51
|
Song Q, Feng G, Zhang J, Xia X, Ji M, Lv L, Ping Y. NMDA Receptor-mediated Ca2+ Influx in the Absence of Mg2+ Block Disrupts Rest: Activity Rhythms in Drosophila. Sleep 2018; 40:4330652. [PMID: 29029290 DOI: 10.1093/sleep/zsx166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Study Objectives The correlated activation of pre- and postsynaptic neurons is essential for the NMDA receptor-mediated Ca2+ influx by removing Mg2+ from block site and NMDA receptors have been implicated in phase resetting of circadian clocks. So we assessed rest:activity rhythms in Mg2+ block defective animals. Methods Using Drosophila locomotor monitoring system, we checked circadian rest:activity rhythms of different mutants under constant darkness (DD) and light:dark (LD) conditions. We recorded NMDA receptor-mediated currents or Ca2+ increase in neurons using patch-clamp and Ca2+ imaging techniques. Results We found that Mg2+ block defective mutant flies were completely arrhythmic under DD. To further understand the role of Mg2+ block in daily circadian rest:activity, we observed the mutant files under LD cycles, and we found severely reduced morning anticipation and advanced evening peak compared to control flies. We also used tissue-specific expression of Mg2+ block defective NMDA receptors and demonstrated pigment-dispersing factor receptor (PDFR)-expressing circadian neurons were implicated in mediating the circadian rest:activity deficits. Endogenous functional NMDA receptors are expressed in most Drosophila neurons, including in a subgroup of dorsal neurons (DN1s). Subsequently, we determined that the uncorrelated extra Ca2+ influx may act in part through Ca2+/Calmodulin (CaM)-stimulated PDE1c pathway leading to morning behavior phenotypes. Conclusions These results demonstrate that Mg2+ block of NMDA receptors at resting potential is essential for the daily circadian rest:activity rhythms and we propose that Mg2+ block functions to suppress CaM-stimulated PDE1c activation at resting potential, thus regulating Ca2+ and cyclic AMP oscillations in circadian and sleep circuits.
Collapse
Affiliation(s)
- Qian Song
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Ge Feng
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaxing Zhang
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Xuechun Xia
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Min Ji
- Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Lei Lv
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Yong Ping
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders (No.13dz2260500), Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
52
|
Yamada Y, Prosser RA. Copper in the suprachiasmatic circadian clock: A possible link between multiple circadian oscillators. Eur J Neurosci 2018; 51:47-70. [PMID: 30269387 DOI: 10.1111/ejn.14181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 09/05/2018] [Accepted: 09/17/2018] [Indexed: 01/07/2023]
Abstract
The mammalian circadian clock in the suprachiasmatic nucleus (SCN) is very robust, able to coordinate our daily physiological and behavioral rhythms with exquisite accuracy. Simultaneously, the SCN clock is highly sensitive to environmental timing cues such as the solar cycle. This duality of resiliency and sensitivity may be sustained in part by a complex intertwining of three cellular oscillators: transcription/translation, metabolic/redox, and membrane excitability. We suggest here that one of the links connecting these oscillators may be forged from copper (Cu). Cellular Cu levels are highly regulated in the brain and peripherally, and Cu affects cellular metabolism, redox state, cell signaling, and transcription. We have shown that both Cu chelation and application induce nighttime phase shifts of the SCN clock in vitro and that these treatments affect glutamate, N-methyl-D-aspartate receptor, and associated signaling processes differently. More recently we found that Cu induces mitogen-activated protein kinase-dependent phase shifts, while the mechanisms by which Cu removal induces phase shifts remain unclear. Lastly, we have found that two Cu transporters are expressed in the SCN, and that one of these transporters (ATP7A) exhibits a day/night rhythm. Our results suggest that Cu homeostasis is tightly regulated in the SCN, and that changes in Cu levels may serve as a time cue for the circadian clock. We discuss these findings in light of the existing literature and current models of multiple coupled circadian oscillators in the SCN.
Collapse
Affiliation(s)
- Yukihiro Yamada
- Department of Biochemistry & Cellular and Molecular Biology, NeuroNET Research Center, University of Tennessee, Knoxville, Tennessee
| | - Rebecca A Prosser
- Department of Biochemistry & Cellular and Molecular Biology, NeuroNET Research Center, University of Tennessee, Knoxville, Tennessee
| |
Collapse
|
53
|
Abstract
Background Several immunological functions are dependent on circadian rhythms. However, there are still relatively few studies about circadian rhythms in neuromyelitis optica spectrum disorders (NMOSD) and 2D2 transgenic mice. We explore whether 2D2 mice have abnormalities in circadian rhythms and the potential underlying molecular mechanism. Material/Methods We first observed the wheel-running motion of the control and 2D2 mice using wheel-running measurements. The cytokine levels were also analyzed using enzyme-linked immunosorbent assay (ELISA), and the results of clock gene expressions in the suprachiasmatic nucleus (SCN) were investigated using real-time polymerase chain reaction (real-time PCR). Results The wheel-running rhythm in 2D2 mice differed from that of the controls. The TNF-α and IL-10 rhythms were disrupted in 2D2 mice. Additionally, the rhythm of the clock genes, Per1 and Per2, and expression in the SCN of 2D2 mice were also changed. Conclusions The results presented here indicate that alteration of circadian rhythms in 2D2 mice affects behavior and immune function, and the potential molecular mechanism might be the Per1 and Per2 expression disorders in the SCN. 2D2 mice might be a suitable model for studying circadian disruption in NMOSD.
Collapse
Affiliation(s)
- Huiru Xue
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, China (mainland).,Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Xiuli Cao
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Meini Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| |
Collapse
|
54
|
Yang M, Ren B, Qiao L, Ren B, Hu Y, Zhao R, Ren Z, Du J. Behavior responses of zebrafish (Danio rerio) to aquatic environmental stresses in the characteristic of circadian rhythms. CHEMOSPHERE 2018; 210:129-138. [PMID: 29986218 DOI: 10.1016/j.chemosphere.2018.07.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/03/2018] [Accepted: 07/03/2018] [Indexed: 06/08/2023]
Abstract
As behavior shows a distinct circadian rhythm, it is hypothesized that circadian rhythms based on zebrafish (Danio rerio) behavior responses could be affected by contaminants in this study, and then the behavior strength of zebrafish exposed to 0.005 mg/L Cadmium chloride (CdCl2), 0.01 mg/L Dibasic Sodium Phosphate (Na2HPO4), 0.002 mg/L deltamethrin, and 0.003 mg/L atrazine for 6 days is used to illustrate the possibility of behavior circadian rhythms as an indicator in the environmental stress assessment. Statistical analysis with p < 0.01 shows that a clear difference between average values of BS during dark period (AVD) and those during light period (AVL) could be observed, and 24 h circadian rhythms do exist in zebrafish behavior responses. Both BS values and circadian rhythms of zebrafish can be affected in the aspect of periodicity with clear time delay, which were 1 h delay in CdCl2, 4 h delay in Na2HPO4, 4 h delay in deltamethrin, and 1 h delay in atrazine. Behavior circadian rhythms were disturbed according to the repetitive cycles after autocorrelation analysis, and the toxic effects of different chemicals could be reflected by the profiles of the Self-Organizing Map (SOM), which indicated the circadian rhythm disorder in different degrees. These results deduced from the statistical analysis, autocorrelation and SOM strongly supported that circadian rhythms based on zebrafish BS could be used as an indicator in the environmental stress assessment.
Collapse
Affiliation(s)
- Meiyi Yang
- Institute of Environment and Ecology, Shandong Normal University, Ji'nan 250014, Shandong, China
| | - Baigang Ren
- Institute of Environment and Ecology, Shandong Normal University, Ji'nan 250014, Shandong, China; School of Physics and Electronic Science, Shandong Normal University, Ji'nan 250014, Shandong, China
| | - Linlin Qiao
- Institute of Environment and Ecology, Shandong Normal University, Ji'nan 250014, Shandong, China
| | - Baixiang Ren
- Institute of Environment and Ecology, Shandong Normal University, Ji'nan 250014, Shandong, China
| | - Yongyuan Hu
- Institute of Environment and Ecology, Shandong Normal University, Ji'nan 250014, Shandong, China
| | - Ruibin Zhao
- Institute of Environment and Ecology, Shandong Normal University, Ji'nan 250014, Shandong, China
| | - Zongming Ren
- Institute of Environment and Ecology, Shandong Normal University, Ji'nan 250014, Shandong, China.
| | - Jun Du
- School of Physics and Electronic Science, Shandong Normal University, Ji'nan 250014, Shandong, China
| |
Collapse
|
55
|
Atkins N, Ren S, Hatcher N, Burgoon PW, Mitchell JW, Sweedler JV, Gillette MU. Functional Peptidomics: Stimulus- and Time-of-Day-Specific Peptide Release in the Mammalian Circadian Clock. ACS Chem Neurosci 2018; 9:2001-2008. [PMID: 29901982 DOI: 10.1021/acschemneuro.8b00089] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Daily oscillations of brain and body states are under complex temporal modulation by environmental light and the hypothalamic suprachiasmatic nucleus (SCN), the master circadian clock. To better understand mediators of differential temporal modulation, we characterize neuropeptide releasate profiles by nonselective capture of secreted neuropeptides in an optic nerve horizontal SCN brain slice model. Releasates are collected following electrophysiological stimulation of the optic nerve/retinohypothalamic tract under conditions that alter the phase of the SCN activity state. Secreted neuropeptides are identified by intact mass via matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). We found time-of-day-specific suites of peptides released downstream of optic nerve stimulation. Peptide release was modified differentially with respect to time-of-day by stimulus parameters and by inhibitors of glutamatergic or PACAPergic neurotransmission. The results suggest that SCN physiology is modulated by differential peptide release of both known and unexpected peptides that communicate time-of-day-specific photic signals via previously unreported neuropeptide signatures.
Collapse
|
56
|
SCN VIP Neurons Are Essential for Normal Light-Mediated Resetting of the Circadian System. J Neurosci 2018; 38:7986-7995. [PMID: 30082421 DOI: 10.1523/jneurosci.1322-18.2018] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/10/2018] [Accepted: 07/14/2018] [Indexed: 12/21/2022] Open
Abstract
The suprachiasmatic nucleus (SCN) synchronizes circadian rhythms in behavior and physiology to the external light cycle, but the mechanisms by which this occurs are unclear. As the neuropeptide vasoactive intestinal peptide (VIP) is important for circadian light responses, we tested the hypothesis that rhythmic VIP-producing SCN neurons mediate circadian light responses in male and female mice. Using in vivo fiber photometry over multiple days, we found daily rhythms in spontaneous calcium events of SCN VIP neurons that peaked during the subjective day and were disrupted by constant light. The light-evoked calcium responses peaked around subjective dusk and were greater during the subjective night. Using novel VIP sensor cells, we found that the activity patterns in SCN VIP neurons correlated tightly with spontaneous and NMDA-evoked VIP release. Finally, in vivo hyperpolarization of VIP neurons attenuated light-induced shifts of daily rhythms in locomotion. We conclude that SCN VIP neurons exhibit circadian rhythms in spontaneous and light-responsive activity and are essential for the normal resetting of daily rhythms by environmental light.SIGNIFICANCE STATEMENT Daily rhythms in behavior and physiology, including sleep/wake and hormone release, are synchronized to local time by the master circadian pacemaker, the suprachiasmatic nucleus (SCN). The advent of artificial lighting and, consequently, light exposure at night, is associated with an increased risk of disease due to disrupted circadian rhythms. However, the mechanisms by which the SCN encodes normal and pathological light information are unclear. Here, we find that vasoactive intestinal peptide (VIP)-producing SCN neurons exhibit daily rhythms in neuronal activity and VIP release, and that blocking the activity of these neurons attenuates light-induced phase shifts. We conclude that rhythmic VIP neurons are an essential component of the circadian light transduction pathway.
