1
|
King MW, Jacob S, Sharma A, Lawrence JH, Weaver DR, Musiek ES. Circadian rhythms and the light-dark cycle interact to regulate amyloid plaque accumulation and tau phosphorylation in 5xFAD mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.645805. [PMID: 40236233 PMCID: PMC11996435 DOI: 10.1101/2025.03.31.645805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Background Circadian disruption has long been appreciated as a downstream consequence of Alzheimer's Disease in humans. However, an upstream role for behavioral circadian disruption in regulating AD pathology remains an open question. Methods To determine the role of the central circadian clock in the suprachiasmatic nucleus (SCN) in regulating amyloid pathology, we crossed the 5xFAD amyloid mouse model with mice harboring deletion of the critical clock gene Bmal1 in GABAergic neurons using VGAT-iCre, which is expressed in >95% of SCN cells. To examine the role the light-dark cycle in this process, we aged these mice in either regular 12:12 light-dark (LD) or constant darkness (DD) conditions. Transcriptional, behavioral, and physiological rhythms were examined in VGAT-iCre; 5xFAD; Bmal1 fl/fl (VGAT-BMAL1KO;5xFAD) mice under varying light conditions. Amyloid plaque deposition, peri-plaque tau phosphorylation, and other pathology was examined by immunohistochemistry, and transcriptomic changes were examined by high-throughput qPCR. Results VGAT-BMAL1KO;5xFAD mice showed loss of SCN BMAL1 expression and severe disruption of behavioral rhythms in both LD and DD, with loss of day-night rhythms in consolidated sleep and blunting of rhythmic clock gene expression in the brain. Surprisingly, VGAT-BMAL1KO;5xFAD mice kept under LD showed reduced total plaque accumulation and peri-plaque tau phosphorylation, compared to Cre-negative controls. These changes were gated by the light-dark cycle, as they were absent in VGAT-BMAL1KO;5xFAD mice kept in DD conditions. Total plaque accumulation was also reduced in control 5xFAD mice kept in DD as compared to LD, suggesting a general effect of light-dark cycle on amyloid aggregation. Expression of murine presenilin 1 (Psen1) -- which catalyzes the processing of sAPPβ into Aβ -- as well as APP cleavage to C-terminal fragments, were suppressed in VGAT-BMAL1KO;5xFAD under LD conditions. Conclusions These studies elucidated an interaction between the circadian clock in GABAergic neurons and the light-dark cycle in regulating amyloid pathology and suggest that decoupling the central clock form the light-dark cycle may reduce APP cleavage and plaque formation. These results call into question the proposed simple positive feedback loop between circadian rhythm disruption and Alzheimer's Disease pathology.
Collapse
|
2
|
Rivera Quiles C, Simmons SC, Dodson O, Alday M, Camacho Fontánez N, Caico S, Garrison A, Shafieichaharberoud F, Huang X, Hu Q, Heller EA, Mazei-Robison MS. Identification of a Novel Population of Neuromedin S Expressing Neurons in the Ventral Tegmental Area That Promote Morphine-Elicited Behavior. J Neurosci 2025; 45:e1662242025. [PMID: 39929726 PMCID: PMC11949474 DOI: 10.1523/jneurosci.1662-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 01/02/2025] [Accepted: 01/08/2025] [Indexed: 03/28/2025] Open
Abstract
Opioid use disorder constitutes a major health and economic burden, but our limited understanding of the underlying neurobiology impedes better interventions. Alteration in the activity and output of dopamine (DA) neurons in the ventral tegmental area (VTA) contributes to drug effects, but the mechanisms underlying these changes remain relatively unexplored. We used translating ribosome affinity purification (TRAP) and RNA sequencing to identify gene expression changes in mouse VTA DA neurons following chronic morphine exposure. We found that expression of the neuropeptide neuromedin S (NMS) is robustly increased in VTA DA neurons by morphine. Using an NMS-iCre driver line, we confirmed that a subset of VTA neurons express NMS and that chemogenetic modulation of VTA NMS neuron activity altered morphine responses in male and female mice. Specifically, VTA NMS neuronal activation promoted morphine locomotor activity while inhibition reduced morphine locomotor activity and conditioned place preference. Interestingly, these effects appear specific to morphine, as modulation of VTA NMS activity did not affect cocaine behaviors, consistent with our data that cocaine administration does not increase VTA Nms expression. Chemogenetic manipulation of VTA neurons that express glucagon-like peptide, a transcript also robustly increased in VTA DA neurons by morphine, does not alter morphine-elicited behavior, further highlighting the functional relevance of VTA NMS-expressing neurons. Together, our current data suggest that NMS-expressing neurons represent a novel subset of VTA neurons that may be functionally relevant for morphine responses and support the utility of cell-type-specific analyses like TRAP to identify neuronal adaptations underlying substance use disorder.
Collapse
Affiliation(s)
- Cristina Rivera Quiles
- Departments of Neuroscience Program, Michigan State University, East Lansing, Michigan 48824
| | - Sarah C Simmons
- Departments of Neuroscience Program, Michigan State University, East Lansing, Michigan 48824
| | - Olivia Dodson
- Departments of Neuroscience Program, Michigan State University, East Lansing, Michigan 48824
| | - Milagros Alday
- Departments of Neuroscience Program, Michigan State University, East Lansing, Michigan 48824
| | - Nicole Camacho Fontánez
- Departments of Neuroscience Program, Michigan State University, East Lansing, Michigan 48824
| | - Samantha Caico
- Departments of Neuroscience Program, Michigan State University, East Lansing, Michigan 48824
| | - Amber Garrison
- Departments of Neuroscience Program, Michigan State University, East Lansing, Michigan 48824
| | | | - Xuefei Huang
- Chemistry, Michigan State University, East Lansing, Michigan 48824
| | - Qiwen Hu
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Elizabeth A Heller
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Michelle S Mazei-Robison
- Departments of Neuroscience Program, Michigan State University, East Lansing, Michigan 48824
- Physiology, Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
3
|
Liu T, He W, Zhong Z, Lu C, Wu L, Wang Z, Smith WK, Shi Q, Long Q, Wang H. The circadian clock orchestrates spermatogonial differentiation and fertilization by regulating retinoic acid signaling in vertebrates. Natl Sci Rev 2025; 12:nwae456. [PMID: 40051524 PMCID: PMC11884735 DOI: 10.1093/nsr/nwae456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 10/05/2024] [Accepted: 11/13/2024] [Indexed: 03/09/2025] Open
Abstract
The circadian clock generates and maintains ∼24-hour oscillations in almost all organs. The testis, however, remains mysterious, without a clear understanding of its circadian functions. Our time-series transcriptome analysis reveals more than 1000 rhythmically expressed genes in the zebrafish and mouse testes, respectively. Canonical circadian clock genes are rhythmically expressed in Sertoli cells and regulate retinoic acid (RA) production, which is also evidenced by their co-expression with RA synthesis genes in single Sertoli cells. Genetic and pharmacological manipulations and temporal desynchronization revealed that the circadian clock-regulated RA signaling synchronizes spermatogonial differentiation via zbtb16a and promotes fertilization via izumo1 in zebrafish. Our findings indicate that the testicular circadian clock contributes to reproduction in a cell-specific manner through RA signaling, highlighting circadian roles in male fertility.
Collapse
Affiliation(s)
- Taole Liu
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Wei He
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Zhaomin Zhong
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Chenchen Lu
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Lianxin Wu
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Ziming Wang
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - William Kojo Smith
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Quan Shi
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Qiaoming Long
- Cam-Su Genomic Resource Center, Soochow University, Suzhou 215123, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| |
Collapse
|
4
|
Tofani GSS, Clarke G, Cryan JF. I "Gut" Rhythm: the microbiota as a modulator of the stress response and circadian rhythms. FEBS J 2025; 292:1454-1479. [PMID: 39841560 PMCID: PMC11927059 DOI: 10.1111/febs.17400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/20/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025]
Abstract
Modern habits are becoming more and more disruptive to health. As our days are often filled with circadian disruption and stress exposures, we need to understand how our responses to these external stimuli are shaped and how their mediators can be targeted to promote health. A growing body of research demonstrates the role of the gut microbiota in influencing brain function and behavior. The stress response and circadian rhythms, which are essential to maintaining appropriate responses to the environment, are known to be impacted by the gut microbiota. Gut microbes have been shown to alter the host's response to stress and modulate circadian rhythmicity. Although studies demonstrated strong links between the gut microbiota, circadian rhythms and the stress response, such studies were conducted in an independent manner not conducive to understanding the interface between these factors. Due to the interconnected nature of the stress response and circadian rhythms, in this review we explore how the gut microbiota may play a role in regulating the integration of stress and circadian signals in mammals and the consequences for brain health and disease.
Collapse
Affiliation(s)
- Gabriel S. S. Tofani
- APC MicrobiomeUniversity College CorkIreland
- Department of Anatomy & NeuroscienceUniversity College CorkIreland
| | - Gerard Clarke
- APC MicrobiomeUniversity College CorkIreland
- Department of Psychiatry & Neurobehavioural ScienceUniversity College CorkIreland
| | - John F. Cryan
- APC MicrobiomeUniversity College CorkIreland
- Department of Anatomy & NeuroscienceUniversity College CorkIreland
| |
Collapse
|
5
|
Wu YE, De Luca R, Broadhurst RY, Venner A, Sohn LT, Bandaru SS, Schwalbe DC, Campbell J, Arrigoni E, Fuller PM. Suprachiasmatic Neuromedin-S Neurons Regulate Arousal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.22.639648. [PMID: 40027719 PMCID: PMC11870627 DOI: 10.1101/2025.02.22.639648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Mammalian circadian rhythms, which orchestrate the daily temporal structure of biological processes, including the sleep-wake cycle, are primarily regulated by the circadian clock in the hypothalamic suprachiasmatic nucleus (SCN). The SCN clock is also implicated in providing an arousal 'signal,' particularly during the wake-maintenance zone (WMZ) of our biological day, essential for sustaining normal levels of wakefulness in the presence of mounting sleep pressure. Here we identify a role for SCN Neuromedin-S (SCN NMS ) neurons in regulating the level of arousal, especially during the WMZ. We used chemogenetic and optogenetic methods to activate SCN NMS neurons in vivo, which potently drove wakefulness. Fiber photometry confirmed the wake-active profile of SCN NM neurons. Genetically ablating SCN NMS neurons disrupted the sleep-wake cycle, reducing wakefulness during the dark period and abolished the circadian rhythm of body temperature. SCN NMS neurons target the dorsomedial hypothalamic nucleus (DMH), and photostimulation of their terminals within the DMH rapidly produces arousal from sleep. Pre-synaptic inputs to SCN NMS neurons were also identified, including regions known to influence SCN clock regulation. Unexpectedly, we discovered strong input from the preoptic area (POA), which itself receives substantial inhibitory input from the DMH, forming a possible arousal-promoting circuit (SCN->DMH->POA->SCN). Finally, we analyzed the transcriptional profile of SCN NMS neurons via single-nuclei RNA-Seq, revealing three distinct subtypes. Our findings link molecularly-defined SCN neurons to sleep-wake patterns, body temperature rhythms, and arousal control. Significance Statement Our study's findings provide a cellular and neurobiological understanding of how Neuromedin-S (NMS)-containing SCN neurons contribute to regulating circadian rhythms, sleep-wake patterns, body temperature, and arousal control in mammals. This research illuminates the circuit, cellular, and synaptic mechanisms through which SCN neurons regulate daily cycles of wakefulness and sleep, with implications for understanding and potentially manipulating these processes in health and disease.
