Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Gen Physiol ; 155(9)2023 09 04.
Article in English | MEDLINE | ID: mdl-37516908

ABSTRACT

Considerable evidence suggests that day-night rhythms in the functional expression of subthreshold potassium (K+) channels regulate daily oscillations in the spontaneous firing rates of neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals. The K+ conductance(s) driving these daily rhythms in the repetitive firing rates of SCN neurons, however, have not been identified. To test the hypothesis that subthreshold Kv12.1/Kv12.2-encoded K+ channels play a role, we obtained current-clamp recordings from SCN neurons in slices prepared from adult mice harboring targeted disruptions in the Kcnh8 (Kv12.1-/-) or Kcnh3 (Kv12.2-/-) locus. We found that mean nighttime repetitive firing rates were higher in Kv12.1-/- and Kv12.2-/- than in wild type (WT), SCN neurons. In marked contrast, mean daytime repetitive firing rates were similar in Kv12.1-/-, Kv12.2-/-, and WT SCN neurons, and the day-night difference in mean repetitive firing rates, a hallmark feature of WT SCN neurons, was eliminated in Kv12.1-/- and Kv12.2-/- SCN neurons. Similar results were obtained with in vivo shRNA-mediated acute knockdown of Kv12.1 or Kv12.2 in adult SCN neurons. Voltage-clamp experiments revealed that Kv12-encoded current densities in WT SCN neurons are higher at night than during the day. In addition, the pharmacological block of Kv12-encoded currents increased the mean repetitive firing rate of nighttime, but not daytime, in WT SCN neurons. Dynamic clamp-mediated subtraction of modeled Kv12-encoded currents also selectively increased the mean repetitive firing rates of nighttime WT SCN neurons. Despite the elimination of the nighttime decrease in the mean repetitive firing rates of SCN neurons, however, locomotor (wheel-running) activity remained rhythmic in Kv12.1-/-, Kv12.2-/-, and Kv12.1-targeted shRNA-expressing, and Kv12.2-targeted shRNA-expressing animals.


Subject(s)
Suprachiasmatic Nucleus Neurons , Animals , Mice , Mammals , Neurons , Potassium , RNA, Small Interfering , Suprachiasmatic Nucleus
2.
bioRxiv ; 2023 Feb 02.
Article in English | MEDLINE | ID: mdl-36778242

ABSTRACT

Considerable evidence suggests that day-night rhythms in the functional expression of subthreshold potassium (K + ) channels regulate daily oscillations in the rates of spontaneous action potential firing of neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals. The K + conductance(s) driving these daily rhythms in repetitive firing rates, however, have not been identified. To test the hypothesis that subthreshold Kv12.1/Kv12.2-encoded K + channels play a role, we obtained current-clamp recordings from SCN neurons in slices prepared from adult mice harboring targeted disruptions in the Kcnh8 (Kv12.1 -/- ) or Kcnh3 (Kv12.2 -/- ) locus. We found that mean nighttime repetitive firing rates were higher in Kv12.1 -/- and Kv12.2 -/- , than in wild type (WT), SCN neurons. In marked contrast, mean daytime repetitive firing rates were similar in Kv12.1 -/- , Kv12.2 -/- and WT SCN neurons, and the day-night difference in mean repetitive firing rates, a hallmark feature of WT SCN neurons, was eliminated in Kv12.1 -/- and Kv12.2 -/- SCN neurons. Similar results were obtained with in vivo shRNA-mediated acute knockdown of Kv12.1 or Kv12.2 in adult SCN neurons. Voltage-clamp experiments revealed that Kv12-encoded current densities in WT SCN neurons are higher at night than during the day. In addition, pharmacological block of Kv12-encoded currents increased the mean repetitive firing rate of nighttime, but not daytime, in WT SCN neurons. Dynamic clamp-mediated subtraction of modeled Kv12-encoded currents also selectively increased the mean repetitive firing rates of nighttime WT SCN neurons. Despite the elimination of nighttime decrease in the mean repetitive firing rates of SCN neurons, however, locomotor (wheel-running) activity remained rhythmic in Kv12.1 -/- , Kv12.2 -/- , Kv12.1-targeted shRNA-expressing, and Kv12.2-targeted shRNA-expressing animals.