Collapse
|
57
|
Pfeffer M, Zimmermann Z, Gispert S, Auburger G, Korf HW, von Gall C. Impaired Photic Entrainment of Spontaneous Locomotor Activity in Mice Overexpressing Human Mutant α-Synuclein. Int J Mol Sci 2018; 19:E1651. [PMID: 29865270 PMCID: PMC6032049 DOI: 10.3390/ijms19061651] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 01/17/2023] Open
Abstract
Parkinson's disease (PD) is characterized by distinct motor and non-motor symptoms. Sleep disorders are the most frequent and challenging non-motor symptoms in PD patients, and there is growing evidence that they are a consequence of disruptions within the circadian system. PD is characterized by a progressive degeneration of the dorsal vagal nucleus and midbrain dopaminergic neurons together with an imbalance of many other neurotransmitters. Mutations in α-synuclein (SNCA), a protein modulating SNARE complex-dependent neurotransmission, trigger dominantly inherited PD variants and sporadic cases of PD. The A53T SNCA missense mutation is associated with an autosomal dominant early-onset familial PD. To test whether this missense mutation affects the circadian system, we analyzed the spontaneous locomotor behavior of non-transgenic wildtype mice and transgenic mice overexpressing mutant human A53T α-synuclein (A53T). The mice were subjected to entrained- and free-running conditions as well as to experimental jet lag. Furthermore, the vesicular glutamate transporter 2 (VGLUT2) in the suprachiasmatic nucleus (SCN) was analyzed by immunohistochemistry. Free-running circadian rhythm and, thus, circadian rhythm generation, were not affected in A53T mice. A53T mice entrained to the light⁻dark cycle, however, with an advanced phase angle of 2.65 ± 0.5 h before lights off. Moreover, re-entrainment after experimental jet lag was impaired in A53T mice. Finally, VGLUT2 immunoreaction was reduced in the SCN of A53T mice. These data suggest an impaired light entrainment of the circadian system in A53T mice.
Collapse
Affiliation(s)
- Martina Pfeffer
- Institut für Anatomie II, Fachbereich Medizin, Heinrich Heine Universität, Universitätsstr. 1, D-40225 Düsseldorf, Germany.
| | - Zuzana Zimmermann
- Dr. Senckenbergische Anatomie II, Fachbereich Medizin, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany.
| | - Suzana Gispert
- Experimental Neurology, Department of Neurology, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany.
| | - Georg Auburger
- Experimental Neurology, Department of Neurology, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany.
| | - Horst-Werner Korf
- Institut für Anatomie I, Fachbereich Medizin, Heinrich Heine Universität, Universitätsstr. 1, D-40225 Düsseldorf, Germany.
| | - Charlotte von Gall
- Institut für Anatomie II, Fachbereich Medizin, Heinrich Heine Universität, Universitätsstr. 1, D-40225 Düsseldorf, Germany.
| |
Collapse
|
58
|
Cooper JM, Halter KA, Prosser RA. Circadian rhythm and sleep-wake systems share the dynamic extracellular synaptic milieu. Neurobiol Sleep Circadian Rhythms 2018; 5:15-36. [PMID: 31236509 PMCID: PMC6584685 DOI: 10.1016/j.nbscr.2018.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/06/2018] [Accepted: 04/10/2018] [Indexed: 01/23/2023] Open
Abstract
The mammalian circadian and sleep-wake systems are closely aligned through their coordinated regulation of daily activity patterns. Although they differ in their anatomical organization and physiological processes, they utilize overlapping regulatory mechanisms that include an assortment of proteins and molecules interacting within the extracellular space. These extracellular factors include proteases that interact with soluble proteins, membrane-attached receptors and the extracellular matrix; and cell adhesion molecules that can form complex scaffolds connecting adjacent neurons, astrocytes and their respective intracellular cytoskeletal elements. Astrocytes also participate in the dynamic regulation of both systems through modulating neuronal appositions, the extracellular space and/or through release of gliotransmitters that can further contribute to the extracellular signaling processes. Together, these extracellular elements create a system that integrates rapid neurotransmitter signaling across longer time scales and thereby adjust neuronal signaling to reflect the daily fluctuations fundamental to both systems. Here we review what is known about these extracellular processes, focusing specifically on areas of overlap between the two systems. We also highlight questions that still need to be addressed. Although we know many of the extracellular players, far more research is needed to understand the mechanisms through which they modulate the circadian and sleep-wake systems.
Collapse
Key Words
- ADAM, A disintegrin and metalloproteinase
- AMPAR, AMPA receptor
- Astrocytes
- BDNF, brain-derived neurotrophic factor
- BMAL1, Brain and muscle Arnt-like-1 protein
- Bmal1, Brain and muscle Arnt-like-1 gene
- CAM, cell adhesion molecules
- CRY, cryptochrome protein
- Cell adhesion molecules
- Circadian rhythms
- Cry, cryptochrome gene
- DD, dark-dark
- ECM, extracellular matrix
- ECS, extracellular space
- EEG, electroencephalogram
- Endo N, endoneuraminidase N
- Extracellular proteases
- GFAP, glial fibrillary acidic protein
- IL, interleukin
- Ig, immunoglobulin
- LC, locus coeruleus
- LD, light-dark
- LH, lateral hypothalamus
- LRP-1, low density lipoprotein receptor-related protein 1
- LTP, long-term potentiation
- MMP, matrix metalloproteinases
- NCAM, neural cell adhesion molecule protein
- NMDAR, NMDA receptor
- NO, nitric oxide
- NST, nucleus of the solitary tract
- Ncam, neural cell adhesion molecule gene
- Nrl, neuroligin gene
- Nrx, neurexin gene
- P2, purine type 2 receptor
- PAI-1, plasminogen activator inhibitor-1
- PER, period protein
- PPT, peduculopontine tegmental nucleus
- PSA, polysialic acid
- Per, period gene
- REMS, rapid eye movement sleep
- RSD, REM sleep disruption
- SCN, suprachiasmatic nucleus
- SWS, slow wave sleep
- Sleep-wake system
- Suprachiasmatic nucleus
- TNF, tumor necrosis factor
- TTFL, transcriptional-translational negative feedback loop
- VIP, vasoactive intestinal polypeptide
- VLPO, ventrolateral preoptic
- VP, vasopressin
- VTA, ventral tegmental area
- dNlg4, drosophila neuroligin-4 gene
- nNOS, neuronal nitric oxide synthase gene
- nNOS, neuronal nitric oxide synthase protein
- tPA, tissue-type plasminogen activator
- uPA, urokinase-type plasminogen activator
- uPAR, uPA receptor
Collapse
|
59
|
Deboer T. Sleep homeostasis and the circadian clock: Do the circadian pacemaker and the sleep homeostat influence each other's functioning? Neurobiol Sleep Circadian Rhythms 2018; 5:68-77. [PMID: 31236513 PMCID: PMC6584681 DOI: 10.1016/j.nbscr.2018.02.003] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 02/23/2018] [Accepted: 02/23/2018] [Indexed: 12/13/2022] Open
Abstract
Sleep is regulated by a homeostatic and a circadian process. Together these two processes determine most aspects of sleep and related variables like sleepiness and alertness. The two processes are known to be able to work independently, but also to both influence sleep and sleep related variables in an additive or more complex manner. The question remains whether the two processes are directly influencing each other. The present review summarizes evidence from behavioural and electroencephalographic determined sleep, electrophysiology, gene knock out mouse models, and mathematical modelling to explore whether sleep homeostasis can influence circadian clock functioning and vice versa. There is a multitude of data available showing parallel action or influence of sleep homeostatic mechanisms and the circadian clock on several objective and subjective variables related to sleep and alertness. However, the evidence of a direct influence of the circadian clock on sleep homeostatic mechanisms is sparse and more research is needed, particularly applying longer sleep deprivations that include a second night. The strongest evidence of an influence of sleep homeostatic mechanisms on clock functioning comes from sleep deprivation experiments, demonstrating an attenuation of phase shifts of the circadian rhythm to light pulses when sleep homeostatic pressure is increased. The data suggest that the circadian clock is less susceptible to light when sleep pressure is high. The available data indicate that a strong central clock will induce periods of deep sleep, which in turn will strengthen clock function. Both are therefore important for health and wellbeing. Weakening of one will also hamper functioning of the other. Shift work and jet lag are situations where one tries to adapt to zeitgebers in a condition where sleep is compromised. Adaptation to zeitgebers may be improved by introducing nap schedules to reduce sleep pressure, and through that increasing clock susceptibility to light.
Collapse
|
60
|
Photoperiod-Induced Neuroplasticity in the Circadian System. Neural Plast 2018; 2018:5147585. [PMID: 29681926 PMCID: PMC5851158 DOI: 10.1155/2018/5147585] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 01/11/2018] [Indexed: 01/01/2023] Open
Abstract
Seasonal changes in light exposure have profound effects on behavioral and physiological functions in many species, including effects on mood and cognitive function in humans. The mammalian brain's master circadian clock, the suprachiasmatic nucleus (SCN), transmits information about external light conditions to other brain regions, including some implicated in mood and cognition. Although the detailed mechanisms are not yet known, the SCN undergoes highly plastic changes at the cellular and network levels under different light conditions. We therefore propose that the SCN may be an essential mediator of the effects of seasonal changes of day length on mental health. In this review, we explore various forms of neuroplasticity that occur in the SCN and other brain regions to facilitate seasonal adaptation, particularly altered phase distribution of cellular circadian oscillators in the SCN and changes in hypothalamic neurotransmitter expression.
Collapse
|
61
|
Biello SM, Bonsall DR, Atkinson LA, Molyneux PC, Harrington ME, Lall GS. Alterations in glutamatergic signaling contribute to the decline of circadian photoentrainment in aged mice. Neurobiol Aging 2018; 66:75-84. [PMID: 29547750 DOI: 10.1016/j.neurobiolaging.2018.02.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/06/2018] [Accepted: 02/13/2018] [Indexed: 12/20/2022]
Abstract
Robust physiological circadian rhythms form an integral part of well-being. The aging process has been found to negatively impact systems that drive circadian physiology, typically manifesting as symptoms associated with abnormal/disrupted sleeping patterns. Here, we investigated the age-related decline in light-driven circadian entrainment in male C57BL/6J mice. We compared light-driven resetting of circadian behavioral activity in young (1-2 months) and old (14-18 months) mice and explored alterations in the glutamatergic pathway at the level of the circadian pacemaker, the suprachiasmatic nucleus (SCN). Aged animals showed a significant reduction in sensitivity to behavioral phase resetting by light. We show that this change was through alterations in N-Methyl-D-aspartate (NMDA) signaling at the SCN, where NMDA, a glutamatergic agonist, was less potent in inducing clock resetting. Finally, we show that this shift in NMDA sensitivity was through the reduced SCN expression of this receptor's NR2B subunit. Only in young animals did an NR2B antagonist attenuate behavioral resetting. These results can help target treatments that aim to improve both physiological and behavioral circadian entrainment in aged populations.