Collapse
Affiliation(s)
- Yu-Er Wu
- Department of Neurological Surgery, University of California, Davis School of Medicine; Davis, CA 95618, USA
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA 02215, USA
| | - Roberto De Luca
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA 02215, USA
| | - Rebecca Y. Broadhurst
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA 02215, USA
| | - Anne Venner
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA 02215, USA
| | - Lauren T. Sohn
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA 02215, USA
| | - Sathyajit S. Bandaru
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA 02215, USA
| | - Dana C. Schwalbe
- Department of Biology, University of Virginia; Charlottesville, VA 22904, USA
| | - John Campbell
- Department of Biology, University of Virginia; Charlottesville, VA 22904, USA
| | - Elda Arrigoni
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA 02215, USA
| | - Patrick M Fuller
- Department of Neurological Surgery, University of California, Davis School of Medicine; Davis, CA 95618, USA
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA 02215, USA
| |
Collapse
|
6
|
Rahim AR, Will V, Myung J. Mood variation under dual regulation of circadian clock and light. Chronobiol Int 2025; 42:162-184. [PMID: 39840618 DOI: 10.1080/07420528.2025.2455144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/16/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025]
Abstract
The intricate relationship between circadian rhythms and mood is well-established. Disturbances in circadian rhythms and sleep often precede the development of mood disorders, such as major depressive disorder (MDD), bipolar disorder (BD), and seasonal affective disorder (SAD). Two primary factors, intrinsic circadian clocks and light, drive the natural fluctuations in mood throughout the day, mirroring the patterns of sleepiness and wakefulness. Nearly all organisms possess intrinsic circadian clocks that coordinate daily rhythms, with light serving as the primary environmental cue to synchronize these internal timekeepers with the 24-hour cycle. Additionally, light directly influences mood states. Disruptions to circadian rhythms, such as those caused by jet lag, shift work, or reduced daylight hours, can trigger or exacerbate mood symptoms. The complex and often subtle connections between circadian disruptions and mood dysregulation suggest that focusing solely on individual clock genes is insufficient to fully understand their etiology and progression. Instead, mood instability may arise from systemic misalignments between external cycles and the internal synchronization of circadian clocks. Here, we synthesize past research on the independent contributions of circadian clocks and light to mood regulation, drawing particularly on insights from animal studies that illuminate fundamental mechanisms relevant to human health.
Collapse
Affiliation(s)
- Amalia Ridla Rahim
- Laboratory of Braintime, Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, Taipei, Taiwan
| | - Veronica Will
- Laboratory of Braintime, Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, Taipei, Taiwan
| | - Jihwan Myung
- Laboratory of Braintime, Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences (GIMS), Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
7
|
Jászberényi M, Thurzó B, Jayakumar AR, Schally AV. The Aggravating Role of Failing Neuropeptide Networks in the Development of Sporadic Alzheimer's Disease. Int J Mol Sci 2024; 25:13086. [PMID: 39684795 DOI: 10.3390/ijms252313086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease imposes an increasing burden on aging Western societies. The disorder most frequently appears in its sporadic form, which can be caused by environmental and polygenic factors or monogenic conditions of incomplete penetrance. According to the authors, in the majority of cases, Alzheimer's disease represents an aggravated form of the natural aging of the central nervous system. It can be characterized by the decreased elimination of amyloid β1-42 and the concomitant accumulation of degradation-resistant amyloid plaques. In the present paper, the dysfunction of neuropeptide regulators, which contributes to the pathophysiologic acceleration of senile dementia, is reviewed. However, in the present review, exclusively those neuropeptides or neuropeptide families are scrutinized, and the authors' investigations into their physiologic and pathophysiologic activities have made significant contributions to the literature. Therefore, the pathophysiologic role of orexins, neuromedins, RFamides, corticotrope-releasing hormone family, growth hormone-releasing hormone, gonadotropin-releasing hormone, ghrelin, apelin, and natriuretic peptides are discussed in detail. Finally, the therapeutic potential of neuropeptide antagonists and agonists in the inhibition of disease progression is discussed here.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
| | - Balázs Thurzó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
- Emergency Patient Care Unit, Albert Szent-Györgyi Health Centre, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew V Schally
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
8
|
Klett N, Gompf HS, Allen CN, Cravetchi O, Hablitz LM, Gunesch AN, Irwin RP, Todd WD, Saper CB, Fuller PM. GABAergic signalling in the suprachiasmatic nucleus is required for coherent circadian rhythmicity. Eur J Neurosci 2024; 60:6652-6667. [PMID: 39558544 PMCID: PMC11612841 DOI: 10.1111/ejn.16582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 09/11/2024] [Accepted: 10/07/2024] [Indexed: 11/20/2024]
Abstract
The suprachiasmatic nucleus is the circadian pacemaker of the mammalian brain. Suprachiasmatic nucleus neurons display synchronization of their firing frequency on a circadian timescale, which is required for the pacemaker function of the suprachiasmatic nucleus. However, the mechanisms by which suprachiasmatic nucleus neurons remain synchronized in vivo are poorly understood, although synaptic communication is considered indispensable. Suprachiasmatic nucleus neurons contain the neurotransmitter GABA and express GABA receptors. This has inspired the hypothesis that GABA signalling may play a central role in network synchronization, although this remains untested in vivo. Here, using local genetic deletion, we show that disruption of GABA synaptic transmission within the suprachiasmatic nucleus of adult mice results in the eventual deterioration of physiological and behavioural rhythmicity in vivo and concomitant cellular desynchrony in vitro. These findings suggest that intercellular GABA signalling is essential for behavioural rhythmicity and cellular synchrony of the suprachiasmatic nucleus neural network.
Collapse
Affiliation(s)
- Nathan Klett
- Oregon Institute for Occupational Health SciencesUSA
- Neuroscience Graduate ProgramUSA
| | - Heinrich S. Gompf
- Department of Neurological SurgeryUniversity of California, DavisDavisCAUSA
| | - Charles N. Allen
- Oregon Institute for Occupational Health SciencesUSA
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandORUSA
| | | | - Lauren M. Hablitz
- Oregon Institute for Occupational Health SciencesUSA
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandORUSA
- Present address:
Center for Translational NeuromedicineUniversity of Rochester Medical CenterRochesterNYUSA
| | | | | | - William D. Todd
- Department of Neurology, Division of Sleep Medicine, and Program in NeuroscienceBeth Israel Deaconess Medical Center, Harvard Medical School BostonMAUSA
- Present address:
Department of Zoology and PhysiologyUniversity of WyomingLaramieWYUSA
| | - Clifford B. Saper
- Department of Neurology, Division of Sleep Medicine, and Program in NeuroscienceBeth Israel Deaconess Medical Center, Harvard Medical School BostonMAUSA
| | - Patrick M. Fuller
- Department of Neurological SurgeryUniversity of California, DavisDavisCAUSA
| |
Collapse
|
9
|
Duong TVQ, Yaw AM, Zhou G, Sina N, Cherukuri AS, Nguyen D, Cataldo K, Ly N, Sen A, Sempere L, Detrie C, Seiler R, Olomu IN, Cortese R, Long R, Hoffmann HM. Interaction between time-of-day and oxytocin efficacy in mice and humans with and without gestational diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.16.622460. [PMID: 39605626 PMCID: PMC11601330 DOI: 10.1101/2024.11.16.622460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Management of labor in women with diabetes is challenging due to the high risk of peri- and postpartum complications. To avoid cesarean section and assist with labor progression, Pitocin, a synthetic oxytocin, is frequently used to induce and augment labor. However, the efficacy of Pitocin is often compromised in diabetic pregnancies, leading to increased cesarian delivery. As diabetes deregulates the body's circadian timekeeping system and the time-of-day of the first Pitocin administration contributes to labor duration, our objective was to determine how the time of day and the circadian clock gene, Bmal1, gates oxytocin efficacy. Our studies in mice show that, compared to the rest phase of the day (lights on), the uterotonic efficacy of oxytocin is significantly increased during the active phase (lights off). Using in vitro studies, a myometrium-specific Bmal1 conditional knockout mouse model, and a mouse model of food-induced gestational diabetes, we find that Bmal1 is crucial for maintaining oxytocin receptor expression and response in the myometrium in mice. These findings also translate to humans, where oxytocin-induced human myometrial cell contraction is time-of-day dependent, and retrospective clinical data suggest that administration of Pitocin in the morning should be considered for pregnant women with gestational diabetes.
Collapse
Affiliation(s)
- Thu Van-Quynh Duong
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
- Department of Obstetrics and Gynecology, McLaren Greater Lansing Hospital, Lansing, MI, USA, 48910
| | - Alexandra M. Yaw
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Guoli Zhou
- Center for Statistical Training & Consulting (CSTAT), Michigan State University, East Lansing, MI, USA
| | - Niharika Sina
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Aneesh Sai Cherukuri
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Duong Nguyen
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Kylie Cataldo
- Department of Pediatrics; Department of Obstetrics, Gynecology, and Women’s Health. University of Missouri. Columbia, MO, 65212, USA
| | - Nicollette Ly
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Aritro Sen
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Lorenzo Sempere
- Precision Health Program; Department of Radiology, Michigan State University, East Lansing, MI, USA
| | - Cara Detrie
- Department of Obstetrics and Gynecology, McLaren Greater Lansing Hospital, Lansing, MI, USA, 48910
| | - Robert Seiler
- Department of Obstetrics and Gynecology, McLaren Greater Lansing Hospital, Lansing, MI, USA, 48910
| | - I. Nicholas Olomu
- Department of Pediatrics & Human Development, Division of Neonatal-Perinatal Medicine, Michigan State University, East Lansing MI, 48824
| | - Rene Cortese
- Department of Pediatrics; Department of Obstetrics, Gynecology, and Women’s Health. University of Missouri. Columbia, MO, 65212, USA
| | - Robert Long
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hanne M. Hoffmann
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| |
Collapse
|
10
|
Barsanele PS, de Assis LVM, da Silva JJ, Furtado EMDO, Fernandes P, Cipolla-Neto J, Poletini MO, Moraes MN. Glaucoma-inducing retinal ganglion cell degeneration alters diurnal rhythm of key molecular components of the central clock and locomotor activity in mice. FASEB J 2024; 38:e70109. [PMID: 39441606 DOI: 10.1096/fj.202401105r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/06/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024]
Abstract
Glaucoma is a chronic optic neuropathy characterized by the progressive degeneration of retinal ganglion cells (RGC). These cells play a crucial role in transmitting visual and non-visual information to brain regions, including the suprachiasmatic nucleus (SCN), responsible for synchronizing biological rhythms. To understand how glaucoma affects circadian rhythm synchronization, we investigated potential changes in the molecular clock machinery in the SCN. We found that the progressive increase in intraocular pressure (IOP) negatively correlated with spontaneous locomotor activity (SLA). Transcriptome analysis revealed significant alterations in the SCN of glaucomatous mice, including downregulation of genes associated with circadian rhythms. In fact, we showed a loss of diurnal oscillation in the expression of vasoactive intestinal peptide (Vip), its receptor (Vipr2), and period 1 (Per1) in the SCN of glaucomatous mice. These findings were supported by the 7-h phase shift in the peak expression of arginine vasopressin (Avp) in the SCN of mice with glaucoma. Despite maintaining a 24-h period under both light/dark (LD) and constant dark (DD) conditions, glaucomatous mice exhibited altered SLA rhythms, characterized by decreased amplitude. Taken altogether, our findings provide evidence of how glaucoma affects the regulation of the central circadian clock and its consequence on the regulation of circadian rhythms.