3.
Liver Int ; 43(3): 673-683, 2023 03.
Article in English | MEDLINE | ID: mdl-36367321

ABSTRACT

Patients with cirrhosis exhibit features of circadian disruption. Hyperammonaemia has been suggested to impair both homeostatic and circadian sleep regulation. Here, we tested if hyperammonaemia directly disrupts circadian rhythm generation in the central pacemaker, the suprachiasmatic nuclei (SCN) of the hypothalamus. Wheel-running activity was recorded from mice fed with a hyperammonaemic or normal diet for ~35 days in a 12:12 light-dark (LD) cycle followed by ~15 days in constant darkness (DD). The expression of the clock protein PERIOD2 (PER2) was recorded from SCN explants before, during and after ammonia exposure, ±glutamate receptor antagonists. In LD, hyperammonaemic mice advanced their daily activity onset time by ~1 h (16.8 ± 0.3 vs. 18.1 ± 0.04 h, p = .009) and decreased their total activity, concentrating it during the first half of the night. In DD, hyperammonaemia reduced the amplitude of daily activity (551.5 ± 27.7 vs. 724.9 ± 59 counts, p = .007), with no changes in circadian period. Ammonia (≥0.01 mM) rapidly and significantly reduced PER2 amplitude, and slightly increased circadian period. The decrease in PER2 amplitude correlated with decreased synchrony among circadian cells in the SCN and increased extracellular glutamate, which was rescued by AMPA glutamate receptor antagonists. These data suggest that hyperammonaemia affects circadian regulation of rest-activity behaviour by increasing extracellular glutamate in the SCN.


Subject(s)
Glutamic Acid , Hyperammonemia , Mice , Animals , Ammonia , Excitatory Amino Acid Antagonists , Circadian Rhythm/physiology
4.
Nat Commun ; 12(1): 5763, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34599158

ABSTRACT

Signals from the central circadian pacemaker, the suprachiasmatic nucleus (SCN), must be decoded to generate daily rhythms in hormone release. Here, we hypothesized that the SCN entrains rhythms in the paraventricular nucleus (PVN) to time the daily release of corticosterone. In vivo recording revealed a critical circuit from SCN vasoactive intestinal peptide (SCNVIP)-producing neurons to PVN corticotropin-releasing hormone (PVNCRH)-producing neurons. PVNCRH neurons peak in clock gene expression around midday and in calcium activity about three hours later. Loss of the clock gene Bmal1 in CRH neurons results in arrhythmic PVNCRH calcium activity and dramatically reduces the amplitude and precision of daily corticosterone release. SCNVIP activation reduces (and inactivation increases) corticosterone release and PVNCRH calcium activity, and daily SCNVIP activation entrains PVN clock gene rhythms by inhibiting PVNCRH neurons. We conclude that daily corticosterone release depends on coordinated clock gene and neuronal activity rhythms in both SCNVIP and PVNCRH neurons.


Subject(s)
Circadian Rhythm/physiology , Glucocorticoids/metabolism , Neurons/physiology , Paraventricular Hypothalamic Nucleus/physiology , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Calcium/metabolism , Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Corticosterone/pharmacology , Corticotropin-Releasing Hormone/metabolism , Feces/chemistry , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Photometry , Suprachiasmatic Nucleus/physiology
5.
J R Soc Interface ; 18(182): 20210454, 2021 09.
Article in English | MEDLINE | ID: mdl-34520693

ABSTRACT

In the suprachiasmatic nucleus (SCN), γ-aminobutyric acid (GABA) is a primary neurotransmitter. GABA can signal through two types of GABAA receptor subunits, often referred to as synaptic GABAA (gamma subunit) and extra-synaptic GABAA (delta subunit). To test the functional roles of these distinct GABAA in regulating circadian rhythms, we developed a multicellular SCN model where we could separately compare the effects of manipulating GABA neurotransmitter or receptor dynamics. Our model predicted that blocking GABA signalling modestly increased synchrony among circadian cells, consistent with published SCN pharmacology. Conversely, the model predicted that lowering GABAA receptor density reduced firing rate, circadian cell fraction, amplitude and synchrony among individual neurons. When we tested these predictions, we found that the knockdown of delta GABAA reduced the amplitude and synchrony of clock gene expression among cells in SCN explants. The model further predicted that increasing gamma GABAA densities could enhance synchrony, as opposed to increasing delta GABAA densities. Overall, our model reveals how blocking GABAA receptors can modestly increase synchrony, while increasing the relative density of gamma over delta subunits can dramatically increase synchrony. We hypothesize that increased gamma GABAA density in the winter could underlie the tighter phase relationships among SCN cells.