Collapse
Affiliation(s)
| | - David R Bonsall
- Medway School of Pharmacy, University of Kent, Chatham, UK; Neuroscience Program, Smith College, Northampton, MA, USA
| | | | | | | | - Gurprit S Lall
- Medway School of Pharmacy, University of Kent, Chatham, UK.
| |
Collapse
|
62
|
Fisk AS, Tam SKE, Brown LA, Vyazovskiy VV, Bannerman DM, Peirson SN. Light and Cognition: Roles for Circadian Rhythms, Sleep, and Arousal. Front Neurol 2018; 9:56. [PMID: 29479335 PMCID: PMC5811463 DOI: 10.3389/fneur.2018.00056] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/22/2018] [Indexed: 01/12/2023] Open
Abstract
Light exerts a wide range of effects on mammalian physiology and behavior. As well as synchronizing circadian rhythms to the external environment, light has been shown to modulate autonomic and neuroendocrine responses as well as regulating sleep and influencing cognitive processes such as attention, arousal, and performance. The last two decades have seen major advances in our understanding of the retinal photoreceptors that mediate these non-image forming responses to light, as well as the neural pathways and molecular mechanisms by which circadian rhythms are generated and entrained to the external light/dark (LD) cycle. By contrast, our understanding of the mechanisms by which lighting influences cognitive processes is more equivocal. The effects of light on different cognitive processes are complex. As well as the direct effects of light on alertness, indirect effects may also occur due to disrupted circadian entrainment. Despite the widespread use of disrupted LD cycles to study the role circadian rhythms on cognition, the different experimental protocols used have subtly different effects on circadian function which are not always comparable. Moreover, these protocols will also disrupt sleep and alter physiological arousal, both of which are known to modulate cognition. Studies have used different assays that are dependent on different cognitive and sensory processes, which may also contribute to their variable findings. Here, we propose that studies addressing the effects of different lighting conditions on cognitive processes must also account for their effects on circadian rhythms, sleep, and arousal if we are to fully understand the physiological basis of these responses.
Collapse
Affiliation(s)
- Angus S Fisk
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Shu K E Tam
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Laurence A Brown
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Vladyslav V Vyazovskiy
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Stuart N Peirson
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
63
|
Abraham U, Schlichting JK, Kramer A, Herzel H. Quantitative analysis of circadian single cell oscillations in response to temperature. PLoS One 2018; 13:e0190004. [PMID: 29293562 PMCID: PMC5749732 DOI: 10.1371/journal.pone.0190004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 12/06/2017] [Indexed: 11/18/2022] Open
Abstract
Body temperature rhythms synchronize circadian oscillations in different tissues, depending on the degree of cellular coupling: the responsiveness to temperature is higher when single circadian oscillators are uncoupled. So far, the role of coupling in temperature responsiveness has only been studied in organotypic tissue slices of the central circadian pacemaker, because it has been assumed that peripheral target organs behave like uncoupled multicellular oscillators. Since recent studies indicate that some peripheral tissues may exhibit cellular coupling as well, we asked whether peripheral network dynamics also influence temperature responsiveness. Using a novel technique for long-term, high-resolution bioluminescence imaging of primary cultured cells, exposed to repeated temperature cycles, we were able to quantitatively measure period, phase, and amplitude of central (suprachiasmatic nuclei neuron dispersals) and peripheral (mouse ear fibroblasts) single cell oscillations in response to temperature. Employing temperature cycles of different lengths, and different cell densities, we found that some circadian characteristics appear cell-autonomous, e.g. period responses, while others seem to depend on the quality/degree of cellular communication, e.g. phase relationships, robustness of the oscillation, and amplitude. Overall, our findings indicate a strong dependence on the cell's ability for intercellular communication, which is not only true for neuronal pacemakers, but, importantly, also for cells in peripheral tissues. Hence, they stress the importance of comparative studies that evaluate the degree of coupling in a given tissue, before it may be used effectively as a target for meaningful circadian manipulation.
Collapse
Affiliation(s)
- Ute Abraham
- Laboratory of Chronobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Achim Kramer
- Laboratory of Chronobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hanspeter Herzel
- Institute for Theoretical Biology, Humboldt-University, Berlin, Germany
| |
Collapse
|
64
|
Purnell BS, Hajek MA, Buchanan GF. Time-of-day influences on respiratory sequelae following maximal electroshock-induced seizures in mice. J Neurophysiol 2017; 118:2592-2600. [PMID: 28794189 PMCID: PMC5668461 DOI: 10.1152/jn.00039.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 08/07/2017] [Accepted: 08/08/2017] [Indexed: 11/22/2022] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is the leading cause of death in refractory epilepsy patients. Although specific mechanisms underlying SUDEP are not well understood, evidence suggests most SUDEP occurs due to seizure-induced respiratory arrest. SUDEP also tends to happen at night. Although this may be due to circumstances in which humans find themselves at night, such as being alone without supervision or sleeping prone, or to independent influences of sleep state, there are a number of reasons why the night (i.e., circadian influences) could be an independent risk factor for SUDEP. We explored this possibility. Adult male WT mice were instrumented for EEG, EMG, and EKG recording and subjected to maximal electroshock (MES) seizures during wakefulness, non-rapid eye movement (NREM) sleep, and rapid eye movement (REM) sleep during the nighttime/dark phase. These data were compared with data collected following seizures induced during the daytime/light phase. Seizures induced during the nighttime were similar in severity and duration to those induced during the daytime; however, seizures induced during the nighttime were associated with a lesser degree of respiratory dysregulation and postictal EEG suppression. Seizures induced during REM sleep during the nighttime were universally fatal, as is seen when seizures are induced during REM during the daytime. Taken together, these data implicate a role for time of day in influencing the physiological consequences of seizures that may contribute to seizure-induced death.NEW & NOTEWORTHY Sudden unexpected death in epilepsy (SUDEP) is the leading cause of death in patients with refractory epilepsy. SUDEP frequently occurs during the night, which has been attributed to an effect of sleep. We have shown that sleep state does indeed influence survival following a seizure. That SUDEP occurs during the night could also implicate a circadian influence. In this study we found that time of day independently affects the physiological consequences of seizures.
Collapse
Affiliation(s)
- Benton S Purnell
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Michael A Hajek
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Gordon F Buchanan
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa Carver College of Medicine, Iowa City, Iowa;
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, Iowa; and
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
65
|
Abstract
The small GTPase Ras is a universal eukaryotic cytoplasmic membrane-anchored protein, which regulates diverse downstream signal transduction pathways that play an important role in the proper functioning of neurons. Ras activity is a central regulator of structural and functional synaptic plasticity in the adult nervous system, where it channels neuronal responses to various extracellular cues allowing the organism to adapt to complex environmental stimuli. The suprachiasmatic nucleus (SCN) is the principle pacemaker of the circadian clock, and the circadian and photic regulation of Ras activity in the SCN is an important modulator of the clockwork. We have generated transgenic mouse expressing constitutively active V12-H-Ras selectively in neurons via a synapsin I promoter (synRas mice), which serves as a suitable model to study the role of neuronal Ras signaling. Modulation of Ras activity affects ERK1,2/CREB signaling and glycogen synthase kinase-3 beta expression in the SCN, which in turn modify the photoentrainment of the clock and the fine tuning the circadian period length. The main focus of this review is to offer an overview of the function of Ras signaling in the circadian rhythm and its potential role in learning and memory consolidation.
Collapse
Affiliation(s)
- Tsvetan Serchov
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Rolf Heumann
- Biochemistry II, Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
66
|
Lommen J, Stahr A, Ingenwerth M, Ali AAH, von Gall C. Time-of-day-dependent expression of purinergic receptors in mouse suprachiasmatic nucleus. Cell Tissue Res 2017; 369:579-590. [PMID: 28547658 PMCID: PMC5579179 DOI: 10.1007/s00441-017-2634-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/26/2017] [Indexed: 12/22/2022]
Abstract
Purinergic P2X and P2Y receptors are involved in mediating intercellular signalling via purines such as adenosine triphosphate (ATP). P2X and P2Y receptors have been implicated in numerous body functions including learning, memory and sleep. All of these body functions show time-of–day-dependent variations controlled by the master circadian oscillator located in the suprachiasmatic nucleus (SCN). Evidence exists for a role of purinergic signalling in intercellular coupling within SCN. However, few studies have been performed on the expression of purinergic receptors in SCN. Therefore, we analyse the expression of seven P2X (P2X1–7) and eight P2Y (P2Y1–2, 4, 6, 11–14) receptors in mouse SCN and address their time-of-day-dependent variation by using immunohistochemistry and real-time polymerase chain reaction. At the early light phase, P2X and P2Y receptors show a low to moderate, homogenously distributed immunoreaction throughout SCN. P2Y13 reveals strong immunoreaction in fibres within the core region of SCN. From the fifteen analysed P2 receptors, seven exhibit a time-of-day-dependent variation in SCN. P2X1 immunoreaction is very low in the early light phase with a minor increase at the end of the dark phase. P2X4 immunoreaction strongly increases during the dark phase in soma cells in the core region and in a dense network of fibres in the shell region of SCN. P2X3 immunoreaction is moderately elevated during the dark phase. Conversely, immunoreaction for P2Y2, P2Y12 and P2Y14 moderately increases at the early light phase and P2Y6 immunoreaction displays a moderate increase at the mid-light phase. Thus, this study demonstrates a time-of-day-dependent variation of P2 receptors in mouse SCN.
Collapse
Affiliation(s)
- Julian Lommen
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Anna Stahr
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Marc Ingenwerth
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany.,Institute of Pathology, University of Duisburg-Essen, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Amira A H Ali
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
67
|
Tsuji T, Allchorne AJ, Zhang M, Tsuji C, Tobin VA, Pineda R, Raftogianni A, Stern JE, Grinevich V, Leng G, Ludwig M. Vasopressin casts light on the suprachiasmatic nucleus. J Physiol 2017; 595:3497-3514. [PMID: 28402052 DOI: 10.1113/jp274025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 02/21/2017] [Indexed: 12/31/2022] Open
Abstract
KEY POINTS A subpopulation of retinal ganglion cells expresses the neuropeptide vasopressin. These retinal ganglion cells project predominately to our biological clock, the suprachiasmatic nucleus (SCN). Light-induced vasopressin release enhances the responses of SCN neurons to light. It also enhances expression of genes involved in photo-entrainment of biological rhythms. ABSTRACT In all animals, the transition between night and day engages a host of physiological and behavioural rhythms. These rhythms depend not on the rods and cones of the retina, but on retinal ganglion cells (RGCs) that detect the ambient light level in the environment. These project to the suprachiasmatic nucleus (SCN) of the hypothalamus to entrain circadian rhythms that are generated within the SCN. The neuropeptide vasopressin has an important role in this entrainment. Many SCN neurons express vasopressin, and it has been assumed that the role of vasopressin in the SCN reflects the activity of these cells. Here we show that vasopressin is also expressed in many retinal cells that project to the SCN. Light-evoked vasopressin release contributes to the responses of SCN neurons to light, and enhances expression of the immediate early gene c-fos in the SCN, which is involved in photic entrainment of circadian rhythms.