Collapse
Affiliation(s)
- Pietra Souza Barsanele
- Laboratório de Cronobiologia Molecular, Departamento de Ciências Biológicas, Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, Brazil
- Programa de Pós-graduação em Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Neurobiologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Juliano Jefferson da Silva
- Laboratório de Neurobiologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Eliz Maria de Oliveira Furtado
- Laboratório de Cronobiologia Molecular, Departamento de Ciências Biológicas, Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, Brazil
- Programa de Pós-graduação em Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Paola Fernandes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - José Cipolla-Neto
- Laboratório de Neurobiologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Maristela Oliveira Poletini
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Nathália Moraes
- Laboratório de Cronobiologia Molecular, Departamento de Ciências Biológicas, Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, Brazil
- Programa de Pós-graduação em Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Neurobiologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
11
|
McManus D, Patton AP, Smyllie NJ, Chin JW, Hastings MH. PERfect Day: reversible and dose-dependent control of circadian time-keeping in the mouse suprachiasmatic nucleus by translational switching of PERIOD2 protein expression. Eur J Neurosci 2024; 60:5537-5552. [PMID: 39300693 PMCID: PMC7617102 DOI: 10.1111/ejn.16537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024]
Abstract
The biological clock of the suprachiasmatic nucleus (SCN) orchestrates circadian (approximately daily) rhythms of behaviour and physiology that underpin health. SCN cell-autonomous time-keeping revolves around a transcriptional/translational feedback loop (TTFL) within which PERIOD (PER1,2) and CRYPTOCHROME (CRY1,2) proteins heterodimerise and suppress trans-activation of their encoding genes (Per1,2; Cry1,2). To explore its contribution to SCN time-keeping, we used adeno-associated virus-mediated translational switching to express PER2 (tsPER2) in organotypic SCN slices carrying bioluminescent TTFL circadian reporters. Translational switching requires provision of the non-canonical amino acid, alkyne lysine (AlkK), for protein expression. Correspondingly, AlkK, but not vehicle, induced constitutive expression of tsPER2 in SCN neurons and reversibly and dose-dependently suppressed pPer1-driven transcription in PER-deficient (Per1,2-null) SCN, illustrating the potency of PER2 in negative regulation within the TTFL. Constitutive expression of tsPER2, however, failed to initiate circadian oscillations in arrhythmic PER-deficient SCN. In rhythmic, PER-competent SCN, AlkK dose-dependently reduced the amplitude of PER2-reported oscillations as inhibition by tsPER2 progressively damped the TTFL. tsPER2 also dose-dependently lengthened the period of the SCN TTFL and neuronal calcium rhythms. Following wash-out of AlkK to remove tsPER2, the SCN regained TTFL amplitude and period. Furthermore, SCN retained their pre-washout phase: the removal of tsPER2 did not phase-shift the TTFL. Given that constitutive tsCRY1 can regulate TTFL amplitude and period, but also reset TTFL phase and initiate rhythms in CRY-deficient SCN, these results reveal overlapping and distinct properties of PER2 and CRY1 within the SCN, and emphasise the utility of translational switching to explore the functions of circadian proteins.
Collapse
Affiliation(s)
- David McManus
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Andrew P Patton
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Nicola J Smyllie
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Jason W Chin
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Michael H Hastings
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
12
|
Neitz AF, Carter BM, Ceriani MF, Ellisman MH, de la Iglesia HO. Suprachiasmatic nucleus VIPergic fibers show a circadian rhythm of expansion and retraction. Curr Biol 2024; 34:4056-4061.e2. [PMID: 39127047 PMCID: PMC11387125 DOI: 10.1016/j.cub.2024.07.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/19/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024]
Abstract
In animals, overt circadian rhythms of physiology and behavior are centrally regulated by a circadian clock located in specific brain regions. In the fruit fly Drosophila and in mammals, these clocks rely on single-cell oscillators, but critical for their function as central circadian pacemakers are network properties that change dynamically throughout the circadian cycle as well as in response to environmental stimuli.1,2,3 In the fly, this plasticity involves circadian rhythms of expansion and retraction of clock neuron fibers.4,5,6,7,8,9,10,11,12,13,14 Whether these drastic structural changes are a universal property of central neuronal pacemakers is unknown. To address this question, we studied neurons of the mouse suprachiasmatic nucleus (SCN) that express vasoactive intestinal polypeptide (VIP), which are critical for the SCN to function as a central circadian pacemaker. By targeting the expression of the fluorescent protein tdTomato to these neurons and using tissue clearing techniques to visualize all SCN VIPergic neurons and their fibers, we show that, similar to clock neurons in the fly, VIPergic fibers undergo a daily rhythm of expansion and retraction, with maximal branching during the day. This rhythm is circadian, as it persists under constant environmental conditions and is present in both males and females. We propose that circadian structural remodeling of clock neurons represents a key feature of central circadian pacemakers that is likely critical to regulate network properties, the response to environmental stimuli, and the regulation of circadian outputs.
Collapse
Affiliation(s)
- Alexandra F Neitz
- Department of Biology, University of Washington, Seattle, WA 98195-1800, USA; Molecular & Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA
| | - Bryn M Carter
- Department of Biology, University of Washington, Seattle, WA 98195-1800, USA
| | | | - Mark H Ellisman
- National Center for Molecular Imaging Research, Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0608, USA
| | - Horacio O de la Iglesia
- Department of Biology, University of Washington, Seattle, WA 98195-1800, USA; Molecular & Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
13
|
Curtis L, Piggins HD. Diverse genetic alteration dysregulates neuropeptide and intracellular signalling in the suprachiasmatic nuclei. Eur J Neurosci 2024; 60:3921-3945. [PMID: 38924215 DOI: 10.1111/ejn.16443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/12/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
In mammals, intrinsic 24 h or circadian rhythms are primarily generated by the suprachiasmatic nuclei (SCN). Rhythmic daily changes in the transcriptome and proteome of SCN cells are controlled by interlocking transcription-translation feedback loops (TTFLs) of core clock genes and their proteins. SCN cells function as autonomous circadian oscillators, which synchronize through intercellular neuropeptide signalling. Physiological and behavioural rhythms can be severely disrupted by genetic modification of a diverse range of genes and proteins in the SCN. With the advent of next generation sequencing, there is unprecedented information on the molecular profile of the SCN and how it is affected by genetically targeted alteration. However, whether the expression of some genes is more readily affected by genetic alteration of the SCN is unclear. Here, using publicly available datasets from recent RNA-seq assessments of the SCN from genetically altered and control mice, we evaluated whether there are commonalities in transcriptome dysregulation. This was completed for four different phases across the 24 h cycle and was augmented by Gene Ontology Molecular Function (GO:MF) and promoter analysis. Common differentially expressed genes (DEGs) and/or enriched GO:MF terms included signalling molecules, their receptors, and core clock components. Finally, examination of the JASPAR database indicated that E-box and CRE elements in the promoter regions of several commonly dysregulated genes. From this analysis, we identify differential expression of genes coding for molecules involved in SCN intra- and intercellular signalling as a potential cause of abnormal circadian rhythms.
Collapse
Affiliation(s)
- Lucy Curtis
- School of Biological Sciences, University of Bristol, Bristol, UK
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, UK
| | - Hugh D Piggins
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
14
|
Yamaguchi Y. Arginine vasopressin: Critical regulator of circadian homeostasis. Peptides 2024; 177:171229. [PMID: 38663583 DOI: 10.1016/j.peptides.2024.171229] [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: 02/29/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 04/30/2024]
Abstract
Circadian rhythms optimally regulate numerous physiological processes in an organism and synchronize them with the external environment. The suprachiasmatic nucleus (SCN), the center of the circadian clock in mammals, is composed of multiple cell types that form a network that provides the basis for the remarkable stability of the circadian clock. Among the neuropeptides expressed in the SCN, arginine vasopressin (AVP) has attracted much attention because of its deep involvement in the function of circadian rhythms, as elucidated in particular by studies using genetically engineered mice. This review briefly summarizes the current knowledge on the peptidergic distribution and topographic neuronal organization in the SCN, the molecular mechanisms of the clock genes, and the relationship between the SCN and peripheral clocks. With respect to the physiological roles of AVP and AVP-expressing neurons, in addition to a sex-dependent action of AVP in the SCN, studies using AVP receptor knockout mice and mice genetically manipulated to alter the clock properties of AVP neurons are summarized here, highlighting its importance in maintaining circadian homeostasis and its potential as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Yoshiaki Yamaguchi
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Japan.
| |
Collapse
|
15
|
Bonnefont X. Cell Signaling in the Circadian Pacemaker: New Insights from in vivo Imaging. Neuroendocrinology 2024; 115:103-110. [PMID: 38754404 DOI: 10.1159/000539344] [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: 03/29/2024] [Accepted: 05/12/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND "One for all, and all for one," the famous rallying cry of the Three Musketeers, in Alexandre Dumas's popular novel, certainly applies to the 20,000 cells composing the suprachiasmatic nuclei (SCN). These cells work together to form the central clock that coordinates body rhythms in tune with the day-night cycle. Like virtually every body cell, individual SCN cells exhibit autonomous circadian oscillations, but this rhythmicity only reaches a high level of precision and robustness when the cells are coupled with their neighbors. Therefore, understanding the functional network organization of SCN cells beyond their core rhythmicity is an important issue in circadian biology. SUMMARY The present review summarizes the main results from our recent study demonstrating the feasibility of recording SCN cells in freely moving mice and the significance of variations in intracellular calcium over several timescales. KEY MESSAGE We discuss how in vivo imaging at the cell level will be pivotal to interrogate the mammalian master clock, in an integrated context that preserves the SCN network organization, with intact inputs and outputs.
Collapse
Affiliation(s)
- Xavier Bonnefont
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- BioCampus Montpellier, Université de Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
16
|
Bass J. Interorgan rhythmicity as a feature of healthful metabolism. Cell Metab 2024; 36:655-669. [PMID: 38335957 PMCID: PMC10990795 DOI: 10.1016/j.cmet.2024.01.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024]
Abstract
The finding that animals with circadian gene mutations exhibit diet-induced obesity and metabolic syndrome with hypoinsulinemia revealed a distinct role for the clock in the brain and peripheral tissues. Obesogenic diets disrupt rhythmic sleep/wake patterns, feeding behavior, and transcriptional networks, showing that metabolic signals reciprocally control the clock. Providing access to high-fat diet only during the sleep phase (light period) in mice accelerates weight gain, whereas isocaloric time-restricted feeding during the active period enhances energy expenditure due to circadian induction of adipose thermogenesis. This perspective focuses on advances and unanswered questions in understanding the interorgan circadian control of healthful metabolism.
Collapse
Affiliation(s)
- Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
17
|
Ono D, Weaver DR, Hastings MH, Honma KI, Honma S, Silver R. The Suprachiasmatic Nucleus at 50: Looking Back, Then Looking Forward. J Biol Rhythms 2024; 39:135-165. [PMID: 38366616 PMCID: PMC7615910 DOI: 10.1177/07487304231225706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
It has been 50 years since the suprachiasmatic nucleus (SCN) was first identified as the central circadian clock and 25 years since the last overview of developments in the field was published in the Journal of Biological Rhythms. Here, we explore new mechanisms and concepts that have emerged in the subsequent 25 years. Since 1997, methodological developments, such as luminescent and fluorescent reporter techniques, have revealed intricate relationships between cellular and network-level mechanisms. In particular, specific neuropeptides such as arginine vasopressin, vasoactive intestinal peptide, and gastrin-releasing peptide have been identified as key players in the synchronization of cellular circadian rhythms within the SCN. The discovery of multiple oscillators governing behavioral and physiological rhythms has significantly advanced our understanding of the circadian clock. The interaction between neurons and glial cells has been found to play a crucial role in regulating these circadian rhythms within the SCN. Furthermore, the properties of the SCN network vary across ontogenetic stages. The application of cell type-specific genetic manipulations has revealed components of the functional input-output system of the SCN and their correlation with physiological functions. This review concludes with the high-risk effort of identifying open questions and challenges that lie ahead.