Subject(s)
Suprachiasmatic Nucleus , gamma-Aminobutyric Acid , Circadian Rhythm , Neurons , Receptors, GABA
6.
Proc Natl Acad Sci U S A ; 115(37): 9300-9305, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150403

ABSTRACT

Extracting complex interactions (i.e., dynamic topologies) has been an essential, but difficult, step toward understanding large, complex, and diverse systems including biological, financial, and electrical networks. However, reliable and efficient methods for the recovery or estimation of network topology remain a challenge due to the tremendous scale of emerging systems (e.g., brain and social networks) and the inherent nonlinearity within and between individual units. We develop a unified, data-driven approach to efficiently infer connections of networks (ICON). We apply ICON to determine topology of networks of oscillators with different periodicities, degree nodes, coupling functions, and time scales, arising in silico, and in electrochemistry, neuronal networks, and groups of mice. This method enables the formulation of these large-scale, nonlinear estimation problems as a linear inverse problem that can be solved using parallel computing. Working with data from networks, ICON is robust and versatile enough to reliably reveal full and partial resonance among fast chemical oscillators, coherent circadian rhythms among hundreds of cells, and functional connectivity mediating social synchronization of circadian rhythmicity among mice over weeks.


Subject(s)
Models, Theoretical
7.
eNeuro ; 4(3)2017.
Article in English | MEDLINE | ID: mdl-28560311

ABSTRACT

Rapidly activating and inactivating A-type K+ currents (IA) encoded by Kv4.2 and Kv4.3 pore-forming (α) subunits of the Kv4 subfamily are key regulators of neuronal excitability. Previous studies have suggested a role for Kv4.1 α-subunits in regulating the firing properties of mouse suprachiasmatic nucleus (SCN) neurons. To test this, we utilized an RNA-interference strategy to knockdown Kv4.1, acutely and selectively, in the SCN. Current-clamp recordings revealed that the in vivo knockdown of Kv4.1 significantly (p < 0.0001) increased mean ± SEM repetitive firing rates in SCN neurons during the day (6.4 ± 0.5 Hz) and at night (4.3 ± 0.6 Hz), compared with nontargeted shRNA-expressing SCN neurons (day: 3.1 ± 0.5 Hz; night: 1.6 ± 0.3 Hz). IA was also significantly (p < 0.05) reduced in Kv4.1-targeted shRNA-expressing SCN neurons (day: 80.3 ± 11.8 pA/pF; night: 55.3 ± 7.7 pA/pF), compared with nontargeted shRNA-expressing (day: 121.7 ± 10.2 pA/pF; night: 120.6 ± 16.5 pA/pF) SCN neurons. The magnitude of the effect of Kv4.1-targeted shRNA expression on firing rates and IA was larger at night. In addition, Kv4.1-targeted shRNA expression significantly (p < 0.001) increased mean ± SEM nighttime input resistance (Rin; 2256 ± 166 MΩ), compared to nontargeted shRNA-expressing SCN neurons (1143 ± 93 MΩ). Additional experiments revealed that acute knockdown of Kv4.1 significantly (p < 0.01) shortened, by ∼0.5 h, the circadian period of spontaneous electrical activity, clock gene expression and locomotor activity demonstrating a physiological role for Kv4.1-encoded IA channels in regulating circadian rhythms in neuronal excitability and behavior.


