian central circadian clock is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. The SCN contains multiple circadian oscillators which synchronize with each other via several neurotransmitters. Importantly, an inhibitory neurotransmitter, γ-amino butyric acid (GABA), is expressed in almost all SCN neurons. In this review, we discuss how GABA influences circadian rhythms in the SCN. Excitatory and inhibitory effects of GABA may depend on intracellular Cl- concentration, in which several factors such as day-length, time of day, development, and region in the SCN may be involved. GABA also mediates oscillatory coupling of the circadian rhythms in the SCN. Recent genetic approaches reveal that GABA refines circadian output rhythms, but not circadian oscillations in the SCN. Since several efferent projections of the SCN have been suggested, GABA might work downstream of neuronal pathways from the SCN which regulate the temporal order of physiology and behavior.
Collapse
Affiliation(s)
- Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ken-Ichi Honma
- Research and Education Center for Brain Science, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Sato Honma
- Research and Education Center for Brain Science, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
90
|
Mieda M. The central circadian clock of the suprachiasmatic nucleus as an ensemble of multiple oscillatory neurons. Neurosci Res 2020; 156:24-31. [DOI: 10.1016/j.neures.2019.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/09/2019] [Indexed: 10/26/2022]
|
91
|
Tackenberg MC, Hughey JJ, McMahon DG. Distinct Components of Photoperiodic Light Are Differentially Encoded by the Mammalian Circadian Clock. J Biol Rhythms 2020; 35:353-367. [PMID: 32527181 DOI: 10.1177/0748730420929217] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Seasonal light cycles influence multiple physiological functions and are mediated through photoperiodic encoding by the circadian system. Despite our knowledge of the strong connection between seasonal light input and downstream circadian changes, less is known about the specific components of seasonal light cycles that are encoded and induce persistent changes in the circadian system. Using combinations of 3 T cycles (23, 24, 26 h) and 2 photoperiods per T cycle (long and short, with duty cycles scaled to each T cycle), we investigate the after-effects of entrainment to these 6 light cycles. We measure locomotor behavior duration (α), period (τ), and entrained phase angle (ψ) in vivo and SCN phase distribution (σφ), τ, and ψ ex vivo to refine our understanding of critical light components for influencing particular circadian properties. We find that both photoperiod and T-cycle length drive determination of in vivo ψ but differentially influence after-effects in α and τ, with photoperiod driving changes in α and photoperiod length and T-cycle length combining to influence τ. Using skeleton photoperiods, we demonstrate that in vivo ψ is determined by both parametric and nonparametric components, while changes in α are driven nonparametrically. Within the ex vivo SCN, we find that ψ and σφ of the PER2∷LUCIFERASE rhythm follow closely with their likely behavioral counterparts (ψ and α of the locomotor activity rhythm) while also confirming previous reports of τ after-effects of gene expression rhythms showing negative correlations with behavioral τ after-effects in response to T cycles. We demonstrate that within-SCN σφ changes, thought to underlie α changes in vivo, are induced primarily nonparametrically. Taken together, our results demonstrate that distinct components of seasonal light input differentially influence ψ, α, and τ and suggest the possibility of separate mechanisms driving the persistent changes in circadian behaviors mediated by seasonal light.
Collapse
Affiliation(s)
| | - Jacob J Hughey
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee.,Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Douglas G McMahon
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee.,Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
92
|
Takahashi TM, Sunagawa GA, Soya S, Abe M, Sakurai K, Ishikawa K, Yanagisawa M, Hama H, Hasegawa E, Miyawaki A, Sakimura K, Takahashi M, Sakurai T. A discrete neuronal circuit induces a hibernation-like state in rodents. Nature 2020; 583:109-114. [PMID: 32528181 DOI: 10.1038/s41586-020-2163-6] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 03/04/2020] [Indexed: 02/07/2023]
Abstract
Hibernating mammals actively lower their body temperature to reduce energy expenditure when facing food scarcity1. This ability to induce a hypometabolic state has evoked great interest owing to its potential medical benefits2,3. Here we show that a hypothalamic neuronal circuit in rodents induces a long-lasting hypothermic and hypometabolic state similar to hibernation. In this state, although body temperature and levels of oxygen consumption are kept very low, the ability to regulate metabolism still remains functional, as in hibernation4. There was no obvious damage to tissues and organs or abnormalities in behaviour after recovery from this state. Our findings could enable the development of a method to induce a hibernation-like state, which would have potential applications in non-hibernating mammalian species including humans.