Collapse
Affiliation(s)
- Daisuke Ono
- Stress Recognition and Response, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - David R Weaver
- Department of Neurobiology and NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Michael H Hastings
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Ken-Ichi Honma
- Research and Education Center for Brain Science, Hokkaido University, Sapporo, Japan
- Center for Sleep and Circadian Rhythm Disorders, Sapporo Hanazono Hospital, Sapporo, Japan
| | - Sato Honma
- Research and Education Center for Brain Science, Hokkaido University, Sapporo, Japan
- Center for Sleep and Circadian Rhythm Disorders, Sapporo Hanazono Hospital, Sapporo, Japan
| | - Rae Silver
- Stress Recognition and Response, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Neuroscience & Behavior, Barnard College and Department of Psychology, Columbia University, New York City, New York, USA
| |
Collapse
|
18
|
Liao M, Gao X, Chen C, Li Q, Guo Q, Huang H, Zhang E, Ju D. Integrated neural tracing and in-situ barcoded sequencing reveals the logic of SCN efferent circuits in regulating circadian behaviors. SCIENCE CHINA. LIFE SCIENCES 2024; 67:518-528. [PMID: 38057622 DOI: 10.1007/s11427-023-2420-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/30/2023] [Indexed: 12/08/2023]
Abstract
The circadian clock coordinates rhythms in numerous physiological processes to maintain organismal homeostasis. Since the suprachiasmatic nucleus (SCN) is widely accepted as the circadian pacemaker, it is critical to understand the neural mechanisms by which rhythmic information is transferred from the SCN to peripheral clocks. Here, we present the first comprehensive map of SCN efferent connections and suggest a molecular logic underlying these projections. The SCN projects broadly to most major regions of the brain, rather than solely to the hypothalamus and thalamus. The efferent projections from different subtypes of SCN neurons vary in distance and intensity, and blocking synaptic transmission of these circuits affects circadian rhythms in locomotion and feeding to different extents. We also developed a barcoding system to integrate retrograde tracing with in-situ sequencing, allowing us to link circuit anatomy and spatial patterns of gene expression. Analyses using this system revealed that brain regions functioning downstream of the SCN receive input from multiple neuropeptidergic cell types within the SCN, and that individual SCN neurons generally project to a single downstream brain region. This map of SCN efferent connections provides a critical foundation for future investigations into the neural circuits underlying SCN-mediated rhythms in physiology. Further, our new barcoded tracing method provides a tool for revealing the molecular logic of neuronal circuits within heterogeneous brain regions.
Collapse
Affiliation(s)
- Meimei Liao
- College of Biological Sciences, China Agriculture University, Beijing, 100193, China
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Xinwei Gao
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Chen Chen
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Qi Li
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- Tsinghua Institute of Multidisciplinar^ Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Qingchun Guo
- Chinese Institute for Brain Research, Beijing, 102206, China
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China
| | - He Huang
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 401336, China
| | - Erquan Zhang
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- Tsinghua Institute of Multidisciplinar^ Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Dapeng Ju
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 401336, China.
| |
Collapse
|
19
|
Bussi IL, Neitz AF, Sanchez REA, Casiraghi LP, Moldavan M, Kunda D, Allen CN, Evans JA, de la Iglesia HO. Expression of the vesicular GABA transporter within neuromedin S + neurons sustains behavioral circadian rhythms. Proc Natl Acad Sci U S A 2023; 120:e2314857120. [PMID: 38019855 PMCID: PMC10710084 DOI: 10.1073/pnas.2314857120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is the site of a central circadian clock that orchestrates overt rhythms of physiology and behavior. Circadian timekeeping requires intercellular communication among SCN neurons, and multiple signaling pathways contribute to SCN network coupling. Gamma-aminobutyric acid (GABA) is produced by virtually all SCN neurons, and previous work demonstrates that this transmitter regulates coupling in the adult SCN but is not essential for the nucleus to sustain overt circadian rhythms. Here, we show that the deletion of the gene that codes for the GABA vesicular transporter Vgat from neuromedin-S (NMS)+ neurons-a subset of neurons critical for SCN function-causes arrhythmia of locomotor activity and sleep. Further, NMS-Vgat deletion impairs intrinsic clock gene rhythms in SCN explants cultured ex vivo. Although vasoactive intestinal polypeptide (VIP) is critical for SCN function, Vgat deletion from VIP-expressing neurons did not lead to circadian arrhythmia in locomotor activity rhythms. Likewise, adult SCN-specific deletion of Vgat led to mild impairment of behavioral rhythms. Our results suggest that while the removal of GABA release from the adult SCN does not affect the pacemaker's ability to sustain overt circadian rhythms, its removal from a critical subset of neurons within the SCN throughout development removes the nucleus ability to sustain circadian rhythms. Our findings support a model in which SCN GABA release is critical for the developmental establishment of intercellular network properties that define the SCN as a central pacemaker.
Collapse
Affiliation(s)
- Ivana L. Bussi
- Department of Biology, University of Washington, Seattle, WA98195-1800
| | - Alexandra F. Neitz
- Department of Biology, University of Washington, Seattle, WA98195-1800
- Molecular and Cellular Biology in Seattle, University of Washington and Fred Hutch, Seattle, WA98195-7275
| | - Raymond E. A. Sanchez
- Department of Biology, University of Washington, Seattle, WA98195-1800
- Graduate Program in Neuroscience, University of Washington, Seattle, WA98195
| | | | - Michael Moldavan
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR97239
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR97239
| | - Divya Kunda
- Department of Biology, University of Washington, Seattle, WA98195-1800
| | - Charles N. Allen
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR97239
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR97239
| | - Jennifer A. Evans
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Horacio O. de la Iglesia
- Department of Biology, University of Washington, Seattle, WA98195-1800
- Molecular and Cellular Biology in Seattle, University of Washington and Fred Hutch, Seattle, WA98195-7275
- Graduate Program in Neuroscience, University of Washington, Seattle, WA98195
| |
Collapse
|
20
|
Onodera K, Tsuno Y, Hiraoka Y, Tanaka K, Maejima T, Mieda M. In vivo recording of the circadian calcium rhythm in Prokineticin 2 neurons of the suprachiasmatic nucleus. Sci Rep 2023; 13:16974. [PMID: 37813987 PMCID: PMC10562406 DOI: 10.1038/s41598-023-44282-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/05/2023] [Indexed: 10/11/2023] Open
Abstract
Prokineticin 2 (Prok2) is a small protein expressed in a subpopulation of neurons in the suprachiasmatic nucleus (SCN), the primary circadian pacemaker in mammals. Prok2 has been implicated as a candidate output molecule from the SCN to control multiple circadian rhythms. Genetic manipulation specific to Prok2-producing neurons would be a powerful approach to understanding their function. Here, we report the generation of Prok2-tTA knock-in mice expressing the tetracycline transactivator (tTA) specifically in Prok2 neurons and an application of these mice to in vivo recording of Ca2+ rhythms in these neurons. First, the specific and efficient expression of tTA in Prok2 neurons was verified by crossing the mice with EGFP reporter mice. Prok2-tTA mice were then used to express a fluorescent Ca2+ sensor protein to record the circadian Ca2+ rhythm in SCN Prok2 neurons in vivo. Ca2+ in these cells showed clear circadian rhythms in both light-dark and constant dark conditions, with their peaks around midday. Notably, the hours of high Ca2+ nearly coincided with the rest period of the behavioral rhythm. These observations fit well with the predicted function of Prok2 neurons as a candidate output pathway of the SCN by suppressing locomotor activity during both daytime and subjective daytime.
Collapse
Affiliation(s)
- Kaito Onodera
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yuichi Hiraoka
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan.
| |
Collapse
|
21
|
Davidson AJ, Beckner D, Bonnefont X. A Journey in the Brain's Clock: In Vivo Veritas? BIOLOGY 2023; 12:1136. [PMID: 37627020 PMCID: PMC10452196 DOI: 10.3390/biology12081136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023]
Abstract
The suprachiasmatic nuclei (SCN) of the hypothalamus contain the circadian pacemaker that coordinates mammalian rhythms in tune with the day-night cycle. Understanding the determinants of the intrinsic rhythmicity of this biological clock, its outputs, and resetting by environmental cues, has been a longstanding goal of the field. Integrated techniques of neurophysiology, including lesion studies and in vivo multi-unit electrophysiology, have been key to characterizing the rhythmic nature and outputs of the SCN in animal models. In parallel, reduced ex vivo and in vitro approaches have permitted us to unravel molecular, cellular, and multicellular mechanisms underlying the pacemaker properties of the SCN. New questions have emerged in recent years that will require combining investigation at a cell resolution within the physiological context of the living animal: What is the role of specific cell subpopulations in the SCN neural network? How do they integrate various external and internal inputs? What are the circuits involved in controlling other body rhythms? Here, we review what we have already learned about the SCN from in vivo studies, and how the recent development of new genetically encoded tools and cutting-edge imaging technology in neuroscience offers chronobiologists the opportunity to meet these challenges.
Collapse
Affiliation(s)
- Alec J. Davidson
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Delaney Beckner
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Xavier Bonnefont
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
| |
Collapse
|
22
|
Tsuno Y, Peng Y, Horike SI, Wang M, Matsui A, Yamagata K, Sugiyama M, Nakamura TJ, Daikoku T, Maejima T, Mieda M. In vivo recording of suprachiasmatic nucleus dynamics reveals a dominant role of arginine vasopressin neurons in circadian pacesetting. PLoS Biol 2023; 21:e3002281. [PMID: 37643163 PMCID: PMC10465001 DOI: 10.1371/journal.pbio.3002281] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 07/28/2023] [Indexed: 08/31/2023] Open
Abstract
The central circadian clock of the suprachiasmatic nucleus (SCN) is a network consisting of various types of neurons and glial cells. Individual cells have the autonomous molecular machinery of a cellular clock, but their intrinsic periods vary considerably. Here, we show that arginine vasopressin (AVP) neurons set the ensemble period of the SCN network in vivo to control the circadian behavior rhythm. Artificial lengthening of cellular periods by deleting casein kinase 1 delta (CK1δ) in the whole SCN lengthened the free-running period of behavior rhythm to an extent similar to CK1δ deletion specific to AVP neurons. However, in SCN slices, PER2::LUC reporter rhythms of these mice only partially and transiently recapitulated the period lengthening, showing a dissociation between the SCN shell and core with a period instability in the shell. In contrast, in vivo calcium rhythms of both AVP and vasoactive intestinal peptide (VIP) neurons in the SCN of freely moving mice demonstrated stably lengthened periods similar to the behavioral rhythm upon AVP neuron-specific CK1δ deletion, without changing the phase relationships between each other. Furthermore, optogenetic activation of AVP neurons acutely induced calcium increase in VIP neurons in vivo. These results indicate that AVP neurons regulate other SCN neurons, such as VIP neurons, in vivo and thus act as a primary determinant of the SCN ensemble period.