Subject(s)
Circadian Rhythm/physiology , Motor Activity/physiology , Neurons/metabolism , Period Circadian Proteins/metabolism , Shal Potassium Channels/metabolism , Suprachiasmatic Nucleus/metabolism , Action Potentials/physiology , Animals , Cells, Cultured , Gene Knockdown Techniques , Male , Mice, Inbred C57BL , Mice, Transgenic , Patch-Clamp Techniques , Period Circadian Proteins/genetics , Photoperiod , RNA Interference , Shal Potassium Channels/genetics , Tissue Culture Techniques
8.
J Biol Rhythms ; 32(1): 35-47, 2017 02.
Article in English | MEDLINE | ID: mdl-28326909

ABSTRACT

Although the suprachiasmatic nucleus (SCN) has long been considered the master circadian clock in mammals, the topology of the connections that synchronize daily rhythms among SCN cells is not well understood. We combined experimental and computational methods to infer the directed interactions that mediate circadian synchrony between regions of the SCN. We analyzed PERIOD2 (PER2) expression from SCN slices during and after treatment with tetrodotoxin, allowing us to map connections as cells resynchronized their daily cycling following blockade and restoration of cell-cell communication. Using automated analyses, we found that cells in the dorsal SCN stabilized their periods slower than those in the ventral SCN. A phase-amplitude computational model of the SCN revealed that, to reproduce the experimental results: (1) the ventral SCN had to be more densely connected than the dorsal SCN and (2) the ventral SCN needed strong connections to the dorsal SCN. Taken together, these results provide direct evidence that the ventral SCN entrains the dorsal SCN in constant conditions.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Luciferases/metabolism , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/physiology , Algorithms , Animals , Arginine Vasopressin/metabolism , Luciferases/genetics , Luminescent Measurements/methods , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Period Circadian Proteins/genetics , Sodium Channel Blockers/pharmacology , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/metabolism , Tetrodotoxin/pharmacology , Vasoactive Intestinal Peptide/metabolism
9.
Proc Natl Acad Sci U S A ; 113(16): 4512-7, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27044085

ABSTRACT

In the mammalian suprachiasmatic nucleus (SCN), noisy cellular oscillators communicate within a neuronal network to generate precise system-wide circadian rhythms. Although the intracellular genetic oscillator and intercellular biochemical coupling mechanisms have been examined previously, the network topology driving synchronization of the SCN has not been elucidated. This network has been particularly challenging to probe, due to its oscillatory components and slow coupling timescale. In this work, we investigated the SCN network at a single-cell resolution through a chemically induced desynchronization. We then inferred functional connections in the SCN by applying the maximal information coefficient statistic to bioluminescence reporter data from individual neurons while they resynchronized their circadian cycling. Our results demonstrate that the functional network of circadian cells associated with resynchronization has small-world characteristics, with a node degree distribution that is exponential. We show that hubs of this small-world network are preferentially located in the central SCN, with sparsely connected shells surrounding these cores. Finally, we used two computational models of circadian neurons to validate our predictions of network structure.


Subject(s)
Circadian Clocks/physiology , Nerve Net/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Genes, Reporter , Mice, Transgenic , Nerve Net/cytology , Suprachiasmatic Nucleus/cytology
10.
Eur J Neurosci ; 42(10): 2833-42, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26417679

ABSTRACT

The suprachiasmatic nucleus (SCN) in mammals is the master clock which regulates circadian rhythms. Neural activity of SCN neurons is synchronized to external light through the retinohypothalamic tract (RHT). The paraventricular thalamic nucleus (PVT) is a neural structure that receives synaptic inputs from, and projects back to, the SCN. Lesioning the anterior PVT (aPVT) modifies the behavioral phase response curve induced by short pulses of bright light. In order to study the influence of the aPVT on SCN neural activity, we addressed whether the stimulation of the aPVT can modulate the electrical response of the SCN to either retinal or RHT stimulation. Using in vitro and in vivo recordings, we found a large population of SCN neurons responsive to the stimulation of either aPVT or RHT pathways. Furthermore, we found that simultaneous stimulation of the aPVT and the RHT increased neuronal responsiveness and spontaneous firing rate (SFR) in neurons with a low basal SFR (which also have more negative membrane potentials), such as quiescent and arrhythmic neurons, but no change was observed in neurons with rhythmic firing patterns and more depolarized membrane potentials. These results suggest that inputs from the aPVT could shift the membrane potential of an SCN neuron to values closer to its firing threshold and thus contribute to integration of the response of the circadian clock to light.