Collapse
Affiliation(s)
- Takahiro Tsuji
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - Andrew J Allchorne
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - Meng Zhang
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - Chiharu Tsuji
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - Vicky A Tobin
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - Rafael Pineda
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - Androniki Raftogianni
- Schaller Research Group on Neuropeptides, German Cancer Research Centre DKFZ, Central Institute of Mental Health, and University of Heidelberg, Heidelberg, Germany
| | - Javier E Stern
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - Valery Grinevich
- Schaller Research Group on Neuropeptides, German Cancer Research Centre DKFZ, Central Institute of Mental Health, and University of Heidelberg, Heidelberg, Germany
| | - Gareth Leng
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - Mike Ludwig
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
68
|
Barca-Mayo O, Pons-Espinal M, Follert P, Armirotti A, Berdondini L, De Pietri Tonelli D. Astrocyte deletion of Bmal1 alters daily locomotor activity and cognitive functions via GABA signalling. Nat Commun 2017; 8:14336. [PMID: 28186121 PMCID: PMC5309809 DOI: 10.1038/ncomms14336] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 12/19/2016] [Indexed: 02/08/2023] Open
Abstract
Circadian rhythms are controlled by a network of clock neurons in the central pacemaker, the suprachiasmatic nucleus (SCN). Core clock genes, such as Bmal1, are expressed in SCN neurons and in other brain cells, such as astrocytes. However, the role of astrocytic clock genes in controlling rhythmic behaviour is unknown. Here we show that ablation of Bmal1 in GLAST-positive astrocytes alters circadian locomotor behaviour and cognition in mice. Specifically, deletion of astrocytic Bmal1 has an impact on the neuronal clock through GABA signalling. Importantly, pharmacological modulation of GABAA-receptor signalling completely rescues the behavioural phenotypes. Our results reveal a crucial role of astrocytic Bmal1 for the coordination of neuronal clocks and propose a new cellular target, astrocytes, for neuropharmacology of transient or chronic perturbation of circadian rhythms, where alteration of astrocytic clock genes might contribute to the impairment of the neurobehavioural outputs such as cognition.
Collapse
Affiliation(s)
- Olga Barca-Mayo
- Neurobiology of miRNA Lab, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
- NetS3 Lab, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Meritxell Pons-Espinal
- Neurobiology of miRNA Lab, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Philipp Follert
- Neurobiology of miRNA Lab, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Andrea Armirotti
- D3 PharmaChemistry, Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Luca Berdondini
- NetS3 Lab, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Davide De Pietri Tonelli
- Neurobiology of miRNA Lab, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| |
Collapse
|
69
|
Abstract
Daily activity rhythms that are dominated by internal clocks are called circadian rhythms. A central clock is located in the suprachiasmatic nucleus of the hypothalamus, and peripheral clocks are located in most mammalian peripheral cells. The central clock is entrained by light/dark cycles, whereas peripheral clocks are entrained by feeding cycles. The effects of nutrients on the central and peripheral clocks have been investigated during the past decade and much interaction between them has come to light. For example, a high-fat diet prolongs the period of circadian behavior, a ketogenic diet advances the onset of locomotor activity rhythms, and a high-salt diet advances the phase of peripheral molecular clocks. Moreover, some food factors such as caffeine, nobiletin, and resveratrol, alter molecular and/or behavioral circadian rhythms. Here, we review nutrients and food factors that modulate mammalian circadian clocks from the cellular to the behavioral level.
Collapse
Affiliation(s)
- Hideaki Oike
- a Food Research Institute, National Agriculture and Food Research Organization , Tsukuba , Japan
| |
Collapse
|
70
|
Cooper JM, Rastogi A, Krizo JA, Mintz EM, Prosser RA. Urokinase-type plasminogen activator modulates mammalian circadian clock phase regulation in tissue-type plasminogen activator knockout mice. Eur J Neurosci 2017; 45:805-815. [PMID: 27992087 DOI: 10.1111/ejn.13511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/12/2016] [Accepted: 12/14/2016] [Indexed: 12/20/2022]
Abstract
Glutamate phase shifts the circadian clock in the mammalian suprachiasmatic nucleus (SCN) by activating NMDA receptors. Tissue-type plasminogen activator (tPA) gates phase shifts by activating plasmin to generate m(ature) BDNF, which binds TrkB receptors allowing clock phase shifts. Here, we investigate phase shifting in tPA knockout (tPA-/- ; B6.129S2-Plattm1Mlg /J) mice, and identify urokinase-type plasminogen activator (uPA) as an additional circadian clock regulator. Behavioral activity rhythms in tPA-/- mice entrain to a light-dark (LD) cycle and phase shift in response to nocturnal light pulses with no apparent loss in sensitivity. When the LD cycle is inverted, tPA-/- mice take significantly longer to entrain than C57BL/6J wild-type (WT) mice. SCN brain slices from tPA-/- mice exhibit entrained neuronal activity rhythms and phase shift in response to nocturnal glutamate with no change in dose-dependency. Pre-treating slices with the tPA/uPA inhibitor, plasminogen activator inhibitor-1 (PAI-1), inhibits glutamate-induced phase delays in tPA-/- slices. Selective inhibition of uPA with UK122 prevents glutamate-induced phase resetting in tPA-/- but not WT SCN slices. tPA expression is higher at night than the day in WT SCN, while uPA expression remains constant in WT and tPA-/- slices. Casein-plasminogen zymography reveals that neither tPA nor uPA total proteolytic activity is under circadian control in WT or tPA-/- SCN. Finally, tPA-/- SCN tissue has lower mBDNF levels than WT tissue, while UK122 does not affect mBDNF levels in either strain. Together, these results suggest that either tPA or uPA can support photic/glutamatergic phase shifts of the SCN circadian clock, possibly acting through distinct mechanisms.
Collapse
Affiliation(s)
- Joanna M Cooper
- Department of Biochemistry and Cellular and Molecular Biology, NeuroNET Research Center, M407 Walters Life Sciences, University of Tennessee, Knoxville, TN, 37996-0001, USA
| | - Ashutosh Rastogi
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Jessica A Krizo
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Eric M Mintz
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Rebecca A Prosser
- Department of Biochemistry and Cellular and Molecular Biology, NeuroNET Research Center, M407 Walters Life Sciences, University of Tennessee, Knoxville, TN, 37996-0001, USA
| |
Collapse
|
71
|
Albers HE, Walton JC, Gamble KL, McNeill JK, Hummer DL. The dynamics of GABA signaling: Revelations from the circadian pacemaker in the suprachiasmatic nucleus. Front Neuroendocrinol 2017; 44:35-82. [PMID: 27894927 PMCID: PMC5225159 DOI: 10.1016/j.yfrne.2016.11.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/16/2016] [Accepted: 11/22/2016] [Indexed: 12/31/2022]
Abstract
Virtually every neuron within the suprachiasmatic nucleus (SCN) communicates via GABAergic signaling. The extracellular levels of GABA within the SCN are determined by a complex interaction of synthesis and transport, as well as synaptic and non-synaptic release. The response to GABA is mediated by GABAA receptors that respond to both phasic and tonic GABA release and that can produce excitatory as well as inhibitory cellular responses. GABA also influences circadian control through the exclusively inhibitory effects of GABAB receptors. Both GABA and neuropeptide signaling occur within the SCN, although the functional consequences of the interactions of these signals are not well understood. This review considers the role of GABA in the circadian pacemaker, in the mechanisms responsible for the generation of circadian rhythms, in the ability of non-photic stimuli to reset the phase of the pacemaker, and in the ability of the day-night cycle to entrain the pacemaker.
Collapse
Affiliation(s)
- H Elliott Albers
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States.
| | - James C Walton
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - John K McNeill
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Daniel L Hummer
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Department of Psychology, Morehouse College, Atlanta, GA 30314, United States
| |
Collapse
|
72
|
Ruby CL, Palmer KN, Zhang J, Risinger MO, Butkowski MA, Swartzwelder HS. Differential Sensitivity to Ethanol-Induced Circadian Rhythm Disruption in Adolescent and Adult Mice. Alcohol Clin Exp Res 2016; 41:187-196. [PMID: 27997028 DOI: 10.1111/acer.13275] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/19/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND Growing evidence supports a central role for the circadian system in alcohol use disorders, but few studies have examined this relationship during adolescence. In mammals, circadian rhythms are regulated by the suprachiasmatic nucleus, a biological clock whose timing is synchronized (reset) to the environment primarily by light (photic) input. Alcohol (ethanol [EtOH]) disrupts circadian timing in part by attenuating photic phase-resetting responses in adult rodents. However, circadian rhythms change throughout life and it is not yet known whether EtOH has similar effects on circadian regulation during adolescence. METHODS General circadian locomotor activity was monitored in male C57BL6/J mice beginning in adolescence (P27) or adulthood (P61) in a 12-hour light, 12-hour dark photocycle for ~2 weeks to establish baseline circadian activity measures. On the day of the experiment, mice received an acute injection of EtOH (1.5 g/kg, i.p.) or equal volume saline 15 minutes prior to a 30-minute light pulse at Zeitgeber Time 14 (2 hours into the dark phase) and then were released into constant darkness (DD) for ~2 weeks to assess phase-resetting responses. Control mice of each age-group received injections but no light pulse prior to DD. RESULTS While adults showed the expected decrease in photic phase-delays induced by acute EtOH, this effect was absent in adolescent mice. Adolescents also showed baseline differences in circadian rhythmicity compared to adults, including advanced photocycle entrainment, larger photic phase-delays, a shorter free-running (endogenous) circadian period, and greater circadian rhythm amplitude. CONCLUSIONS Collectively, our results indicate that adolescent mice are less sensitive to the effect of EtOH on circadian photic phase-resetting and that their daily activity rhythms are markedly different than those of adults.
Collapse
Affiliation(s)
- Christina L Ruby
- Department of Biology, Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Kaitlyn N Palmer
- Department of Biology, Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Jiawen Zhang
- Department of Biology, Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Megan O Risinger
- Department of Biology, Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Melissa A Butkowski
- Department of Biology, Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - H Scott Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina.,Neurobiology Research Laboratory, VA Medical Center, Durham, North Carolina
| |
Collapse
|
73
|
Ibata Y, Tanaka M, Tamada Y, Hayashi S, Kawakami F, Takamatsu T, Hisa Y, Okamura H. REVIEW ■ : The Suprachiasmatic Nucleus: A Circadian Oscillator. Neuroscientist 2016. [DOI: 10.1177/107385849700300409] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The suprachiasmatic nucleus (SCN) is considered to be a circadian oscillator that regulates a set of phys iological aspects of behavior, including sleep-wakefulness and hormone release in mammalian species. In this review, we describe recent research that has begun to reveal the functional organization of the SCN. The SCN, which consists of a bilateral pair of tiny nuclei located just above the optic chiasm, contains several kinds of peptidergic neurons, but vasoactive intestinal peptide (VIP), arginine vasopressin (AVP), and somatostatin (SOM) neurons are the main components. VIP neurons and AVP neurons show distinctly different locations in the SCN; the former are found in the ventrolateral portion, whereas the latter are localized in the dorsomedial portion. VIP neurons receive all neuronal inputs from other regions of the CNS, such as those evoked by photic stimulation via the retinal ganglion cells and those relayed by 5HT inner vation from the raphe nuclei. VIP neurons relay their information to other kinds of neurons in the SCN, such as AVP and SOM neurons. VIP neurons, thus, may play a significant role in entrainment of circadian rhythm. VIP, AVP, SOM, and their mRNAs show rhythmic fluctuations that are predicted by this model; VIP and its mRNA show diurnal variation under the influence of photic stimulation, whereas AVP, SOM, and their mRNAs show endogenous rhythms. Immediate early genes (lEGs), such as c-fos mRNA, are also expressed in VIP neurons in the SCN, and IEG expression in the cells appears to be modified by photic stimuli. Together with transplantation studies showing that exogenous SCN tissue tends to restore circadian rhythm in arrhythmic animals, these results are beginning to clarify the function of the SCN in setting, maintaining, and resetting the biological clock. NEUROSCIENTIST 3:215-225, 1997
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hitoshi Okamura
- Department of Anatomy & Brain Science (HO) Kobe University School of Medicine Kusunokicho, Chuoku, Kobe
| |
Collapse
|
74
|
Abstract
The SCN of the mammalian hypothalamus comprises a self-sustained, biological clock that generates endogenous ca. 24-h (circadian) rhythms. Circadian rhythmicity in the SCN originates from the interaction of a defined set of “clock genes” that participate in transcription/translation feedback loops. In order for the SCN to serve as an internal clock that times an internal day corresponding to the external solar day, the intracellular molecular oscillations must be output as physiological signals and be reset by appropriate environmental inputs. Here, the authors consider the mechanisms by which the SCN circadian pacemaker encodes rhythmic output and light input. In particular, they focus on the ionic mechanisms by which SCN neurons encode clock gene output as circa-dian rhythms in spike frequency, as well as cellular and molecular mechanisms by which SCN neurons encode circadian light input through phase heterogeneity in the SCN network. The authors propose that there are 2 distinct classes of ionic mechanisms supporting spike frequency rhythms output—modulation of basal membrane potential and conductance versus modulation of spike production—whereas light input is transformed by cellular communication within the SCN network and encoded by the relative phase relationships among SCN neurons.