Collapse
Affiliation(s)
- Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yubo Peng
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shin-ichi Horike
- Division of Integrated Omics Research, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Mohan Wang
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Ayako Matsui
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kanato Yamagata
- Child Brain Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Mizuki Sugiyama
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Takahiro J. Nakamura
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
23
|
Yang ND, Mellor RL, Hermanstyne TO, Nerbonne JM. Effects of NALCN-Encoded Na + Leak Currents on the Repetitive Firing Properties of SCN Neurons Depend on K +-Driven Rhythmic Changes in Input Resistance. J Neurosci 2023; 43:5132-5141. [PMID: 37339878 PMCID: PMC10342223 DOI: 10.1523/jneurosci.0182-23.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/02/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
Neurons in the suprachiasmatic nucleus (SCN) generate circadian changes in the rates of spontaneous action potential firing that regulate and synchronize daily rhythms in physiology and behavior. Considerable evidence suggests that daily rhythms in the repetitive firing rates (higher during the day than at night) of SCN neurons are mediated by changes in subthreshold potassium (K+) conductance(s). An alternative "bicycle" model for circadian regulation of membrane excitability in clock neurons, however, suggests that an increase in NALCN-encoded sodium (Na+) leak conductance underlies daytime increases in firing rates. The experiments reported here explored the role of Na+ leak currents in regulating daytime and nighttime repetitive firing rates in identified adult male and female mouse SCN neurons: vasoactive intestinal peptide-expressing (VIP+), neuromedin S-expressing (NMS+) and gastrin-releasing peptide-expressing (GRP+) cells. Whole-cell recordings from VIP+, NMS+, and GRP+ neurons in acute SCN slices revealed that Na+ leak current amplitudes/densities are similar during the day and at night, but have a larger impact on membrane potentials in daytime neurons. Additional experiments, using an in vivo conditional knockout approach, demonstrated that NALCN-encoded Na+ currents selectively regulate daytime repetitive firing rates of adult SCN neurons. Dynamic clamp-mediated manipulation revealed that the effects of NALCN-encoded Na+ currents on the repetitive firing rates of SCN neurons depend on K+ current-driven changes in input resistances. Together, these findings demonstrate that NALCN-encoded Na+ leak channels contribute to regulating daily rhythms in the excitability of SCN neurons by a mechanism that depends on K+ current-mediated rhythmic changes in intrinsic membrane properties.SIGNIFICANCE STATEMENT Elucidating the ionic mechanisms responsible for generating daily rhythms in the rates of spontaneous action potential firing of neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals, is an important step toward understanding how the molecular clock controls circadian rhythms in physiology and behavior. While numerous studies have focused on identifying subthreshold K+ channel(s) that mediate day-night changes in the firing rates of SCN neurons, a role for Na+ leak currents has also been suggested. The results of the experiments presented here demonstrate that NALCN-encoded Na+ leak currents differentially modulate daily rhythms in the daytime/nighttime repetitive firing rates of SCN neurons as a consequence of rhythmic changes in subthreshold K+ currents.
Collapse
Affiliation(s)
- Nien-Du Yang
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri 63110
| | | | - Tracey O Hermanstyne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Jeanne M Nerbonne
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri 63110
- Department of Medicine, Cardiovascular Division
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
24
|
Francis TC, Porcu A. Emotionally clocked out: cell-type specific regulation of mood and anxiety by the circadian clock system in the brain. Front Mol Neurosci 2023; 16:1188184. [PMID: 37441675 PMCID: PMC10333695 DOI: 10.3389/fnmol.2023.1188184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/29/2023] [Indexed: 07/15/2023] Open
Abstract
Circadian rhythms are self-sustained oscillations of biological systems that allow an organism to anticipate periodic changes in the environment and optimally align feeding, sleep, wakefulness, and the physiological and biochemical processes that support them within the 24 h cycle. These rhythms are generated at a cellular level by a set of genes, known as clock genes, which code for proteins that inhibit their own transcription in a negative feedback loop and can be perturbed by stress, a risk factor for the development of mood and anxiety disorders. A role for circadian clocks in mood and anxiety has been suggested for decades on the basis of clinical observations, and the dysregulation of circadian rhythms is a prominent clinical feature of stress-related disorders. Despite our understanding of central clock structure and function, the effect of circadian dysregulation in different neuronal subtypes in the suprachiasmatic nucleus (SCN), the master pacemaker region, as well as other brain systems regulating mood, including mesolimbic and limbic circuits, is just beginning to be elucidated. In the brain, circadian clocks regulate neuronal physiological functions, including neuronal activity, synaptic plasticity, protein expression, and neurotransmitter release which in turn affect mood-related behaviors via cell-type specific mechanisms. Both animal and human studies have revealed an association between circadian misalignment and mood disorders and suggest that internal temporal desynchrony might be part of the etiology of psychiatric disorders. To date, little work has been conducted associating mood-related phenotypes to cell-specific effects of the circadian clock disruptions. In this review, we discuss existing literature on how clock-driven changes in specific neuronal cell types might disrupt phase relationships among cellular communication, leading to neuronal circuit dysfunction and changes in mood-related behavior. In addition, we examine cell-type specific circuitry underlying mood dysfunction and discuss how this circuitry could affect circadian clock. We provide a focus for future research in this area and a perspective on chronotherapies for mood and anxiety disorders.
Collapse
Affiliation(s)
- T. Chase Francis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Alessandra Porcu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
25
|
Imai N. Molecular and Cellular Neurobiology of Circadian and Circannual Rhythms in Migraine: A Narrative Review. Int J Mol Sci 2023; 24:10092. [PMID: 37373239 DOI: 10.3390/ijms241210092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Migraine-a primary headache-has circadian and circannual rhythms in the onset of attacks. The circadian and circannual rhythms involve the hypothalamus, which is strongly associated with pain processing in migraines. Moreover, the role of melatonin in circadian rhythms has been implied in the pathophysiology of migraines. However, the prophylactic effect of melatonin in migraines is controversial. Calcitonin gene-related peptide (CGRP) has recently attracted attention in the pathophysiology and treatment of migraines. Pituitary adenylate cyclase-activating peptide (PACAP)-a neuropeptide identical to CGRP-is a potential therapeutic target after CGRP. PACAP is involved in the regulation of circadian entrainment to light. This review provides an overview of circadian and circannual rhythms in the hypothalamus and describes the relationship between migraines and the molecular and cellular neurobiology of circadian and circannual rhythms. Furthermore, the potential clinical applications of PACAP are presented.
Collapse
Affiliation(s)
- Noboru Imai
- Department of Neurology and Headache Center, Japanese Red Cross Shizuoka Hospital, Shizuoka 420-0853, Japan
| |
Collapse
|
26
|
Abstract
Sonic Hedgehog signaling and primary cilia control the core mammalian circadian clock.
Collapse
Affiliation(s)
- Dong Won Kim
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
27
|
Tu HQ, Li S, Xu YL, Zhang YC, Li PY, Liang LY, Song GP, Jian XX, Wu M, Song ZQ, Li TT, Hu HB, Yuan JF, Shen XL, Li JN, Han QY, Wang K, Zhang T, Zhou T, Li AL, Zhang XM, Li HY. Rhythmic cilia changes support SCN neuron coherence in circadian clock. Science 2023; 380:972-979. [PMID: 37262147 DOI: 10.1126/science.abm1962] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 04/13/2023] [Indexed: 06/03/2023]
Abstract
The suprachiasmatic nucleus (SCN) drives circadian clock coherence through intercellular coupling, which is resistant to environmental perturbations. We report that primary cilia are required for intercellular coupling among SCN neurons to maintain the robustness of the internal clock in mice. Cilia in neuromedin S-producing (NMS) neurons exhibit pronounced circadian rhythmicity in abundance and length. Genetic ablation of ciliogenesis in NMS neurons enabled a rapid phase shift of the internal clock under jet-lag conditions. The circadian rhythms of individual neurons in cilia-deficient SCN slices lost their coherence after external perturbations. Rhythmic cilia changes drive oscillations of Sonic Hedgehog (Shh) signaling and clock gene expression. Inactivation of Shh signaling in NMS neurons phenocopied the effects of cilia ablation. Thus, cilia-Shh signaling in the SCN aids intercellular coupling.
Collapse
Affiliation(s)
- Hai-Qing Tu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Sen Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Ling Xu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Cheng Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Pei-Yao Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Li-Yun Liang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Guang-Ping Song
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Xiao Jian
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Min Wu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Zeng-Qing Song
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Ting-Ting Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Huai-Bin Hu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Jin-Feng Yuan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Lin Shen
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Jia-Ning Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Qiu-Ying Han
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Kai Wang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Tao Zhang
- Laboratory Animal Center, Academy of Military Medical Sciences, Beijing, China
| | - Tao Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Ai-Ling Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xue-Min Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hui-Yan Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Medrano M, Allaoui W, Van Bulck M, Thys S, Makrini-Maleville L, Seuntjens E, De Vos WH, Valjent E, Gaszner B, Van Eeckhaut A, Smolders I, De Bundel D. Neuroanatomical characterization of the Nmu-Cre knock-in mice reveals an interconnected network of unique neuropeptidergic cells. Open Biol 2023; 13:220353. [PMID: 37311538 PMCID: PMC10264104 DOI: 10.1098/rsob.220353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/10/2023] [Indexed: 06/15/2023] Open
Abstract
Neuromedin U (NMU) is an evolutionary conserved neuropeptide that has been implicated in multiple processes, such as circadian regulation, energy homeostasis, reward processing and stress coping. Although the central expression of NMU has been addressed previously, the lack of specific and sensitive tools has prevented a comprehensive characterization of NMU-expressing neurons in the brain. We have generated a knock-in mouse model constitutively expressing Cre recombinase under the Nmu promoter. We have validated the model using a multi-level approach based on quantitative reverse-transcription polymerase chain reactions, in situ hybridization, a reporter mouse line and an adenoviral vector driving Cre-dependent expression of a fluorescent protein. Using the Nmu-Cre mouse, we performed a complete characterization of NMU expression in adult mouse brain, unveiling a potential midline NMU modulatory circuit with the ventromedial hypothalamic nucleus (VMH) as a key node. Moreover, immunohistochemical analysis suggested that NMU neurons in the VMH mainly constitute a unique population of hypothalamic cells. Taken together, our results suggest that Cre expression in the Nmu-Cre mouse model largely reflects NMU expression in the adult mouse brain, without altering endogenous NMU expression. Thus, the Nmu-Cre mouse model is a powerful and sensitive tool to explore the role of NMU neurons in mice.
Collapse
Affiliation(s)
- Mireia Medrano
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Wissal Allaoui
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Mathias Van Bulck
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Sofie Thys
- Department of Veterinary Sciences, Laboratory of Cell Biology and Histology and Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, 2610 Antwerp, Belgium
| | | | - Eve Seuntjens
- Department of Biology, Laboratory of Developmental Neurobiology, KU Leuven, 3000 Leuven, Belgium
| | - Winnok H. De Vos
- Department of Veterinary Sciences, Laboratory of Cell Biology and Histology and Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, 2610 Antwerp, Belgium
- μNEURO Research Centre of Excellence, University of Antwerp, 2610 Antwerp, Belgium
- Antwerp Centre for Advanced Microscopy (ACAM), 2610 Wilrijk, Belgium
| | - Emmanuel Valjent
- IGF, Université de Montpellier, CNRS, Inserm, 34094 Montpellier, France
| | - Bálazs Gaszner
- Medical School, Research Group for Mood Disorders, Department of Anatomy and Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Ann Van Eeckhaut
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Ilse Smolders
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Dimitri De Bundel
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| |
Collapse
|
29
|
Joye DAM, Rohr KE, Suenkens K, Wuorinen A, Inda T, Arzbecker M, Mueller E, Huber A, Pancholi H, Blackmore MG, Carmona-Alcocer V, Evans JA. Somatostatin regulates central clock function and circadian responses to light. Proc Natl Acad Sci U S A 2023; 120:e2216820120. [PMID: 37098068 PMCID: PMC10160998 DOI: 10.1073/pnas.2216820120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 03/21/2023] [Indexed: 04/26/2023] Open
Abstract
Daily and annual changes in light are processed by central clock circuits that control the timing of behavior and physiology. The suprachiasmatic nucleus (SCN) in the anterior hypothalamus processes daily photic inputs and encodes changes in day length (i.e., photoperiod), but the SCN circuits that regulate circadian and photoperiodic responses to light remain unclear. Somatostatin (SST) expression in the hypothalamus is modulated by photoperiod, but the role of SST in SCN responses to light has not been examined. Our results indicate that SST signaling regulates daily rhythms in behavior and SCN function in a manner influenced by sex. First, we use cell-fate mapping to provide evidence that SST in the SCN is regulated by light via de novo Sst activation. Next, we demonstrate that Sst -/- mice display enhanced circadian responses to light, with increased behavioral plasticity to photoperiod, jetlag, and constant light conditions. Notably, lack of Sst -/- eliminated sex differences in photic responses due to increased plasticity in males, suggesting that SST interacts with clock circuits that process light differently in each sex. Sst -/- mice also displayed an increase in the number of retinorecipient neurons in the SCN core, which express a type of SST receptor capable of resetting the molecular clock. Last, we show that lack of SST signaling modulates central clock function by influencing SCN photoperiodic encoding, network after-effects, and intercellular synchrony in a sex-specific manner. Collectively, these results provide insight into peptide signaling mechanisms that regulate central clock function and its response to light.