Subject(s)
Midline Thalamic Nuclei/physiology , Neurons/physiology , Retina/physiology , Suprachiasmatic Nucleus/physiology , Action Potentials , Animals , Electric Stimulation , Male , Rats , Rats, Wistar , Visual Pathways/physiology
11.
J Biol Rhythms ; 30(5): 396-407, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26152125

ABSTRACT

Neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals, display daily rhythms in electrical activity with more depolarized resting potentials and higher firing rates during the day than at night. Although these daily variations in the electrical properties of SCN neurons are required for circadian rhythms in physiology and behavior, the mechanisms linking changes in neuronal excitability to the molecular clock are not known. Recently, we reported that mice deficient for either Kcna4 (Kv1.4(-/-)) or Kcnd2 (Kv4.2(-/-); but not Kcnd3, Kv4.3(-/-)), voltage-gated K(+) (Kv) channel pore-forming subunits that encode subthreshold, rapidly activating, and inactivating K(+) currents (IA), have shortened (0.5 h) circadian periods in SCN firing and in locomotor activity compared with wild-type (WT) mice. In the experiments here, we used a mouse (Per2(Luc)) line engineered with a bioluminescent reporter construct, PERIOD2::LUCIFERASE (PER2::LUC), replacing the endogenous Per2 locus, to test the hypothesis that the loss of Kv1.4- or Kv4.2-encoded IA channels also modifies circadian rhythms in the expression of the clock protein PERIOD2 (PER2). We found that SCN explants from Kv1.4(-/-)Per2(Luc) and Kv4.2(-/-) Per2(Luc), but not Kv4.3(-/-)Per2(Luc), mice have significantly shorter (by approximately 0.5 h) circadian periods in PER2 rhythms, compared with explants from Per2(Luc) mice, revealing that the membrane properties of SCN neurons feedback to regulate clock (PER2) expression. The combined loss of both Kv1.4- and Kv4.2-encoded IA channels in Kv1.4(-/-)/Kv4.2(-/-)Per2(Luc) SCN explants did not result in any further alterations in PER2 rhythms. Interestingly, however, mice lacking both Kv1.4 and Kv4.2 show a striking (approximately 1.8 h) advance in their daily activity onset in a light cycle compared with WT mice, suggesting additional roles for Kv1.4- and Kv4.2-encoded IA channels in controlling the light-dependent responses of neurons within and/or outside of the SCN to regulate circadian phase of daily activity.


Subject(s)
Circadian Rhythm/physiology , Kv1.4 Potassium Channel/physiology , Period Circadian Proteins/metabolism , Shal Potassium Channels/physiology , Suprachiasmatic Nucleus/physiology , Animals , Circadian Rhythm/genetics , Ion Channel Gating/genetics , Ion Channel Gating/physiology , Kv1.4 Potassium Channel/genetics , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements/methods , Male , Membrane Potentials/genetics , Membrane Potentials/physiology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Motor Activity/genetics , Motor Activity/physiology , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques , Period Circadian Proteins/genetics , Shal Potassium Channels/genetics , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/metabolism , Tissue Culture Techniques
12.
J Neurosci ; 34(17): 6040-6, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24760863

ABSTRACT

Accumulating evidence suggests that the olfactory bulbs (OBs) function as an independent circadian system regulating daily rhythms in olfactory performance. However, the cells and signals in the olfactory system that generate and coordinate these circadian rhythms are unknown. Using real-time imaging of gene expression, we found that the isolated olfactory epithelium and OB, but not the piriform cortex, express similar, sustained circadian rhythms in PERIOD2 (PER2). In vivo, PER2 expression in the OB of mice is circadian, approximately doubling with a peak around subjective dusk. Furthermore, mice exhibit circadian rhythms in odor detection performance with a peak at approximately subjective dusk. We also found that circadian rhythms in gene expression and odor detection performance require vasoactive intestinal polypeptide (VIP) or its receptor VPAC2R. VIP is expressed, in a circadian manner, in interneurons in the external plexiform and periglomerular layers, whereas VPAC2R is expressed in mitral and external tufted cells in the OB. Together, these results indicate that VIP signaling modulates the output from the OB to maintain circadian rhythms in the mammalian olfactory system.