Collapse
Affiliation(s)
- Sandra J Kuhlman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | |
Collapse
|
75
|
Abstract
Drawing upon the premise that there is a parsimony to nature, the author proposes that heme moieties and bile pigments in animals mediate some non-visual influences of light upon neuroactive gases and biological rhythms. Primary abnormalities in synthesis and regulation of blood and bile may, therefore, cause some neuropsychiatric illnesses. Light may play a critical role in neurotransmission. NEUROSCIENTIST 2:207-210, 1996
Collapse
|
76
|
Abstract
The hypothalamic suprachiasmatic nucleus (SCN) has a pivotal role in the mammalian circadian clock. SCN neurons generate circadian rhythms in action potential firing frequencies and neurotransmitter release, and the core oscillation is thought to be driven by “clock gene” transcription-translation feedback loops. Cytosolic Ca2+mobilization followed by stimulation of various receptors has been shown to reset the gene transcription cycles in SCN neurons, whereas contribution of steady-state cytosolic Ca2+levels to the rhythm generation is unclear. Recently, circadian rhythms in cytosolic Ca2+levels have been demonstrated in cultured SCN neurons. The circadian Ca2+rhythms are driven by the release of Ca2+from ryanodine-sensitive internal stores and resistant to the blockade of action potentials. These results raise the possibility that gene translation/transcription loops may interact with autonomous Ca2+oscillations in the production of circadian rhythms in SCN neurons.
Collapse
Affiliation(s)
- Masayuki Ikeda
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Suita, Osaka, Japan.
| |
Collapse
|
77
|
Rebouças ECC, Leal S, Silva SM, Sá SI. Changes in the female arcuate nucleus morphology and neurochemistry after chronic ethanol consumption and long-term withdrawal. J Chem Neuroanat 2016; 77:30-40. [PMID: 27154870 DOI: 10.1016/j.jchemneu.2016.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/02/2016] [Accepted: 05/02/2016] [Indexed: 12/26/2022]
Abstract
Ethanol is a macronutrient whose intake is a form of ingestive behavior, sharing physiological mechanisms with food intake. Chronic ethanol consumption is detrimental to the brain, inducing gender-dependent neuronal damage. The hypothalamic arcuate nucleus (ARN) is a modulator of food intake that expresses feeding-regulatory neuropeptides, such as alpha melanocyte-stimulating hormone (α-MSH) and neuropeptide Y (NPY). Despite its involvement in pathways associated with eating disorders and ethanol abuse, the impact of ethanol consumption and withdrawal in the ARN structure and neurochemistry in females is unknown. We used female rat models of 20% ethanol consumption for six months and of subsequent ethanol withdrawal for two months. Food intake and body weights were measured. ARN morphology was stereologically analyzed to estimate its volume, total number of neurons and total number of neurons expressing NPY, α-MSH, tyrosine hydroxylase (TH) and estrogen receptor alpha (ERα). Ethanol decreased energy intake and body weights. However, it did not change the ARN morphology or the expression of NPY, α-MSH and TH, while increasing ERα expression. Withdrawal induced a significant volume and neuron loss that was accompanied by an increase in NPY expression without affecting α-MSH and TH expression. These findings indicate that the female ARN is more vulnerable to withdrawal than to excess alcohol. The data also support the hypothesis that the same pathways that regulate the expression of NPY and α-MSH in long-term ethanol intake may regulate food intake. The present model of long-term ethanol intake and withdrawal induces new physiological conditions with adaptive responses.
Collapse
Affiliation(s)
- Elce C C Rebouças
- Department of Natural Sciences, State University of Southwestern Bahia, Praça Primavera, 40-Bairro Primavera, Itapetinga, BA 45700-000, Brazil; Department of Anatomy, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal.
| | - Sandra Leal
- Department of Anatomy, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal; Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Department of Sciences, Instituto Universitário de Ciências da Saúde (IUCS), CESPU, CRL, R. Central da Gandra 1317, 4585-116 Gandra, Portugal.
| | - Susana M Silva
- Department of Anatomy, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal.
| | - Susana I Sá
- Department of Anatomy, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal.
| |
Collapse
|
78
|
Serchov T, Jilg A, Wolf CT, Radtke I, Stehle JH, Heumann R. Ras Activity Oscillates in the Mouse Suprachiasmatic Nucleus and Modulates Circadian Clock Dynamics. Mol Neurobiol 2016; 53:1843-1855. [PMID: 25762011 DOI: 10.1007/s12035-015-9135-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/22/2015] [Indexed: 12/17/2022]
Abstract
Circadian rhythms, generated in the mouse suprachiasmatic nucleus (SCN), are synchronized to the environmental day-night changes by photic input. The activation of the extracellular signal-regulated kinases 1 and 2 (ERK1,2) and cAMP response element-binding protein (CREB)-mediated transcription play a critical role in this photoentrainment. The small GTPase Ras is one of the major upstream regulators of the ERK1,2/CREB pathway. In contrast to the well-described role of Ras in structural and functional synaptic plasticity in the adult mouse brain, the physiological regulation of Ras by photic sensory input is yet unknown. Here, we describe for the first time a circadian rhythm of Ras activity in the mouse SCN. Using synRas transgenic mice, expressing constitutively activated V12-Ha-Ras selectively in neurons, we demonstrate that enhanced Ras activation causes shortening of the circadian period length. We found upregulated expression and decreased inhibitory phosphorylation of the circadian period length modulator, glycogen synthase kinase-3 beta (GSK3β), in the SCN of synRas mice. Conversely, downregulation of Ras activity by blocking its function with an antibody in oscillating cell cultures reduced protein levels and increased phosphorylation of GSK3β and lengthened the period of BMAL1 promoter-driven luciferase activity. Furthermore, enhanced Ras activity in synRas mice resulted in a potentiation of light-induced phase delays at early subjective night, and increased photic induction of pERK1,2/pCREB and c-Fos. In contrast, at late subjective night, photic activation of Ras/ERK1,2/CREB in synRas mice was not sufficient to stimulate c-Fos protein expression and phase advance the clock. Taken together, our results demonstrate that Ras activity fine tunes the period length and modulates photoentrainment of the circadian clock.
Collapse
Affiliation(s)
- Tsvetan Serchov
- Department of Molecular Neurobiochemistry, Ruhr-University, 44780, Bochum, Germany
- International Graduate School of Neuroscience, Ruhr-University, 44780, Bochum, Germany
| | - Antje Jilg
- Institute of Anatomy III, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Christian T Wolf
- Department of Molecular Neurobiochemistry, Ruhr-University, 44780, Bochum, Germany
| | - Ina Radtke
- Department of Molecular Neurobiochemistry, Ruhr-University, 44780, Bochum, Germany
| | - Jörg H Stehle
- Institute of Anatomy III, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Rolf Heumann
- Department of Molecular Neurobiochemistry, Ruhr-University, 44780, Bochum, Germany.
- International Graduate School of Neuroscience, Ruhr-University, 44780, Bochum, Germany.
| |
Collapse
|
79
|
Borbély AA, Daan S, Wirz-Justice A, Deboer T. The two-process model of sleep regulation: a reappraisal. J Sleep Res 2016; 25:131-43. [PMID: 26762182 DOI: 10.1111/jsr.12371] [Citation(s) in RCA: 809] [Impact Index Per Article: 89.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 01/01/2023]
Abstract
In the last three decades the two-process model of sleep regulation has served as a major conceptual framework in sleep research. It has been applied widely in studies on fatigue and performance and to dissect individual differences in sleep regulation. The model posits that a homeostatic process (Process S) interacts with a process controlled by the circadian pacemaker (Process C), with time-courses derived from physiological and behavioural variables. The model simulates successfully the timing and intensity of sleep in diverse experimental protocols. Electrophysiological recordings from the suprachiasmatic nuclei (SCN) suggest that S and C interact continuously. Oscillators outside the SCN that are linked to energy metabolism are evident in SCN-lesioned arrhythmic animals subjected to restricted feeding or methamphetamine administration, as well as in human subjects during internal desynchronization. In intact animals these peripheral oscillators may dissociate from the central pacemaker rhythm. A sleep/fast and wake/feed phase segregate antagonistic anabolic and catabolic metabolic processes in peripheral tissues. A deficiency of Process S was proposed to account for both depressive sleep disturbances and the antidepressant effect of sleep deprivation. The model supported the development of novel non-pharmacological treatment paradigms in psychiatry, based on manipulating circadian phase, sleep and light exposure. In conclusion, the model remains conceptually useful for promoting the integration of sleep and circadian rhythm research. Sleep appears to have not only a short-term, use-dependent function; it also serves to enforce rest and fasting, thereby supporting the optimization of metabolic processes at the appropriate phase of the 24-h cycle.