Collapse
Affiliation(s)
- Deborah A. M. Joye
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Kayla E. Rohr
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Kimberlee Suenkens
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Alissa Wuorinen
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Thomas Inda
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Madeline Arzbecker
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Emma Mueller
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Alec Huber
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Harshida Pancholi
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | | | | | - Jennifer A. Evans
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| |
Collapse
|
30
|
Piet R. Circadian and kisspeptin regulation of the preovulatory surge. Peptides 2023; 163:170981. [PMID: 36842628 DOI: 10.1016/j.peptides.2023.170981] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 02/28/2023]
Abstract
Fertility in mammals is ultimately controlled by a small population of neurons - the gonadotropin-releasing hormone (GnRH) neurons - located in the ventral forebrain. GnRH neurons control gonadal function through the release of GnRH, which in turn stimulates the secretion of the anterior pituitary gonadotropins luteinizing hormone (LH) and follicle-stimulating hormone (FSH). In spontaneous ovulators, ovarian follicle maturation eventually stimulates, via sex steroid feedback, the mid-cycle surge in GnRH and LH secretion that causes ovulation. The GnRH/LH surge is initiated in many species just before the onset of activity through processes controlled by the central circadian clock, ensuring that the neuroendocrine control of ovulation and sex behavior are coordinated. This review aims to give an overview of anatomical and functional studies that collectively reveal some of the mechanisms through which the central circadian clock regulates GnRH neurons and their afferent circuits to drive the preovulatory surge.
Collapse
Affiliation(s)
- Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States.
| |
Collapse
|
31
|
Adlanmerini M, Lazar MA. The REV-ERB Nuclear Receptors: Timekeepers for the Core Clock Period and Metabolism. Endocrinology 2023; 164:bqad069. [PMID: 37149727 PMCID: PMC10413432 DOI: 10.1210/endocr/bqad069] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/20/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
REV-ERB nuclear receptors are potent transcriptional repressors that play an important role in the core mammalian molecular clock and metabolism. Deletion of both REV-ERBα and its largely redundant isoform REV-ERBβ in a murine tissue-specific manner have shed light on their specific functions in clock mechanisms and circadian metabolism. This review highlights recent findings that establish REV-ERBs as crucial circadian timekeepers in a variety of tissues, regulating overlapping and distinct processes that maintain normal physiology and protect from metabolic dysfunction.
Collapse
Affiliation(s)
- Marine Adlanmerini
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
32
|
Fukada Y. Kinase signaling in distinct neuronal populations in the mouse brain controls sleep homeostasis and the circadian clock. Proc Natl Acad Sci U S A 2023; 120:e2303354120. [PMID: 37018202 PMCID: PMC10104544 DOI: 10.1073/pnas.2303354120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023] Open
Affiliation(s)
- Yoshitaka Fukada
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo113-0033, Japan
- Circadian Clock Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo156-8506, Japan
| |
Collapse
|
33
|
MUW researcher of the month. Wien Klin Wochenschr 2023; 135:217-218. [PMID: 37081182 DOI: 10.1007/s00508-023-02203-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
|
34
|
Asano F, Kim SJ, Fujiyama T, Miyoshi C, Hotta-Hirashima N, Asama N, Iwasaki K, Kakizaki M, Mizuno S, Mieda M, Sugiyama F, Takahashi S, Shi S, Hirano A, Funato H, Yanagisawa M. SIK3-HDAC4 in the suprachiasmatic nucleus regulates the timing of arousal at the dark onset and circadian period in mice. Proc Natl Acad Sci U S A 2023; 120:e2218209120. [PMID: 36877841 PMCID: PMC10089210 DOI: 10.1073/pnas.2218209120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/07/2023] [Indexed: 03/08/2023] Open
Abstract
Mammals exhibit circadian cycles of sleep and wakefulness under the control of the suprachiasmatic nucleus (SCN), such as the strong arousal phase-locked to the beginning of the dark phase in laboratory mice. Here, we demonstrate that salt-inducible kinase 3 (SIK3) deficiency in gamma-aminobutyric acid (GABA)-ergic neurons or neuromedin S (NMS)-producing neurons delayed the arousal peak phase and lengthened the behavioral circadian cycle under both 12-h light:12-h dark condition (LD) and constant dark condition (DD) without changing daily sleep amounts. In contrast, the induction of a gain-of-function mutant allele of Sik3 in GABAergic neurons exhibited advanced activity onset and a shorter circadian period. Loss of SIK3 in arginine vasopressin (AVP)-producing neurons lengthened the circadian cycle, but the arousal peak phase was similar to that in control mice. Heterozygous deficiency of histone deacetylase (HDAC) 4, a SIK3 substrate, shortened the circadian cycle, whereas mice with HDAC4 S245A, which is resistant to phosphorylation by SIK3, delayed the arousal peak phase. Phase-delayed core clock gene expressions were detected in the liver of mice lacking SIK3 in GABAergic neurons. These results suggest that the SIK3-HDAC4 pathway regulates the circadian period length and the timing of arousal through NMS-positive neurons in the SCN.
Collapse
Affiliation(s)
- Fuyuki Asano
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Staci J. Kim
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Tomoyuki Fujiyama
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Chika Miyoshi
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Noriko Hotta-Hirashima
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Nodoka Asama
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Kanako Iwasaki
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Miyo Kakizaki
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa920-8640, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Shoi Shi
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Arisa Hirano
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Hiromasa Funato
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
- Department of Anatomy, Toho University Graduate School of Medicine, Tokyo143-8540, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX75390
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba305-8577, Japan
| |
Collapse
|
35
|
Arginine-vasopressin-expressing neurons in the murine suprachiasmatic nucleus exhibit a circadian rhythm in network coherence in vivo. Proc Natl Acad Sci U S A 2023; 120:e2209329120. [PMID: 36656857 PMCID: PMC9942887 DOI: 10.1073/pnas.2209329120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The suprachiasmatic nucleus (SCN) is composed of functionally distinct subpopulations of GABAergic neurons which form a neural network responsible for synchronizing most physiological and behavioral circadian rhythms in mammals. To date, little is known regarding which aspects of SCN rhythmicity are generated by individual SCN neurons, and which aspects result from neuronal interaction within a network. Here, we utilize in vivo miniaturized microscopy to measure fluorescent GCaMP-reported calcium dynamics in arginine vasopressin (AVP)-expressing neurons in the intact SCN of awake, behaving mice. We report that SCN AVP neurons exhibit periodic, slow calcium waves which we demonstrate, using in vivo electrical recordings, likely reflect burst firing. Further, we observe substantial heterogeneity of function in that AVP neurons exhibit unstable rhythms, and relatively weak rhythmicity at the population level. Network analysis reveals that correlated cellular behavior, or coherence, among neuron pairs also exhibited stochastic rhythms with about 33% of pairs rhythmic at any time. Unlike single-cell variables, coherence exhibited a strong rhythm at the population level with time of maximal coherence among AVP neuronal pairs at CT/ZT 6 and 9, coinciding with the timing of maximal neuronal activity for the SCN as a whole. These results demonstrate robust circadian variation in the coordination between stochastically rhythmic neurons and that interactions between AVP neurons in the SCN may be more influential than single-cell activity in the regulation of circadian rhythms. Furthermore, they demonstrate that cells in this circuit, like those in many other circuits, exhibit profound heterogenicity of function over time and space.
Collapse
|
36
|
Zheng Y, Pan L, Wang F, Yan J, Wang T, Xia Y, Yao L, Deng K, Zheng Y, Xia X, Su Z, Chen H, Lin J, Ding Z, Zhang K, Zhang M, Chen Y. Neural function of Bmal1: an overview. Cell Biosci 2023; 13:1. [PMID: 36593479 PMCID: PMC9806909 DOI: 10.1186/s13578-022-00947-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
Bmal1 (Brain and muscle arnt-like, or Arntl) is a bHLH/PAS domain transcription factor central to the transcription/translation feedback loop of the biologic clock. Although Bmal1 is well-established as a major regulator of circadian rhythm, a growing number of studies in recent years have shown that dysfunction of Bmal1 underlies a variety of psychiatric, neurodegenerative-like, and endocrine metabolism-related disorders, as well as potential oncogenic roles. In this review, we systematically summarized Bmal1 expression in different brain regions, its neurological functions related or not to circadian rhythm and biological clock, and pathological phenotypes arising from Bmal1 knockout. This review also discusses oscillation and rhythmicity, especially in the suprachiasmatic nucleus, and provides perspective on future progress in Bmal1 research.
Collapse
Affiliation(s)
- Yuanjia Zheng
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China ,grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingyun Pan
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Feixue Wang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinglan Yan
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Taiyi Wang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yucen Xia
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Yao
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kelin Deng
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqi Zheng
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoye Xia
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhikai Su
- grid.411866.c0000 0000 8848 7685The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong China
| | - Hongjie Chen
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jie Lin
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenwei Ding
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kaitong Zhang
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meng Zhang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongjun Chen
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China ,grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China ,Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China
| |
Collapse
|
37
|
Abstract
Our physiology and behavior follow precise daily programs that adapt us to the alternating opportunities and challenges of day and night. Under experimental isolation, these rhythms persist with a period of approximately one day (circadian), demonstrating their control by an internal autonomous clock. Circadian time is created at the cellular level by a transcriptional/translational feedback loop (TTFL) in which the protein products of the Period and Cryptochrome genes inhibit their own transcription. Because the accumulation of protein is slow and delayed, the system oscillates spontaneously with a period of ∼24 hours. This cell-autonomous TTFL controls cycles of gene expression in all major tissues and these cycles underpin our daily metabolic programs. In turn, our innumerable cellular clocks are coordinated by a central pacemaker, the suprachiasmatic nucleus (SCN) of the hypothalamus. When isolated in slice culture, the SCN TTFL and its dependent cycles of neural activity persist indefinitely, operating as "a clock in a dish". In vivo, SCN time is synchronized to solar time by direct innervation from specialized retinal photoreceptors. In turn, the precise circadian cycle of action potential firing signals SCN-generated time to hypothalamic and brain stem targets, which co-ordinate downstream autonomic, endocrine, and behavioral (feeding) cues to synchronize and sustain the distributed cellular clock network. Circadian time therefore pervades every level of biological organization, from molecules to society. Understanding its mechanisms offers important opportunities to mitigate the consequences of circadian disruption, so prevalent in modern societies, that arise from shiftwork, aging, and neurodegenerative diseases, not least Huntington's disease.