Subject(s)
Circadian Rhythm/physiology , Olfactory Bulb/metabolism , Olfactory Pathways/metabolism , Smell/physiology , Vasoactive Intestinal Peptide/metabolism , Animals , Cerebral Cortex/metabolism , Male , Mice , Motor Activity/physiology , Olfactory Mucosa/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism
13.
Proc Natl Acad Sci U S A ; 110(46): E4355-61, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24167276

ABSTRACT

Shift work or transmeridian travel can desynchronize the body's circadian rhythms from local light-dark cycles. The mammalian suprachiasmatic nucleus (SCN) generates and entrains daily rhythms in physiology and behavior. Paradoxically, we found that vasoactive intestinal polypeptide (VIP), a neuropeptide implicated in synchrony among SCN cells, can also desynchronize them. The degree and duration of desynchronization among SCN neurons depended on both the phase and the dose of VIP. A model of the SCN consisting of coupled stochastic cells predicted both the phase- and the dose-dependent response to VIP and that the transient phase desynchronization, or "phase tumbling", could arise from intrinsic, stochastic noise in small populations of key molecules (notably, Period mRNA near its daily minimum). The model also predicted that phase tumbling following brief VIP treatment would accelerate entrainment to shifted environmental cycles. We tested this using a prepulse of VIP during the day before a shift in either a light cycle in vivo or a temperature cycle in vitro. Although VIP during the day does not shift circadian rhythms, the VIP pretreatment approximately halved the time required for mice to reentrain to an 8-h shifted light schedule and for SCN cultures to reentrain to a 10-h shifted temperature cycle. We conclude that VIP below 100 nM synchronizes SCN cells and above 100 nM reduces synchrony in the SCN. We show that exploiting these mechanisms that transiently reduce cellular synchrony before a large shift in the schedule of daily environmental cues has the potential to reduce jet lag.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Models, Biological , Signal Transduction/physiology , Suprachiasmatic Nucleus/physiology , Vasoactive Intestinal Peptide/metabolism , Animals , Biological Clocks/drug effects , Circadian Rhythm/drug effects , Dose-Response Relationship, Drug , Luminescent Measurements , Male , Mice , Motor Activity/physiology , Period Circadian Proteins/metabolism , Photoperiod , Temperature , Vasoactive Intestinal Peptide/pharmacology
14.
Exp Neurol ; 243: 21-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23099412

ABSTRACT

Daily rhythms in neural activity underlie circadian rhythms in sleep-wake and other daily behaviors. The cells within the mammalian suprachiasmatic nucleus (SCN) are intrinsically capable of 24-h timekeeping. These cells synchronize with each other and with local environmental cycles to drive coherent rhythms in daily behaviors. Recent studies have identified a small number of neuropeptides critical for this ability to synchronize and sustain coordinated daily rhythms. This review highlights the roles of specific intracellular and intercellular signals within the SCN that underlie circadian synchrony.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Suprachiasmatic Nucleus/physiology , Animals , Humans , Signal Transduction/physiology , Sleep/physiology , Sleep Wake Disorders/physiopathology
15.
J Neurosci ; 32(29): 10045-52, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22815518

ABSTRACT

Neurons in the suprachiasmatic nucleus (SCN) display coordinated circadian changes in electrical activity that are critical for daily rhythms in physiology, metabolism, and behavior. SCN neurons depolarize spontaneously and fire repetitively during the day and hyperpolarize, drastically reducing firing rates, at night. To explore the hypothesis that rapidly activating and inactivating A-type (I(A)) voltage-gated K(+) (Kv) channels, which are also active at subthreshold membrane potentials, are critical regulators of the excitability of SCN neurons, we examined locomotor activity and SCN firing in mice lacking Kv1.4 (Kv1.4(-/-)), Kv4.2 (Kv4.2(-/-)), or Kv4.3 (Kv4.3(-/-)), the pore-forming (α) subunits of I(A) channels. Mice lacking either Kv1.4 or Kv4.2 α subunits have markedly shorter (0.5 h) periods of locomotor activity than wild-type (WT) mice. In vitro extracellular multi-electrode recordings revealed that Kv1.4(-/-) and Kv4.2(-/-) SCN neurons display circadian rhythms in repetitive firing, but with shorter periods (0.5 h) than WT cells. In contrast, the periods of wheel-running activity in Kv4.3(-/-) mice and firing in Kv4.3(-/-) SCN neurons were indistinguishable from WT animals and neurons. Quantitative real-time PCR revealed that the transcripts encoding all three Kv channel α subunits, Kv1.4, Kv4.2, and Kv4.3, are expressed constitutively throughout the day and night in the SCN. Together, these results demonstrate that Kv1.4- and Kv4.2-encoded I(A) channels regulate the intrinsic excitability of SCN neurons during the day and night and determine the period and amplitude of circadian rhythms in SCN neuron firing and locomotor behavior.