Collapse
Affiliation(s)
- Alexander A Borbély
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Serge Daan
- Centre for Behaviour and Neuroscience, University of Groningen, Groningen, the Netherlands
| | - Anna Wirz-Justice
- Centre for Chronobiology, University of Basel Psychiatric Clinics, Basel, Switzerland
| | - Tom Deboer
- Laboratory for Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
80
|
Pembroke WG, Babbs A, Davies KE, Ponting CP, Oliver PL. Temporal transcriptomics suggest that twin-peaking genes reset the clock. eLife 2015; 4. [PMID: 26523393 PMCID: PMC4718813 DOI: 10.7554/elife.10518] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/01/2015] [Indexed: 01/08/2023] Open
Abstract
The mammalian suprachiasmatic nucleus (SCN) drives daily rhythmic behavior and physiology, yet a detailed understanding of its coordinated transcriptional programmes is lacking. To reveal the finer details of circadian variation in the mammalian SCN transcriptome we combined laser-capture microdissection (LCM) and RNA-seq over a 24 hr light / dark cycle. We show that 7-times more genes exhibited a classic sinusoidal expression signature than previously observed in the SCN. Another group of 766 genes unexpectedly peaked twice, near both the start and end of the dark phase; this twin-peaking group is significantly enriched for synaptic transmission genes that are crucial for light-induced phase shifting of the circadian clock. 341 intergenic non-coding RNAs, together with novel exons of annotated protein-coding genes, including Cry1, also show specific circadian expression variation. Overall, our data provide an important chronobiological resource (www.wgpembroke.com/shiny/SCNseq/) and allow us to propose that transcriptional timing in the SCN is gating clock resetting mechanisms. DOI:http://dx.doi.org/10.7554/eLife.10518.001 The daily cycles of life in mammals are driven by a small region of the brain called the suprachiasmatic nucleus (or SCN). The SCN receives signals from sunlight and other environmental factors to help coordinate most aspects of daily biological activity and behaviour. To work correctly, it is essential that the SCN switches certain genes on and off at exactly the right time. However, many questions remain over the identity of these genes and how their levels of activity change during a 24-hour period. When a gene is active (or “being expressed”), it is used as a template to build the molecules of RNA that are needed to make proteins and to help to control how cells work. Pembroke et al. have now sequenced the RNA molecules made in the SCN of mice (which plays the same role as the equivalent human brain region) over a 24-hour period. The mice spent half of each day in the light, and half in the dark. This revealed that the expression levels of over a quarter of all the genes that are found in the SCN fluctuate over a 24-hour period. One particular group of genes peak in activity twice a day; Pembroke et al. suggest that these genes are important for controlling how an animal can adjust its body clock to light. Further research is now needed to find out which of the newly discovered fluctuating genes play the most important roles in daily activity rhythms, and which might play a part in disease. DOI:http://dx.doi.org/10.7554/eLife.10518.002
Collapse
Affiliation(s)
- William G Pembroke
- MRC Functional Genomics Unit, Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Arran Babbs
- MRC Functional Genomics Unit, Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Kay E Davies
- MRC Functional Genomics Unit, Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Chris P Ponting
- MRC Functional Genomics Unit, Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Peter L Oliver
- MRC Functional Genomics Unit, Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
81
|
Poletini MO, Ramos BC, Moraes MN, Castrucci AML. Nonvisual Opsins and the Regulation of Peripheral Clocks by Light and Hormones. Photochem Photobiol 2015; 91:1046-55. [DOI: 10.1111/php.12494] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/23/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Maristela O. Poletini
- Department of Physiology and Biophysics; Institute of Biological Sciences; Federal University of Minas Gerais; Belo Horizonte Brazil
- Department of Physiology; Institute of Biosciences; University of São Paulo; São Paulo Brazil
| | - Bruno C. Ramos
- Department of Physiology; Institute of Biosciences; University of São Paulo; São Paulo Brazil
| | - Maria Nathalia Moraes
- Department of Physiology; Institute of Biosciences; University of São Paulo; São Paulo Brazil
| | - Ana Maria L. Castrucci
- Department of Physiology; Institute of Biosciences; University of São Paulo; São Paulo Brazil
| |
Collapse
|
82
|
Kim YS, Kim YB, Kim WB, Yoon BE, Shen FY, Lee SW, Soong TW, Han HC, Colwell CS, Lee CJ, Kim YI. Histamine resets the circadian clock in the suprachiasmatic nucleus through the H1R-CaV1.3-RyR pathway in the mouse. Eur J Neurosci 2015. [DOI: 10.1111/ejn.13030] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yoon Sik Kim
- Department of Physiology and Neuroscience Research Institute; Korea University College of Medicine; Seoul 136-705 Korea
| | - Young-Beom Kim
- Department of Physiology and Neuroscience Research Institute; Korea University College of Medicine; Seoul 136-705 Korea
| | - Woong Bin Kim
- Department of Physiology and Neuroscience Research Institute; Korea University College of Medicine; Seoul 136-705 Korea
| | - Bo-Eun Yoon
- Center for Neural Science and Center for Functional Connectomics; Korea Institute of Science and Technology; Seoul 136-791 Korea
- Department of Nanobiomedical Science; Dankook University; Chungnam Korea
| | - Feng-Yan Shen
- Department of Physiology and Neuroscience Research Institute; Korea University College of Medicine; Seoul 136-705 Korea
| | - Seung Won Lee
- Department of Physiology and Neuroscience Research Institute; Korea University College of Medicine; Seoul 136-705 Korea
| | - Tuck-Wah Soong
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore Bik MD9; Singapore Singapore
| | - Hee-Chul Han
- Department of Physiology and Neuroscience Research Institute; Korea University College of Medicine; Seoul 136-705 Korea
| | - Christopher S. Colwell
- Department of Psychiatry & Biobehavioral Sciences; University of California-Los Angeles; Los Angeles CA USA
| | - C. Justin Lee
- Center for Neural Science and Center for Functional Connectomics; Korea Institute of Science and Technology; Seoul 136-791 Korea
| | - Yang In Kim
- Department of Physiology and Neuroscience Research Institute; Korea University College of Medicine; Seoul 136-705 Korea
| |
Collapse
|
83
|
Moraes MN, Ramos BC, Poletini MO, Castrucci AML. Melanopsins: Localization and Phototransduction in Xenopus laevis Melanophores. Photochem Photobiol 2015; 91:1133-41. [PMID: 26108966 DOI: 10.1111/php.12484] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 06/12/2015] [Indexed: 11/27/2022]
Abstract
Xenopus laevis melanophores express two melanopsins, Opn4x and Opn4m. We identified Opn4x immunoreactivity throughout the melanophore cytoplasm and in the cell membrane. The strongest immunopositivity for Opn4m was observed in the nuclear region, and no labeling was seen in the cell membrane. This immunodistribution suggests Opn4x as the functional photopigment. In X. laevis melanophores, light triggers pigment dispersion and clock gene induction at blue wavelength, which maximally activates melanopsins. Although light stimulation activates phospholipase C and increases intracellular calcium and cGMP, this nucleotide does not participate in photo-induced melanin dispersion. Nevertheless, the guanylyl cyclase activator YC-1 stimulates Per1 expression, similar to blue light pulse, and the use of pharmacological inhibitors indicates the participation of the phosphoinositide cascade. Since cAMP levels does not change after blue light stimulation, the cAMP/PKA pathway most probably is not involved in blue light induction of Per in X. laevis melanophores. Given the localization of melanopsins and our pharmacological data, the light-induced clock gene expression seems to be mediated by Opn4x through phosphoinositide cascade and rise in cGMP, thus leading to the reset of the biological clock in our model.
Collapse
Affiliation(s)
- Maria Nathália Moraes
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Bruno C Ramos
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Maristela O Poletini
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil.,Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria L Castrucci
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
84
|
Bosler O, Girardet C, Franc JL, Becquet D, François-Bellan AM. Structural plasticity of the circadian timing system. An overview from flies to mammals. Front Neuroendocrinol 2015; 38:50-64. [PMID: 25703789 DOI: 10.1016/j.yfrne.2015.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 12/22/2022]
Abstract
The circadian timing system orchestrates daily variations in physiology and behavior through coordination of multioscillatory cell networks that are highly plastic in responding to environmental changes. Over the last decade, it has become clear that this plasticity involves structural changes and that the changes may be observed not only in central brain regions where the master clock cells reside but also in clock-controlled structures. This review considers experimental data in invertebrate and vertebrate model systems, mainly flies and mammals, illustrating various forms of structural circadian plasticity from cellular to circuit-based levels. It highlights the importance of these plastic events in the functional adaptation of the clock to the changing environment.
Collapse
Affiliation(s)
- Olivier Bosler
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, Faculté de médecine, secteur nord, Boulevard Pierre Dramard, CS 80011, F-13344 Marseille cedex 15, France.
| | - Clémence Girardet
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, Faculté de médecine, secteur nord, Boulevard Pierre Dramard, CS 80011, F-13344 Marseille cedex 15, France.
| | - Jean-Louis Franc
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, Faculté de médecine, secteur nord, Boulevard Pierre Dramard, CS 80011, F-13344 Marseille cedex 15, France
| | - Denis Becquet
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, Faculté de médecine, secteur nord, Boulevard Pierre Dramard, CS 80011, F-13344 Marseille cedex 15, France
| | - Anne-Marie François-Bellan
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, Faculté de médecine, secteur nord, Boulevard Pierre Dramard, CS 80011, F-13344 Marseille cedex 15, France
| |
Collapse
|
85
|
Ramkisoensing A, Meijer JH. Synchronization of Biological Clock Neurons by Light and Peripheral Feedback Systems Promotes Circadian Rhythms and Health. Front Neurol 2015; 6:128. [PMID: 26097465 PMCID: PMC4456861 DOI: 10.3389/fneur.2015.00128] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 05/19/2015] [Indexed: 12/16/2022] Open
Abstract
In mammals, the suprachiasmatic nucleus (SCN) functions as a circadian clock that drives 24-h rhythms in both physiology and behavior. The SCN is a multicellular oscillator in which individual neurons function as cell-autonomous oscillators. The production of a coherent output rhythm is dependent upon mutual synchronization among single cells and requires both synaptic communication and gap junctions. Changes in phase-synchronization between individual cells have consequences on the amplitude of the SCN’s electrical activity rhythm, and these changes play a major role in the ability to adapt to seasonal changes. Both aging and sleep deprivation negatively affect the circadian amplitude of the SCN, whereas behavioral activity (i.e., exercise) has a positive effect on amplitude. Given that the amplitude of the SCN’s electrical activity rhythm is essential for achieving robust rhythmicity in physiology and behavior, the mechanisms that underlie neuronal synchronization warrant further study. A growing body of evidence suggests that the functional integrity of the SCN contributes to health, well-being, cognitive performance, and alertness; in contrast, deterioration of the 24-h rhythm is a risk factor for neurodegenerative disease, cancer, depression, and sleep disorders.
Collapse
Affiliation(s)
- Ashna Ramkisoensing
- Laboratory for Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center , Leiden , Netherlands
| | - Johanna H Meijer
- Laboratory for Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|
86
|
Vosko A, van Diepen HC, Kuljis D, Chiu AM, Heyer D, Terra H, Carpenter E, Michel S, Meijer JH, Colwell CS. Role of vasoactive intestinal peptide in the light input to the circadian system. Eur J Neurosci 2015; 42:1839-48. [PMID: 25885685 DOI: 10.1111/ejn.12919] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 04/11/2015] [Accepted: 04/13/2015] [Indexed: 12/11/2022]
Abstract
The neuropeptide vasoactive intestinal peptide (VIP) is expressed at high levels in a subset of neurons in the ventral region of the suprachiasmatic nucleus (SCN). While VIP is known to be important for the synchronization of the SCN network, the role of VIP in photic regulation of the circadian system has received less attention. In the present study, we found that the light-evoked increase in electrical activity in vivo was unaltered by the loss of VIP. In the absence of VIP, the ventral SCN still exhibited N-methyl-d-aspartate-evoked responses in a brain slice preparation, although the absolute levels of neural activity before and after treatment were significantly reduced. Next, we used calcium imaging techniques to determine if the loss of VIP altered the calcium influx due to retinohypothalamic tract stimulation. The magnitude of the evoked calcium influx was not reduced in the ventral SCN, but did decline in the dorsal SCN regions. We examined the time course of the photic induction of Period1 in the SCN using in situ hybridization in VIP-mutant mice. We found that the initial induction of Period1 was not reduced by the loss of this signaling peptide. However, the sustained increase in Period1 expression (after 30 min) was significantly reduced. Similar results were found by measuring the light induction of cFOS in the SCN. These findings suggest that VIP is critical for longer-term changes within the SCN circuit, but does not play a role in the acute light response.