Collapse
Affiliation(s)
- Andrew P. Patton
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | |
Collapse
|
38
|
Regulation of CRE-Dependent Transcriptional Activity in a Mouse Suprachiasmatic Nucleus Cell Line. Int J Mol Sci 2022; 23:ijms232012226. [PMID: 36293078 PMCID: PMC9602552 DOI: 10.3390/ijms232012226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/23/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
We evaluated the signalling framework of immortalized cells from the hypothalamic suprachiasmatic nucleus (SCN) of the mouse. We selected a vasoactive intestinal peptide (VIP)-positive sub-clone of immortalized mouse SCN-cells stably expressing a cAMP-regulated-element (CRE)-luciferase construct named SCNCRE. We characterized these cells in terms of their status as neuronal cells, as well as for important components of the cAMP-dependent signal transduction pathway and compared them to SCN ex vivo. SCNCRE cells were treated with agents that modulate different intracellular signalling pathways to investigate their potency and timing for transcriptional CRE-dependent signalling. Several activating pathways modulate SCN neuronal signalling via the cAMP-regulated-element (CRE: TGACGCTA) and phosphorylation of transcription factors such as cAMP-regulated-element-binding protein (CREB). CRE-luciferase activity induced by different cAMP-signalling pathway-modulating agents displayed a variety of substance-specific dose and time-dependent profiles and interactions relevant to the regulation of SCN physiology. Moreover, the induction of the protein kinase C (PKC) pathway by phorbol ester application modulates the CRE-dependent signalling pathway as well. In conclusion, the cAMP/PKA- and the PKC-regulated pathways individually and in combination modulate the final CRE-dependent transcriptional output.
Collapse
|
39
|
A hypothalamic dopamine locus for psychostimulant-induced hyperlocomotion in mice. Nat Commun 2022; 13:5944. [PMID: 36209152 PMCID: PMC9547883 DOI: 10.1038/s41467-022-33584-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/22/2022] [Indexed: 11/29/2022] Open
Abstract
The lateral septum (LS) has been implicated in the regulation of locomotion. Nevertheless, the neurons synchronizing LS activity with the brain’s clock in the suprachiasmatic nucleus (SCN) remain unknown. By interrogating the molecular, anatomical and physiological heterogeneity of dopamine neurons of the periventricular nucleus (PeVN; A14 catecholaminergic group), we find that Th+/Dat1+ cells from its anterior subdivision innervate the LS in mice. These dopamine neurons receive dense neuropeptidergic innervation from the SCN. Reciprocal viral tracing in combination with optogenetic stimulation ex vivo identified somatostatin-containing neurons in the LS as preferred synaptic targets of extrahypothalamic A14 efferents. In vivo chemogenetic manipulation of anterior A14 neurons impacted locomotion. Moreover, chemogenetic inhibition of dopamine output from the anterior PeVN normalized amphetamine-induced hyperlocomotion, particularly during sedentary periods. Cumulatively, our findings identify a hypothalamic locus for the diurnal control of locomotion and pinpoint a midbrain-independent cellular target of psychostimulants. The psychostimulant-sensitive neural mechanism linking the circadian clock to locomotion is unknown. Here, hypothalamic A14 neurons are shown to time diurnal activity by entraining the lateral septum, and their activity is shown to be sensitive to amphetamine.
Collapse
|
40
|
Porcu A, Nilsson A, Booreddy S, Barnes SA, Welsh DK, Dulcis D. Seasonal changes in day length induce multisynaptic neurotransmitter switching to regulate hypothalamic network activity and behavior. SCIENCE ADVANCES 2022; 8:eabn9867. [PMID: 36054362 PMCID: PMC10848959 DOI: 10.1126/sciadv.abn9867] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 07/19/2022] [Indexed: 05/18/2023]
Abstract
Seasonal changes in day length (photoperiod) affect numerous physiological functions. The suprachiasmatic nucleus (SCN)-paraventricular nucleus (PVN) axis plays a key role in processing photoperiod-related information. Seasonal variations in SCN and PVN neurotransmitter expression have been observed in humans and animal models. However, the molecular mechanisms by which the SCN-PVN network responds to altered photoperiod is unknown. Here, we show in mice that neuromedin S (NMS) and vasoactive intestinal polypeptide (VIP) neurons in the SCN display photoperiod-induced neurotransmitter plasticity. In vivo recording of calcium dynamics revealed that NMS neurons alter PVN network activity in response to winter-like photoperiod. Chronic manipulation of NMS neurons is sufficient to induce neurotransmitter switching in PVN neurons and affects locomotor activity. Our findings reveal previously unidentified molecular adaptations of the SCN-PVN network in response to seasonality and the role for NMS neurons in adjusting hypothalamic function to day length via a coordinated multisynaptic neurotransmitter switching affecting behavior.
Collapse
Affiliation(s)
- Alessandra Porcu
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA
| | - Anna Nilsson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Sathwik Booreddy
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Samuel A. Barnes
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - David K. Welsh
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA
| | - Davide Dulcis
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
41
|
The duper mutation reveals previously unsuspected functions of Cryptochrome 1 in circadian entrainment and heart disease. Proc Natl Acad Sci U S A 2022; 119:e2121883119. [PMID: 35930669 PMCID: PMC9371649 DOI: 10.1073/pnas.2121883119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The Cryptochrome 1 (Cry1)-deficient duper mutant hamster has a short free-running period in constant darkness (τDD) and shows large phase shifts in response to brief light pulses. We tested whether this measure of the lability of the circadian phase is a general characteristic of Cry1-null animals and whether it indicates resistance to jet lag. Upon advance of the light:dark (LD) cycle, both duper hamsters and Cry1-/- mice re-entrained locomotor rhythms three times as fast as wild types. However, accelerated re-entrainment was dissociated from the amplified phase-response curve (PRC): unlike duper hamsters, Cry1-/- mice show no amplification of the phase response to 15' light pulses. Neither the amplified acute shifts nor the increased rate of re-entrainment in duper mutants is due to acceleration of the circadian clock: when mutants drank heavy water to lengthen the period, these aspects of the phenotype persisted. In light of the health consequences of circadian misalignment, we examined effects of duper and phase shifts on a hamster model of heart disease previously shown to be aggravated by repeated phase shifts. The mutation shortened the lifespan of cardiomyopathic hamsters relative to wild types, but this effect was eliminated when mutants experienced 8-h phase shifts every second week, to which they rapidly re-entrained. Our results reveal previously unsuspected roles of Cry1 in phase shifting and longevity in the face of heart disease. The duper mutant offers new opportunities to understand the basis of circadian disruption and jet lag.
Collapse
|
42
|
Tonsfeldt KJ, Cui LJ, Lee J, Walbeek TJ, Brusman LE, Jin Y, Mieda M, Gorman MR, Mellon PL. Female fertility does not require Bmal1 in suprachiasmatic nucleus neurons expressing arginine vasopressin, vasoactive intestinal peptide, or neuromedin-S. Front Endocrinol (Lausanne) 2022; 13:956169. [PMID: 35992114 PMCID: PMC9389073 DOI: 10.3389/fendo.2022.956169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/06/2022] [Indexed: 01/27/2023] Open
Abstract
Disruptions to the circadian system alter reproductive capacity, particularly in females. Mice lacking the core circadian clock gene, Bmal1, are infertile and have evidence of neuroendocrine disruption including the absence of the preovulatory luteinizing hormone (LH) surge and enhanced responsiveness to exogenous kisspeptin. Here, we explore the role of Bmal1 in suprachiasmatic nucleus (SCN) neuron populations known to project to the neuroendocrine axis. We generated four mouse lines using Cre/Lox technology to create conditional deletion of Bmal1 in arginine vasopressin (Bmal1fl/fl:Avpcre ), vasoactive intestinal peptide (Bmal1fl/fl:Vipcre ), both (Bmal1fl/fl:Avpcre+Vipcre ), and neuromedin-s (Bmal1fl/fl:Nmscre ) neurons. We demonstrate that the loss of Bmal1 in these populations has substantial effects on home-cage circadian activity and temperature rhythms. Despite this, we found that female mice from these lines demonstrated normal estrus cycles, fecundity, kisspeptin responsiveness, and inducible LH surge. We found no evidence of reproductive disruption in constant darkness. Overall, our results indicate that while conditional Bmal1 knockout in AVP, VIP, or NMS neurons is sufficient to disrupted locomotor activity, this disruption is insufficient to recapitulate the neuroendocrine reproductive effects of the whole-body Bmal1 knockout.
Collapse
Affiliation(s)
- Karen J. Tonsfeldt
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, United States
| | - Laura J. Cui
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Jinkwon Lee
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Thijs J. Walbeek
- Department of Psychology, University of California, San Diego, La Jolla, CA, United States
| | - Liza E. Brusman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Ye Jin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Michael R. Gorman
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, United States
- Department of Psychology, University of California, San Diego, La Jolla, CA, United States
| | - Pamela L. Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, United States
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
43
|
Yamaguchi Y, Murai I, Takeda M, Doi S, Seta T, Hanada R, Kangawa K, Okamura H, Miyake T, Doi M. <i>Nmu</i>/<i>Nms</i>/<i>Gpr176</i> Triple-Deficient Mice Show Enhanced Light-Resetting of Circadian Locomotor Activity. Biol Pharm Bull 2022; 45:1172-1179. [DOI: 10.1248/bpb.b22-00260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yoshiaki Yamaguchi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Iori Murai
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Momoko Takeda
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Shotaro Doi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Takehito Seta
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Reiko Hanada
- Department of Neurophysiology, Faculty of Medicine, Oita University
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute
| | - Hitoshi Okamura
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Takahito Miyake
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Masao Doi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
44
|
Kauffman AS. Neuroendocrine mechanisms underlying estrogen positive feedback and the LH surge. Front Neurosci 2022; 16:953252. [PMID: 35968365 PMCID: PMC9364933 DOI: 10.3389/fnins.2022.953252] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/08/2022] [Indexed: 01/26/2023] Open
Abstract
A fundamental principle in reproductive neuroendocrinology is sex steroid feedback: steroid hormones secreted by the gonads circulate back to the brain to regulate the neural circuits governing the reproductive neuroendocrine axis. These regulatory feedback loops ultimately act to modulate gonadotropin-releasing hormone (GnRH) secretion, thereby affecting gonadotropin secretion from the anterior pituitary. In females, rising estradiol (E2) during the middle of the menstrual (or estrous) cycle paradoxically "switch" from being inhibitory on GnRH secretion ("negative feedback") to stimulating GnRH release ("positive feedback"), resulting in a surge in GnRH secretion and a downstream LH surge that triggers ovulation. While upstream neural afferents of GnRH neurons, including kisspeptin neurons in the rostral hypothalamus, are proposed as critical loci of E2 feedback action, the underlying mechanisms governing the shift between E2 negative and positive feedback are still poorly understood. Indeed, the precise cell targets, neural signaling factors and receptors, hormonal pathways, and molecular mechanisms by which ovarian-derived E2 indirectly stimulates GnRH surge secretion remain incompletely known. In many species, there is also a circadian component to the LH surge, restricting its occurrence to specific times of day, but how the circadian clock interacts with endocrine signals to ultimately time LH surge generation also remains a major gap in knowledge. Here, we focus on classic and recent data from rodent models and discuss the consensus knowledge of the neural players, including kisspeptin, the suprachiasmatic nucleus, and glia, as well as endocrine players, including estradiol and progesterone, in the complex regulation and generation of E2-induced LH surges in females.