Subject(s)
Action Potentials/physiology , Circadian Rhythm/physiology , Kv1.4 Potassium Channel/metabolism , Motor Activity/physiology , Neurons/physiology , Shal Potassium Channels/metabolism , Suprachiasmatic Nucleus/physiology , Animals , Ion Channel Gating/physiology , Kv1.4 Potassium Channel/genetics , Male , Membrane Potentials/physiology , Mice , Mice, Knockout , Shal Potassium Channels/genetics
16.
Essays Biochem ; 49(1): 103-17, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21819387

ABSTRACT

In vitro assays have localized circadian pacemakers to individual cells, revealed genetic determinants of rhythm generation, identified molecular players in cell-cell synchronization and determined physiological events regulated by circadian clocks. Although they allow strict control of experimental conditions and reduce the number of variables compared with in vivo studies, they also lack many of the conditions in which cellular circadian oscillators normally function. The present review highlights methods to study circadian timing in cultured mammalian cells and how they have shaped the hypothesis that all cells are capable of circadian rhythmicity.


Subject(s)
Astrocytes/physiology , Circadian Rhythm/physiology , Molecular Imaging/methods , Neurons/physiology , Suprachiasmatic Nucleus/physiology , Action Potentials/physiology , Animals , Cell Line, Tumor , Cells, Cultured , Circadian Clocks/physiology , Electrophysiological Phenomena , Humans , Neurons/metabolism
17.
J Biol Rhythms ; 26(6): 552-60, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22215613

ABSTRACT

The suprachiasmatic nucleus (SCN) regulates a wide range of daily behaviors and has been described as the master circadian pacemaker. The role of daily rhythmicity in other tissues, however, is unknown. We hypothesized that circadian changes in olfactory discrimination depend on a genetic circadian oscillator outside the SCN. We developed an automated assay to monitor olfactory discrimination in individual mice throughout the day. We found olfactory sensitivity increased approximately 6-fold from a minimum during the day to a peak in the early night. This circadian rhythm was maintained in SCN-lesioned mice and mice deficient for the Npas2 gene but was lost in mice lacking Bmal1 or both Per1 and Per2 genes. We conclude that daily rhythms in olfactory sensitivity depend on the expression of canonical clock genes. Olfaction is, thus, the first circadian behavior that is not based on locomotor activity and does not require the SCN.


Subject(s)
CLOCK Proteins/genetics , Circadian Rhythm/physiology , Smell/physiology , Suprachiasmatic Nucleus/physiology , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biological Clocks/physiology , CLOCK Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Suprachiasmatic Nucleus/pathology
18.
J Neurosci ; 26(47): 12219-25, 2006 Nov 22.
Article in English | MEDLINE | ID: mdl-17122046

ABSTRACT

Recently, it has been shown that multiple mammalian cell types express daily rhythms in vitro. Although the suprachiasmatic nucleus (SCN) of the hypothalamus is known to regulate a wide range of circadian behaviors, the role for intrinsic rhythmicity in other tissues is unknown. We tested whether the main olfactory bulb (OB) of mice mediates daily changes in olfaction. We found circadian rhythms in cedar oil-induced c-Fos, a protein marker of cellular excitation, in the mitral and granular layers of the OB and in the piriform cortex (PC). These oscillations persisted in constant darkness with a fourfold change in amplitude and a peak approximately 4 h after the onset of daily locomotor activity. Electrolytic lesions of the SCN abolished circadian locomotor rhythms, but not odor-induced c-Fos rhythms in the OB or PC. Furthermore, removal of the OB abolished spontaneous circadian cycling of c-Fos in the PC, shortened the free-running period of locomotor rhythms, and accelerated re-entrainment after a 6 h advance and slowed re-entrainment after a 6 h delay in the light schedule. OB ablation or odorant altered the amplitude of c-Fos rhythms in the SCN and ablation of one OB abolished c-Fos rhythms in the ipsilateral PC, but not in the contralateral OB and PC. We conclude that the OB comprises a master circadian pacemaker, which enhances olfactory responsivity each night, drives rhythms in the PC, and interacts with the SCN to coordinate other daily behaviors.