Collapse
Affiliation(s)
- Andrew Vosko
- Department of Structural Medicine, Rocky Vista University, Parker, CO, USA
| | - Hester C van Diepen
- Laboratory of Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dika Kuljis
- Department of Psychiatry & Biobehavioral Sciences, University of California - Los Angeles, Los Angeles, CA, 90024, USA
| | - Andrew M Chiu
- Medical Scientist Training Program, Northwestern University, Evanston, IL, USA
| | - Djai Heyer
- Laboratory of Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Huub Terra
- Laboratory of Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ellen Carpenter
- Department of Psychiatry & Biobehavioral Sciences, University of California - Los Angeles, Los Angeles, CA, 90024, USA
| | - Stephan Michel
- Laboratory of Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Johanna H Meijer
- Laboratory of Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Christopher S Colwell
- Department of Psychiatry & Biobehavioral Sciences, University of California - Los Angeles, Los Angeles, CA, 90024, USA
| |
Collapse
|
87
|
Leone MJ, Beaule C, Marpegan L, Simon T, Herzog ED, Golombek DA. Glial and light-dependent glutamate metabolism in the suprachiasmatic nuclei. Chronobiol Int 2015; 32:573-8. [DOI: 10.3109/07420528.2015.1006328] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
88
|
Jones JR, Tackenberg MC, McMahon DG. Manipulating circadian clock neuron firing rate resets molecular circadian rhythms and behavior. Nat Neurosci 2015; 18:373-5. [PMID: 25643294 PMCID: PMC4502919 DOI: 10.1038/nn.3937] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/06/2015] [Indexed: 12/15/2022]
Abstract
To examine the interaction between molecular, electrical and behavioral circadian rhythms, we combined optogenetic manipulation of suprachiasmatic nucleus (SCN) firing rate with bioluminescence imaging and locomotor activity monitoring. Manipulating firing rate reset circadian rhythms both ex vivo and in vivo, and this resetting required spikes and network communication. This suggests that SCN firing rate is fundamental to circadian pacemaking as both an input to and output of the molecular clockworks.
Collapse
Affiliation(s)
- Jeff R. Jones
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN, USA
| | | | - Douglas G. McMahon
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN, USA
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
89
|
Permanently compromised NADPH-diaphorase activity within the osmotically activated supraoptic nucleus after in utero but not adult exposure to Aroclor 1254. Neurotoxicology 2015; 47:37-46. [DOI: 10.1016/j.neuro.2014.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/29/2014] [Accepted: 12/19/2014] [Indexed: 12/30/2022]
|
90
|
Hughes S, Jagannath A, Hankins MW, Foster RG, Peirson SN. Photic regulation of clock systems. Methods Enzymol 2014; 552:125-43. [PMID: 25707275 DOI: 10.1016/bs.mie.2014.10.018] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Circadian rhythms in physiology and behavior provide a selective advantage by enabling organisms to anticipate rhythmic changes in their environment. These rhythms are based upon a molecular clock generated via an intracellular transcriptional-translational feedback loop involving a number of key clock genes. However, to be of practical use, circadian rhythms need to be entrained to the external environment. In mammals, the primary signal for entrainment is light detected by the photoreceptors of the eye. Research on the mechanisms of photic entrainment has identified a novel photoreceptor system in the retina, consisting of photosensitive retinal ganglion cells expressing the photopigment melanopsin. Light input from these retinal photoreceptors reaches the master circadian pacemaker in the suprachiasmatic nuclei (SCN) via the retinohypothalamic tract, where it then interacts with the molecular clock to bring about entrainment. This chapter focuses on the retinal photoreceptors mediating entrainment, and how light information from the retina is transmitted to the SCN, before detailing recent advances in our understanding of how the molecular clock within the SCN is regulated by light input. Finally, the primary assays that have been used to measure photic entrainment are described.
Collapse
Affiliation(s)
- Steven Hughes
- Sleep and Circadian Institute (SCNi), Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, Oxford, United Kingdom
| | - Aarti Jagannath
- Sleep and Circadian Institute (SCNi), Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, Oxford, United Kingdom
| | - Mark W Hankins
- Sleep and Circadian Institute (SCNi), Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, Oxford, United Kingdom
| | - Russell G Foster
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom.
| | - Stuart N Peirson
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
91
|
Virag JAI, Lust RM. Circadian influences on myocardial infarction. Front Physiol 2014; 5:422. [PMID: 25400588 PMCID: PMC4214187 DOI: 10.3389/fphys.2014.00422] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/12/2014] [Indexed: 11/13/2022] Open
Abstract
Components of circadian rhythm maintenance, or "clock genes," are endogenous entrainable oscillations of about 24 h that regulate biological processes and are found in the suprachaismatic nucleus (SCN) and many peripheral tissues, including the heart. They are influenced by external cues, or Zeitgebers, such as light and heat, and can influence such diverse phenomena as cytokine expression immune cells, metabolic activity of cardiac myocytes, and vasodilator regulation by vascular endothelial cells. While it is known that the central master clock in the SCN synchronizes peripheral physiologic rhythms, the mechanisms by which the information is transmitted are complex and may include hormonal, metabolic, and neuronal inputs. Whether circadian patterns are causally related to the observed periodicity of events, or whether they are simply epi-phenomena is not well established, but a few studies suggest that the circadian effects likely are real in their impact on myocardial infarct incidence. Cycle disturbances may be harbingers of predisposition and subsequent response to acute and chronic cardiac injury, and identifying the complex interactions of circadian rhythms and myocardial infarction may provide insights into possible preventative and therapeutic strategies for susceptible populations.
Collapse
Affiliation(s)
- Jitka A I Virag
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Robert M Lust
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| |
Collapse
|
92
|
Iyer R, Wang TA, Gillette MU. Circadian gating of neuronal functionality: a basis for iterative metaplasticity. Front Syst Neurosci 2014; 8:164. [PMID: 25285070 PMCID: PMC4168688 DOI: 10.3389/fnsys.2014.00164] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 08/22/2014] [Indexed: 02/06/2023] Open
Abstract
Brain plasticity, the ability of the nervous system to encode experience, is a modulatory process leading to long-lasting structural and functional changes. Salient experiences induce plastic changes in neurons of the hippocampus, the basis of memory formation and recall. In the suprachiasmatic nucleus (SCN), the central circadian (~24-h) clock, experience with light at night induces changes in neuronal state, leading to circadian plasticity. The SCN's endogenous ~24-h time-generator comprises a dynamic series of functional states, which gate plastic responses. This restricts light-induced alteration in SCN state-dynamics and outputs to the nighttime. Endogenously generated circadian oscillators coordinate the cyclic states of excitability and intracellular signaling molecules that prime SCN receptivity to plasticity signals, generating nightly windows of susceptibility. We propose that this constitutes a paradigm of ~24-h iterative metaplasticity, the repeated, patterned occurrence of susceptibility to induction of neuronal plasticity. We detail effectors permissive for the cyclic susceptibility to plasticity. We consider similarities of intracellular and membrane mechanisms underlying plasticity in SCN circadian plasticity and in hippocampal long-term potentiation (LTP). The emerging prominence of the hippocampal circadian clock points to iterative metaplasticity in that tissue as well. Exploring these links holds great promise for understanding circadian shaping of synaptic plasticity, learning, and memory.
Collapse
Affiliation(s)
- Rajashekar Iyer
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign Urbana, IL, USA
| | - Tongfei A Wang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign Urbana, IL, USA
| | - Martha U Gillette
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign Urbana, IL, USA ; Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign Urbana, IL, USA
| |
Collapse
|
93
|
Zhou L, Gao Q, Zhang P, Guo S, Gu J, Hao W, Cao JM. Activation of growth hormone secretagogue receptor induces time-dependent clock phase delay in mice. Am J Physiol Endocrinol Metab 2014; 307:E515-26. [PMID: 25074983 DOI: 10.1152/ajpendo.00535.2013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Early studies have reported a phase-shifting effect of growth hormone secretagogues (GHSs). This study aimed to determine the mechanism of action of GHSs. We examined the response of the hypothalamic suprachiasmatic nuclei (SCN) to growth hormone releasing peptide-6 (GHRP-6) by assessing effects on the phase of locomotor activity rhythms, SCN neuronal discharges, and the potential signaling pathways involved in the drug action on circadian rhythms. The results showed that bolus administration of GHRP-6 (100 μg/kg ip) at the beginning of subjective night (CT12) induced a phase delay of the free-running rhythms in male C57BL/6J mice under constant darkness, but did not elicit phase shift at other checked circadian time (CT) points. The phase-delay effect of GHRP-6 was abolished by d-(+)-Lys-GHRP-6 (GHS receptor antagonist), KN-93 [calcium/calmodulin-dependent protein kinase II (CaMK) II inhibitor], or anti-phosphorylated (p)-cAMP response element-binding protein (CREB) antibody. Further analyses demonstrated that GHRP-6 at CT12 induced higher calcium mobilization and neuronal discharge in the SCN compared with that at CT6, decreased the levels of glutamate and γ-aminobutyric acid, increased the levels of p-CaMKII, p-CREB, and period 1, and delayed the circadian expressions of circadian locomotor output cycles kaput, Bmal1, and prokineticin 2 in the SCN; these signaling changes resulted in behavioral phase delay. Collectively, GHRP-6 induces a CT-dependent phase delay via activating GHS receptor and the downstream signaling, which is partially similar to the signaling cascade of light-induced phase delay at early night. These novel observations may help to better understand the role of GHSs in circadian physiology.