Collapse
|
45
|
Patton AP, Smyllie NJ, Chesham JE, Hastings MH. Astrocytes Sustain Circadian Oscillation and Bidirectionally Determine Circadian Period, But Do Not Regulate Circadian Phase in the Suprachiasmatic Nucleus. J Neurosci 2022; 42:5522-5537. [PMID: 35610047 PMCID: PMC9295834 DOI: 10.1523/jneurosci.2337-21.2022] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/20/2022] [Accepted: 05/03/2022] [Indexed: 11/21/2022] Open
Abstract
The suprachiasmatic nucleus (SCN) is the master circadian clock of mammals, generating and transmitting an internal representation of environmental time that is produced by the cell-autonomous transcriptional/post-translational feedback loops (TTFLs) of the 10,000 neurons and 3500 glial cells. Recently, we showed that TTFL function in SCN astrocytes alone is sufficient to drive circadian timekeeping and behavior, raising questions about the respective contributions of astrocytes and neurons within the SCN circuit. We compared their relative roles in circadian timekeeping in mouse SCN explants, of either sex. Treatment with the glial-specific toxin fluorocitrate revealed a requirement for metabolically competent astrocytes for circuit-level timekeeping. Recombinase-mediated genetically complemented Cryptochrome (Cry) proteins in Cry1-deficient and/or Cry2-deficient SCNs were used to compare the influence of the TTFLs of neurons or astrocytes in the initiation of de novo oscillation or in pacemaking. While neurons and astrocytes both initiated de novo oscillation and lengthened the period equally, their kinetics were different, with astrocytes taking twice as long. Furthermore, astrocytes could shorten the period, but not as potently as neurons. Chemogenetic manipulation of Gi- and Gq-coupled signaling pathways in neurons acutely advanced or delayed the ensemble phase, respectively. In contrast, comparable manipulations in astrocytes were without effect. Thus, astrocytes can initiate SCN rhythms and bidirectionally control the SCN period, albeit with lower potency than neurons. Nevertheless, their activation does not influence the SCN phase. The emergent SCN properties of high-amplitude oscillation, initiation of rhythmicity, pacemaking, and phase are differentially regulated: astrocytes and neurons sustain the ongoing oscillation, but its phase is determined by neurons.SIGNIFICANCE STATEMENT The hypothalamic suprachiasmatic nucleus (SCN) encodes and disseminates time-of-day information to allow mammals to adapt their physiology to daily environmental cycles. Recent investigations have revealed a role for astrocytes, in addition to neurons, in the regulation of this rhythm. Using pharmacology, genetic complementation, and chemogenetics, we compared the abilities of neurons and astrocytes in determining the emergent SCN properties of high-amplitude oscillation, initiation of rhythmicity, pacemaking, and determination of phase. These findings parameterize the circadian properties of the astrocyte population in the SCN and reveal the types of circadian information that astrocytes and neurons can contribute within their heterogeneous cellular network.
Collapse
Affiliation(s)
- Andrew P Patton
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Nicola J Smyllie
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Johanna E Chesham
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | | |
Collapse
|
46
|
El Cheikh Hussein L, Fontanaud P, Mollard P, Bonnefont X. Nested calcium dynamics support daily cell unity and diversity in the suprachiasmatic nuclei of free-behaving mice. PNAS NEXUS 2022; 1:pgac112. [PMID: 36741435 PMCID: PMC9896879 DOI: 10.1093/pnasnexus/pgac112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023]
Abstract
The suprachiasmatic nuclei (SCN) of the anterior hypothalamus host the circadian pacemaker that synchronizes mammalian rhythms with the day-night cycle. SCN neurons are intrinsically rhythmic, thanks to a conserved cell-autonomous clock mechanism. In addition, circuit-level emergent properties confer a unique degree of precision and robustness to SCN neuronal rhythmicity. However, the multicellular functional organization of the SCN is not yet fully understood. Indeed, although SCN neurons are well-coordinated, experimental evidences indicate that some neurons oscillate out of phase in SCN explants, and possibly to a larger extent in vivo. Here, to tackle this issue we used microendoscopic Ca2+ i imaging and investigated SCN rhythmicity at a single cell resolution in free-behaving mice. We found that SCN neurons in vivo exhibited fast Ca2+ i spikes superimposed upon slow changes in baseline Ca2+ i levels. Both spikes and baseline followed a time-of-day modulation in many neurons, but independently from each other. Daily rhythms in basal Ca2+ i were highly coordinated, while spike activity from the same neurons peaked at multiple times of the light cycle, and unveiled clock-independent coactivity in neuron subsets. Hence, fast Ca2+ i spikes and slow changes in baseline Ca2+ i levels highlighted how multiple individual activity patterns could articulate within the temporal unity of the SCN cell network in vivo, and provided support for a multiplex neuronal code in the circadian pacemaker.
Collapse
Affiliation(s)
- Lama El Cheikh Hussein
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, IGF, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France,BioCampus Montpellier, Université de Montpellier, CNRS, INSERM, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France
| | - Pierre Fontanaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, IGF, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France,BioCampus Montpellier, Université de Montpellier, CNRS, INSERM, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France
| | - Patrice Mollard
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, IGF, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France,BioCampus Montpellier, Université de Montpellier, CNRS, INSERM, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France
| | | |
Collapse
|
47
|
Peng Y, Tsuno Y, Matsui A, Hiraoka Y, Tanaka K, Horike SI, Daikoku T, Mieda M. Cell Type-Specific Genetic Manipulation and Impaired Circadian Rhythms in ViptTA Knock-In Mice. Front Physiol 2022; 13:895633. [PMID: 35592033 PMCID: PMC9110775 DOI: 10.3389/fphys.2022.895633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/18/2022] [Indexed: 11/30/2022] Open
Abstract
The suprachiasmatic nucleus (SCN), the central circadian clock in mammals, is a neural network consisting of various types of GABAergic neurons, which can be differentiated by the co-expression of specific peptides such as vasoactive intestinal peptide (VIP) and arginine vasopressin (AVP). VIP has been considered as a critical factor for the circadian rhythmicity and synchronization of individual SCN neurons. However, the precise mechanisms of how VIP neurons regulate SCN circuits remain incompletely understood. Here, we generated ViptTA knock-in mice that express tetracycline transactivator (tTA) specifically in VIP neurons by inserting tTA sequence at the start codon of Vip gene. The specific and efficient expression of tTA in VIP neurons was verified using EGFP reporter mice. In addition, combined with Avp-Cre mice, ViptTA mice enabled us to simultaneously apply different genetic manipulations to VIP and AVP neurons in the SCN. Immunostaining showed that VIP is expressed at a slightly reduced level in heterozygous ViptTA mice but is completely absent in homozygous mice. Consistently, homozygous ViptTA mice showed impaired circadian behavioral rhythms similar to those of Vip knockout mice, such as attenuated rhythmicity and shortened circadian period. In contrast, heterozygous mice demonstrated normal circadian behavioral rhythms comparable to wild-type mice. These data suggest that ViptTA mice are a valuable genetic tool to express exogenous genes specifically in VIP neurons in both normal and VIP-deficient mice, facilitating the study of VIP neuronal roles in the SCN neural network.
Collapse
Affiliation(s)
- Yubo Peng
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Ayako Matsui
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yuichi Hiraoka
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shin-ichi Horike
- Division of Integrated Omics Research, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- *Correspondence: Michihiro Mieda,
| |
Collapse
|
48
|
Time-restricted feeding entrains long-term behavioral changes through the IGF2-KCC2 pathway. iScience 2022; 25:104267. [PMID: 35521538 PMCID: PMC9062755 DOI: 10.1016/j.isci.2022.104267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 10/13/2021] [Accepted: 04/13/2022] [Indexed: 02/03/2023] Open
Abstract
The suprachiasmatic nucleus (SCN) integrates light and systemic signals from peripheral tissues to coordinate physiology and behavior daily rhythms. However, the contribution that nutrients and feeding patterns provide to the SCN network regulation remains controversial. Here, we found that time-restricted feeding (TRF) in ZT0-4 (Zeitgeber Time) generates a robust and long-term shift in locomotor behavior and increased wakefulness. Intracellular Ca2+ signals in SCN GABAergic neurons of freely moving mice showed significant activation after ZT0-4 TRF treatment. Furthermore, RNA-seq profiling of SCN showed that TRF during ZT0-4 increased Insulin-like Growth Factor 2 (Igf2) expression and dysregulated ion transporters, including the downregulation of Kcc2. SCN neuron-specific loss of function of Kcc2 amplified ZT0-4 TRF induced aftereffect. Moreover, overexpression of IGF2 in SCN GABAergic neurons extended the locomotion range, mirroring the TRF aftereffect. In summary, our study showed that the IGF2-KCC2 pathway plays an important role for TRF induced behavior changes.
Collapse
|
49
|
Luan J, Yang K, Ding Y, Zhang X, Wang Y, Cui H, Zhou D, Chen L, Ma Z, Wang W, Zhang W, Liu X. Valsartan-mediated chronotherapy in spontaneously hypertensive rats via targeting clock gene expression in vascular smooth muscle cells. Arch Physiol Biochem 2022; 128:490-500. [PMID: 31794282 DOI: 10.1080/13813455.2019.1695840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE This study was to investigate the underlying mechanisms of valsartan chronotherapy in regulating blood pressure variability. METHODS RT-PCR was used to assay clock genes expression rhythm in the hypothalamus, aortic vessels, and target organs after valsartan chronotherapy. WB was used to measure Period 1 (Per1), Period 2 (Per2) protein expression in aortic vessels, as well as to measure phosphorylation of 20-kDa regulatory myosin light chain (MLC20) in VSMCs. RESULTS Specific clock genes in the hypothalamus, and Per1 and Per2 in aorta abdominalis, exhibited disordered circadian expression in vivo. Valsartan asleep time administration (VSA) restored circadian clock gene expression in a tissue- and gene-specific manner. In vitro, VSA was more efficient in blocking angiotensin II relative to VWA, which led to differential circadian rhythms of Per1 and Per2, ultimately corrected MLC20 phosphorylation. CONCLUSION VSA may be efficacious in regulating circadian clock genes rhythm, then concomitantly correct circadian blood pressure rhythms.
Collapse
Affiliation(s)
- Jiajie Luan
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Kui Yang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Yanyun Ding
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Xiaotong Zhang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Yaqin Wang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Haiju Cui
- Department of Pharmacy, XuanCheng Vocational and Technical college, XuanCheng, Anhui, P.R. China
| | - Deixi Zhou
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Lu Chen
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Zhangqing Ma
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Wusan Wang
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Wen Zhang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
- School of Pharmacy, Wannan Medical College, Wuhu, P.R. China
| | - Xiaoyun Liu
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| |
Collapse
|
50
|
Shirato K, Sato S. Macrophage Meets the Circadian Clock: Implication of the Circadian Clock in the Role of Macrophages in Acute Lower Respiratory Tract Infection. Front Cell Infect Microbiol 2022; 12:826738. [PMID: 35281442 PMCID: PMC8904936 DOI: 10.3389/fcimb.2022.826738] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
The circadian rhythm is a biological system that creates daily variations of physiology and behavior with a 24-h cycle, which is precisely controlled by the molecular circadian clock. The circadian clock dominates temporal activity of physiological homeostasis at the molecular level, including endocrine secretion, metabolic, immune response, coupled with extrinsic environmental cues (e.g., light/dark cycles) and behavioral cues (e.g., sleep/wake cycles and feeding/fasting cycles). The other side of the clock is that the misaligned circadian rhythm contributes to the onset of a variety of diseases, such as cancer, metabolic diseases, and cardiovascular diseases, the acceleration of aging, and the development of systemic inflammation. The role played by macrophages is a key mediator between circadian disruption and systemic inflammation. At the molecular level, macrophage functions are under the direct control of the circadian clock, and thus the circadian misalignment remodels the phenotype of macrophages toward a ‘killer’ mode. Remarkably, the inflammatory macrophages induce systemic and chronic inflammation, leading to the development of inflammatory diseases and the dampened immune defensive machinery against infectious diseases such as COVID-19. Here, we discuss how the circadian clock regulates macrophage immune functions and provide the potential risk of misaligned circadian rhythms against inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Ken Shirato
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University School of Medicine, Mitaka, Japan
| | - Shogo Sato
- Center for Biological Clocks Research, Department of Biology, Texas A&M University, College Station, TX, United States
- *Correspondence: Shogo Sato,
| |
Collapse
|