Subject(s)
Circadian Rhythm/physiology , Olfactory Bulb/physiology , Olfactory Pathways/physiology , Smell/physiology , Analysis of Variance , Animals , Behavior, Animal/physiology , Cell Count/methods , Circadian Rhythm/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Odorants , Olfactory Bulb/cytology , Olfactory Bulb/drug effects , Olfactory Bulb/injuries , Parietal Lobe/cytology , Parietal Lobe/physiology , Proto-Oncogene Proteins c-fos/metabolism , Suprachiasmatic Nucleus/injuries , Suprachiasmatic Nucleus/physiology , Time Factors
19.
J Neurosci ; 25(38): 8620-6, 2005 Sep 21.
Article in English | MEDLINE | ID: mdl-16177029

ABSTRACT

Behavioral and physiological circadian rhythms in mammals are controlled by a master pacemaker in the hypothalamic suprachiasmatic nuclei (SCN). Recently, circadian oscillations of hormone secretion, clock gene expression, and electrical activity have been demonstrated in explants of other brain regions. This suggests that some extra-SCN brain regions contain a functional, SCN-independent circadian clock, but in vivo evidence for intrinsic pacemaking is still lacking. We developed a novel method to image bioluminescence in vivo from the main olfactory bulbs (OB) of intact and SCN-lesioned (SCNX) Period1::luciferase rats for 2 d in constant darkness. The OBs expressed circadian rhythms in situ with a reliable twofold increase from day to night, similar to the phase and amplitude of ex vivo rhythms. In vivo cycling persisted for at least 1 month in the absence of the SCN. To assess indirectly in vivo rhythmicity of other brain areas, we measured the phase-dependence of their in vitro rhythms on the time of surgery. Surgery reliably reset the phase of the pineal gland and vascular organ of the lamina terminalis (VOLT) harvested from SCNX rats but had little effect on the phase of the OB. We deduce that the SCN and OB contain self-sustained circadian oscillators, whereas the pineal gland and VOLT are weak oscillators that require input from the SCN to show coordinated circadian rhythms. We conclude that the mammalian brain comprises a diverse set of SCN-dependent and SCN-independent circadian oscillators.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Nuclear Proteins/metabolism , Olfactory Bulb/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Animals, Genetically Modified , Cell Cycle Proteins , Darkness , Male , Mice , Motor Activity , Nuclear Proteins/physiology , Olfactory Bulb/chemistry , Period Circadian Proteins , Rats , Suprachiasmatic Nucleus/chemistry
20.
Eur J Neurosci ; 19(4): 898-906, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15009137

ABSTRACT

Circadian pacemakers drive many daily molecular, physiological and behavioural rhythms. We investigated whether the main olfactory bulb is a functional circadian pacemaker in rats. Long-term, multielectrode recordings revealed that individual, cultured bulb neurons expressed near 24-h oscillations in firing rate. Real-time recordings of Period1 gene activity showed that a population of cells within the bulb expressed synchronized rhythmicity starting on embryonic day 19. This rhythmicity was intrinsic to the mitral, and not the granule, cell layer, entrainable to physiological temperature cycles and temperature compensated in its period. However, removal of the olfactory bulbs had no effect on running wheel behaviour. These results indicate that individual mitral/tufted cells are competent circadian pacemakers which normally synchronize to each other. The daily rhythms in gene expression and firing rate intrinsic to the olfactory bulb are not required for circadian patterns of locomotion, indicating that they are involved in rhythms outside the canonical circadian system.


Subject(s)
Circadian Rhythm/physiology , Neurons/metabolism , Olfactory Bulb/metabolism , Animals , Animals, Genetically Modified , Animals, Newborn , Cell Cycle Proteins , Female , Gene Expression Regulation/physiology , Male , Motor Activity/physiology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Period Circadian Proteins , Pregnancy , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...