Collapse
Affiliation(s)
- Lan Zhou
- Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Qian Gao
- Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Peng Zhang
- Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Shu Guo
- Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jingli Gu
- Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wei Hao
- Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Ji-Min Cao
- Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
94
|
van Diepen HC, Lucassen EA, Yasenkov R, Groenen I, Ijzerman AP, Meijer JH, Deboer T. Caffeine increases light responsiveness of the mouse circadian pacemaker. Eur J Neurosci 2014; 40:3504-11. [DOI: 10.1111/ejn.12715] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/28/2014] [Accepted: 08/05/2014] [Indexed: 12/19/2022]
Affiliation(s)
- Hester C. van Diepen
- Laboratory for Neurophysiology; Department of Molecular Cell Biology; Leiden University Medical Centre; PO Box 9600 Mailbox S5-P 2300 RC Leiden The Netherlands
| | - Eliane A. Lucassen
- Laboratory for Neurophysiology; Department of Molecular Cell Biology; Leiden University Medical Centre; PO Box 9600 Mailbox S5-P 2300 RC Leiden The Netherlands
| | - Roman Yasenkov
- Laboratory for Neurophysiology; Department of Molecular Cell Biology; Leiden University Medical Centre; PO Box 9600 Mailbox S5-P 2300 RC Leiden The Netherlands
| | - Inske Groenen
- Laboratory for Neurophysiology; Department of Molecular Cell Biology; Leiden University Medical Centre; PO Box 9600 Mailbox S5-P 2300 RC Leiden The Netherlands
| | - Adriaan P. Ijzerman
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research; Leiden The Netherlands
| | - Johanna H. Meijer
- Laboratory for Neurophysiology; Department of Molecular Cell Biology; Leiden University Medical Centre; PO Box 9600 Mailbox S5-P 2300 RC Leiden The Netherlands
| | - Tom Deboer
- Laboratory for Neurophysiology; Department of Molecular Cell Biology; Leiden University Medical Centre; PO Box 9600 Mailbox S5-P 2300 RC Leiden The Netherlands
| |
Collapse
|
95
|
Ramos BCR, Moraes MNCM, Poletini MO, Lima LHRG, Castrucci AML. From blue light to clock genes in zebrafish ZEM-2S cells. PLoS One 2014; 9:e106252. [PMID: 25184495 PMCID: PMC4153568 DOI: 10.1371/journal.pone.0106252] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/29/2014] [Indexed: 01/22/2023] Open
Abstract
Melanopsin has been implicated in the mammalian photoentrainment by blue light. This photopigment, which maximally absorbs light at wavelengths between 470 and 480 nm depending on the species, is found in the retina of all classes of vertebrates so far studied. In mammals, melanopsin activation triggers a signaling pathway which resets the circadian clock in the suprachiasmatic nucleus (SCN). Unlike mammals, Drosophila melanogaster and Danio rerio do not rely only on their eyes to perceive light, in fact their whole body may be capable of detecting light and entraining their circadian clock. Melanopsin, teleost multiple tissue (tmt) opsin and others such as neuropsin and va-opsin, are found in the peripheral tissues of Danio rerio, however, there are limited data concerning the photopigment/s or the signaling pathway/s directly involved in light detection. Here, we demonstrate that melanopsin is a strong candidate to mediate synchronization of zebrafish cells. The deduced amino acid sequence of melanopsin, although being a vertebrate opsin, is more similar to invertebrate than vertebrate photopigments, and melanopsin photostimulation triggers the phosphoinositide pathway through activation of a G(q/11)-type G protein. We stimulated cultured ZEM-2S cells with blue light at wavelengths consistent with melanopsin maximal absorption, and evaluated the time course expression of per1b, cry1b, per2 and cry1a. Using quantitative PCR, we showed that blue light is capable of slightly modulating per1b and cry1b genes, and drastically increasing per2 and cry1a expression. Pharmacological assays indicated that per2 and cry1a responses to blue light are evoked through the activation of the phosphoinositide pathway, which crosstalks with nitric oxide (NO) and mitogen activated protein MAP kinase (MAPK) to activate the clock genes. Our results suggest that melanopsin may be important in mediating the photoresponse in Danio rerio ZEM-2S cells, and provide new insights about the modulation of clock genes in peripheral clocks.
Collapse
Affiliation(s)
- Bruno C. R. Ramos
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | | | - Maristela O. Poletini
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
- Department of Physiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo H. R. G. Lima
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Ana Maria L. Castrucci
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
96
|
Matta-Camacho E, Banerjee S, Hughes TS, Solt LA, Wang Y, Burris TP, Kojetin DJ. Structure of REV-ERBβ ligand-binding domain bound to a porphyrin antagonist. J Biol Chem 2014; 289:20054-66. [PMID: 24872411 DOI: 10.1074/jbc.m113.545111] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
REV-ERBα and REV-ERBβ are members of the nuclear receptor (NR) superfamily of ligand-regulated transcription factors that play important roles in the regulation of circadian physiology, metabolism, and immune function. Although the REV-ERBs were originally characterized as orphan receptors, recent studies have demonstrated that they function as receptors for heme. Here, we demonstrate that cobalt protoporphyrin IX (CoPP) and zinc protoporphyrin IX (ZnPP) are ligands that bind directly to the REV-ERBs. However, instead of mimicking the agonist action of heme, CoPP and ZnPP function as antagonists of REV-ERB function. This was unexpected because the only distinction between these ligands is the metal ion that is coordinated. To understand the structural basis by which REV-ERBβ can differentiate between a porphyrin agonist and antagonist, we characterized the interaction between REV-ERBβ with heme, CoPP, and ZnPP using biochemical and structural approaches, including x-ray crystallography and NMR. The crystal structure of CoPP-bound REV-ERBβ indicates only minor conformational changes induced by CoPP compared with heme, including the porphyrin ring of CoPP, which adopts a planar conformation as opposed to the puckered conformation observed in the heme-bound REV-ERBβ crystal structure. Thus, subtle changes in the porphyrin metal center and ring conformation may influence the agonist versus antagonist action of porphyrins and when considered with other studies suggest that gas binding to the iron metal center heme may drive alterations in REV-ERB activity.
Collapse
Affiliation(s)
- Edna Matta-Camacho
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and
| | - Subhashis Banerjee
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and
| | - Travis S Hughes
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and
| | - Laura A Solt
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and
| | - Yongjun Wang
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and the Department of Pharmacological and Physiological Sciences, St. Louis University School of Medicine, St. Louis, Missouri 63103
| | - Thomas P Burris
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and the Department of Pharmacological and Physiological Sciences, St. Louis University School of Medicine, St. Louis, Missouri 63103
| | - Douglas J Kojetin
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and
| |
Collapse
|
97
|
Circadian modulation of the Cl(-) equilibrium potential in the rat suprachiasmatic nuclei. BIOMED RESEARCH INTERNATIONAL 2014; 2014:424982. [PMID: 24949446 PMCID: PMC4052495 DOI: 10.1155/2014/424982] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/23/2014] [Accepted: 03/27/2014] [Indexed: 11/18/2022]
Abstract
The suprachiasmatic nuclei (SCN) constitute a circadian clock in mammals, where γ-amino-butyric acid (GABA) neurotransmission prevails and participates in different aspects of circadian regulation. Evidence suggests that GABA has an excitatory function in the SCN in addition to its typical inhibitory role. To examine this possibility further, we determined the equilibrium potential of GABAergic postsynaptic currents (E(GABA)) at different times of the day and in different regions of the SCN, using either perforated or whole cell patch clamp. Our results indicate that during the day most neurons in the dorsal SCN have an E(GABA) close to -30 mV while in the ventral SCN they have an E(GABA) close to -60 mV; this difference reverses during the night, in the dorsal SCN neurons have an E(GABA) of -60 mV and in the ventral SCN they have an E(GABA) of -30 mV. The depolarized equilibrium potential can be attributed to the activity of the Na(+)-K(+)-2Cl(-) (NKCC) cotransporter since the equilibrium potential becomes more negative following addition of the NKCC blocker bumetanide. Our results suggest an excitatory role for GABA in the SCN and further indicate both time (day versus night) and regional (dorsal versus ventral) modulation of E(GABA) in the SCN.
Collapse
|
98
|
A novel mechanism controlling resetting speed of the circadian clock to environmental stimuli. Curr Biol 2014; 24:766-73. [PMID: 24656826 PMCID: PMC3988889 DOI: 10.1016/j.cub.2014.02.027] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 01/10/2014] [Accepted: 02/11/2014] [Indexed: 11/23/2022]
Abstract
Many aspects of mammalian physiology are driven through the coordinated action of internal circadian clocks. Clock speed (period) and phase (temporal alignment) are fundamental to an organism’s ability to synchronize with its environment. In humans, lifestyles that disturb these clocks, such as shift work, increase the incidence of diseases such as cancer and diabetes. Casein kinases 1δ and ε are closely related clock components implicated in period determination. However, CK1δ is so dominant in this regard that it remains unclear what function CK1ε normally serves. Here, we reveal that CK1ε dictates how rapidly the clock is reset by environmental stimuli. Genetic disruption of CK1ε in mice enhances phase resetting of behavioral rhythms to acute light pulses and shifts in light cycle. This impact of CK1ε targeting is recapitulated in isolated brain suprachiasmatic nucleus and peripheral (lung) clocks during NMDA- or temperature-induced phase shift in association with altered PERIOD (PER) protein dynamics. Importantly, accelerated re-entrainment of the circadian system in vivo and in vitro can be achieved in wild-type animals through pharmacological inhibition of CK1ε. These studies therefore reveal a role for CK1ε in stabilizing the circadian clock against phase shift and highlight it as a novel target for minimizing physiological disturbance in shift workers. CK1ε−/− mice exhibit enhanced phase resetting to advancing and delaying stimuli Selective pharmacological inhibition of CK1ε with PF4800567 enhances resetting Accelerated resetting is observed in central and peripheral clocks Accelerated resetting reduces physiological desynchrony
Collapse
|
99
|
Adenosine and glutamate in neuroglial interaction: implications for circadian disorders and alcoholism. ADVANCES IN NEUROBIOLOGY 2014; 11:103-19. [PMID: 25236726 DOI: 10.1007/978-3-319-08894-5_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Recent studies have demonstrated that the function of glia is not restricted to the support of neuronal function. In fact, astrocytes are essential for neuronal activity in the brain and play an important role in the regulation of complex behavior. Astrocytes actively participate in synapse formation and brain information processing by releasing and uptaking glutamate, D-serine, adenosine 5'-triphosphate (ATP), and adenosine. In the central nervous system, adenosine-mediated neuronal activity modulates the actions of other neurotransmitter systems. Adenosinergic fine-tuning of the glutamate system in particular has been shown to regulate circadian rhythmicity and sleep, as well as alcohol-related behavior and drinking. Adenosine gates both photic (light-induced) glutamatergic and nonphotic (alerting) input to the circadian clock located in the suprachiasmatic nucleus of the hypothalamus. Astrocytic, SNARE-mediated ATP release provides the extracellular adenosine that drives homeostatic sleep. Acute ethanol increases extracellular adenosine, which mediates the ataxic and hypnotic/sedative effects of alcohol, while chronic ethanol leads to downregulated adenosine signaling that underlies insomnia, a major predictor of relapse. Studies using mice lacking the equilibrative nucleoside transporter 1 have illuminated how adenosine functions through neuroglial interactions involving glutamate uptake transporter GLT-1 [referred to as excitatory amino acid transporter 2 (EAAT2) in human] and possibly water channel aquaporin 4 to regulate ethanol sensitivity, reward-related motivational processes, and alcohol intake.
Collapse
|
100
|
Irwin RP, Allen CN. Simultaneous electrophysiological recording and calcium imaging of suprachiasmatic nucleus neurons. J Vis Exp 2013:50794. [PMID: 24335611 DOI: 10.3791/50794] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Simultaneous electrophysiological and fluorescent imaging recording methods were used to study the role of changes of membrane potential or current in regulating the intracellular calcium concentration. Changing environmental conditions, such as the light-dark cycle, can modify neuronal and neural network activity and the expression of a family of circadian clock genes within the suprachiasmatic nucleus (SCN), the location of the master circadian clock in the mammalian brain. Excitatory synaptic transmission leads to an increase in the postsynaptic Ca(2+) concentration that is believed to activate the signaling pathways that shifts the rhythmic expression of circadian clock genes. Hypothalamic slices containing the SCN were patch clamped using microelectrodes filled with an internal solution containing the calcium indicator bis-fura-2. After a seal was formed between the microelectrode and the SCN neuronal membrane, the membrane was ruptured using gentle suction and the calcium probe diffused into the neuron filling both the soma and dendrites. Quantitative ratiometric measurements of the intracellular calcium concentration were recorded simultaneously with membrane potential or current. Using these methods it is possible to study the role of changes of the intracellular calcium concentration produced by synaptic activity and action potential firing of individual neurons. In this presentation we demonstrate the methods to simultaneously record electrophysiological activity along with intracellular calcium from individual SCN neurons maintained in brain slices.
Collapse
Affiliation(s)
- Robert P Irwin
- Center for Research on Occupational and Environmental Toxicology, Oregon Health & Science University
| | | |
Collapse
|