Collapse
Affiliation(s)
- Tohru M Takahashi
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Genshiro A Sunagawa
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.
| | - Shingo Soya
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan.,Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Katsuyasu Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Kiyomi Ishikawa
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Hiroshi Hama
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, Wako, Japan
| | - Emi Hasegawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Atsushi Miyawaki
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, Wako, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Takeshi Sakurai
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan. .,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan. .,Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
93
|
Adolescent Dietary Habit-induced Obstetric and Gynecologic Disease (ADHOGD) as a New Hypothesis-Possible Involvement of Clock System. Nutrients 2020; 12:nu12051294. [PMID: 32370105 PMCID: PMC7282263 DOI: 10.3390/nu12051294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
There are growing concerns that poor dietary behaviors at young ages will increase the future risk of chronic diseases in adulthood. We found that female college students who skipped breakfast had higher incidences of dysmenorrhea and irregular menstruation, suggesting that meal skipping affects ovarian and uterine functions. Since dysmenorrhea is more prevalent in those with a past history of dieting, we proposed a novel concept that inadequate dietary habits in adolescence become a trigger for the subsequent development of organic gynecologic diseases. Since inadequate feeding that was limited during the non-active phase impaired reproductive functions in post-adolescent female rats, we hypothesize that circadian rhythm disorders due to breakfast skipping disrupts the hypothalamic–pituitary–ovarian axis, impairs the reproductive rhythm, and leads to ovarian and uterine dysfunction. To explain how reproductive dysfunction is memorized from adolescence to adulthood, we hypothesize that the peripheral clock system also plays a critical role in the latent progression of reproductive diseases together with the central system, and propose naming this concept “adolescent dietary habit-induced obstetric and gynecologic disease (ADHOGD)”. This theory will contribute to analyzing the etiologies of and developing prophylaxes for female reproductive diseases from novel aspects. In this article, we describe the precise outline of the above hypotheses with the supporting evidence in the literature.
Collapse
|
94
|
Wen S, Ma D, Zhao M, Xie L, Wu Q, Gou L, Zhu C, Fan Y, Wang H, Yan J. Spatiotemporal single-cell analysis of gene expression in the mouse suprachiasmatic nucleus. Nat Neurosci 2020; 23:456-467. [DOI: 10.1038/s41593-020-0586-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 01/03/2020] [Indexed: 12/11/2022]
|
95
|
Hastings MH, Smyllie NJ, Patton AP. Molecular-genetic Manipulation of the Suprachiasmatic Nucleus Circadian Clock. J Mol Biol 2020; 432:3639-3660. [PMID: 31996314 DOI: 10.1016/j.jmb.2020.01.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/10/2020] [Accepted: 01/15/2020] [Indexed: 01/08/2023]
Abstract
Circadian (approximately daily) rhythms of physiology and behaviour adapt organisms to the alternating environments of day and night. The suprachiasmatic nucleus (SCN) of the hypothalamus is the principal circadian timekeeper of mammals. The mammalian cell-autonomous circadian clock is built around a self-sustaining transcriptional-translational negative feedback loop (TTFL) in which the negative regulators Per and Cry suppress their own expression, which is driven by the positive regulators Clock and Bmal1. Importantly, such TTFL-based clocks are present in all major tissues across the organism, and the SCN is their central co-ordinator. First, we analyse SCN timekeeping at the cell-autonomous and the circuit-based levels of organisation. We consider how molecular-genetic manipulations have been used to probe cell-autonomous timing in the SCN, identifying the integral components of the clock. Second, we consider new approaches that enable real-time monitoring of the activity of these clock components and clock-driven cellular outputs. Finally, we review how intersectional genetic manipulations of the cell-autonomous clockwork can be used to determine how SCN cells interact to generate an ensemble circadian signal. Critically, it is these network-level interactions that confer on the SCN its emergent properties of robustness, light-entrained phase and precision- properties that are essential for its role as the central co-ordinator. Remaining gaps in knowledge include an understanding of how the TTFL proteins behave individually and in complexes: whether particular SCN neuronal populations act as pacemakers, and if so, by which signalling mechanisms, and finally the nature of the recently discovered role of astrocytes within the SCN network.
Collapse
Affiliation(s)
- Michael H Hastings
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.
| | - Nicola J Smyllie
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Andrew P Patton
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| |
Collapse
|
96
|
Whylings J, Rigney N, Peters NV, de Vries GJ, Petrulis A. Sexually dimorphic role of BNST vasopressin cells in sickness and social behavior in male and female mice. Brain Behav Immun 2020; 83:68-77. [PMID: 31550501 PMCID: PMC6906230 DOI: 10.1016/j.bbi.2019.09.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 08/29/2019] [Accepted: 09/20/2019] [Indexed: 01/23/2023] Open
Abstract
Circumstantial evidence supports the hypothesis that the sexually dimorphic vasopressin (AVP) innervation of the brain tempers sickness behavior in males. Here we test this hypothesis directly, by comparing sickness behavior in animals with or without ablations of BNST AVP cells, a major source of sexually dimorphic AVP in the brain. We treated male and female AVP-iCre+ and AVP-iCre- mice that had been injected with viral Cre-dependent caspase-3 executioner construct into the BNST with lipopolysaccharide (LPS) or sterile saline, followed by behavioral analysis. In all groups, LPS treatment reliably reduced motor behavior, increased anxiety-related behavior, and reduced sucrose preference and consumption. Male mice, whose BNST AVP cells had been ablated (AVP-iCre+), displayed only minor reductions in LPS-induced sickness behavior, whereas their female counterparts displayed, if anything, an increase in sickness behaviors. All saline-treated mice with BNST AVP cell ablations consumed more sucrose than did control mice, and males, but not females, with BNST AVP cell ablations showed reduced preference for novel conspecifics compared to control mice. These data confirm that BNST AVP cells control social behavior in a sexually dimorphic way, but do not play a critical role in altering sickness behavior.
Collapse
Affiliation(s)
- Jack Whylings
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| | - Nicole Rigney
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| | - Nicole V Peters
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Geert J de Vries
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA; Department of Biology, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| | - Aras Petrulis
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| |
Collapse
|
97
|
De Nobrega AK, Luz KV, Lyons LC. Resetting the Aging Clock: Implications for Managing Age-Related Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1260:193-265. [PMID: 32304036 DOI: 10.1007/978-3-030-42667-5_9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Worldwide, individuals are living longer due to medical and scientific advances, increased availability of medical care and changes in public health policies. Consequently, increasing attention has been focused on managing chronic conditions and age-related diseases to ensure healthy aging. The endogenous circadian system regulates molecular, physiological and behavioral rhythms orchestrating functional coordination and processes across tissues and organs. Circadian disruption or desynchronization of circadian oscillators increases disease risk and appears to accelerate aging. Reciprocally, aging weakens circadian function aggravating age-related diseases and pathologies. In this review, we summarize the molecular composition and structural organization of the circadian system in mammals and humans, and evaluate the technological and societal factors contributing to the increasing incidence of circadian disorders. Furthermore, we discuss the adverse effects of circadian dysfunction on aging and longevity and the bidirectional interactions through which aging affects circadian function using examples from mammalian research models and humans. Additionally, we review promising methods for managing healthy aging through behavioral and pharmacological reinforcement of the circadian system. Understanding age-related changes in the circadian clock and minimizing circadian dysfunction may be crucial components to promote healthy aging.
Collapse
Affiliation(s)
- Aliza K De Nobrega
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Kristine V Luz
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Lisa C Lyons
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
98
|
Abstract
The circadian clock is an endogenous, time-tracking system that directs multiple metabolic and physiological functions required for homeostasis. The master or central clock located within the suprachiasmatic nucleus in the hypothalamus governs peripheral clocks present in all systemic tissues, contributing to their alignment and ultimately to temporal coordination of physiology. Accumulating evidence reveals the presence of additional clocks in the brain and suggests the possibility that circadian circuits may feed back to these from the periphery. Here, we highlight recent advances in the communications between clocks and discuss how they relate to circadian physiology and metabolism.
Collapse
Affiliation(s)
- Carolina Magdalen Greco
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, School of Medicine, University of California, Irvine, CA, USA
| | - Paolo Sassone-Corsi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
99
|
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: 0.7] [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
|
100
|
Abnormal Photic Entrainment to Phase-Delaying Stimuli in the R6/2 Mouse Model of Huntington's Disease, despite Retinal Responsiveness to Light. eNeuro 2019; 6:ENEURO.0088-19.2019. [PMID: 31744839 PMCID: PMC6905640 DOI: 10.1523/eneuro.0088-19.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 10/22/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022] Open
Abstract
The circadian clock located in the suprachiasmatic nucleus (SCN) in mammals entrains to ambient light via the retinal photoreceptors. This allows behavioral rhythms to change in synchrony with seasonal and daily changes in light period. Circadian rhythmicity is progressively disrupted in Huntington's disease (HD) and in HD mouse models such as the transgenic R6/2 line. Although retinal afferent inputs to the SCN are disrupted in R6/2 mice at late stages, they can respond to changes in light/dark cycles, as seen in jet lag and 23 h/d paradigms. To investigate photic entrainment and SCN function in R6/2 mice at different stages of disease, we first assessed the effect on locomotor activity of exposure to a 15 min light pulse given at different times of the day. We then placed the mice under five non-standard light conditions. These were light cycle regimes (T-cycles) of T21 (10.5 h light/dark), T22 (11 h light/dark), T26 (13 h light/dark), constant light, or constant dark. We found a progressive impairment in photic synchronization in R6/2 mice when the stimuli required the SCN to lengthen rhythms (phase-delaying light pulse, T26, or constant light), but normal synchronization to stimuli that required the SCN to shorten rhythms (phase-advancing light pulse and T22). Despite the behavioral abnormalities, we found that Per1 and c-fos gene expression remained photo-inducible in SCN of R6/2 mice. Both the endogenous drift of the R6/2 mouse SCN to shorter periods and its inability to adapt to phase-delaying changes will contribute to the HD circadian dysfunction.
Collapse
|