201
|
Ribeiro AC, Kapás L. Day- and nighttime injection of a nitric oxide synthase inhibitor elicits opposite sleep responses in rats. Am J Physiol Regul Integr Comp Physiol 2005; 289:R521-R531. [PMID: 15860646 DOI: 10.1152/ajpregu.00605.2004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies suggest that nitric oxide (NO) may play a role in sleep regulation, particularly in the homeostatic process. The present studies were undertaken to compare the sleep effects of injecting a NO synthase (NOS) inhibitor when homeostatic sleep pressure is naturally highest (light onset) or when it is at its nadir (dark onset) in rats. Sleep, electroencephalogram delta-wave activity during nonrapid eye movement sleep (NREMS), also known as slow-wave activity (SWA), and brain temperature responses to three doses of the NOS inhibitor Nω-nitro-l-arginine methyl ester (l-NAME; 5, 50, and 100 mg/kg) injected intraperitoneally at light or dark onset were examined in rats ( n = 6 to 8). The effects of 5 mg/kg l-NAME were determined in both normal and vagotomized (VX) rats. Light onset administration of 50 mg/kg l-NAME decreased NREMS amounts and suppressed SWA and increased rapid eye movement sleep (REMS) amounts. At dark onset, l-NAME injection also dose dependently suppressed SWA; however, unlike light onset injections, both NREMS and REMS amounts were increased after all three doses. Sleep responses to 5 mg/kg l-NAME were not different in control and VX rats, suggesting that the sleep effects of l-NAME are not mediated through the activation of sensory vagal mechanisms. The present findings suggest that timing of the injection is a major determinant of the sleep responses observed after systemic l-NAME injection in rats.
Collapse
Affiliation(s)
- Ana C Ribeiro
- Dept. of Biological Sciences, Fordham University, 441 E. Fordham Road, Bronx, NY 10458, USA
| | | |
Collapse
|
202
|
Engel L, Lorenzkowski V, Langer C, Rohleder N, Spessert R. The photoperiod entrains the molecular clock of the rat pineal. Eur J Neurosci 2005; 21:2297-304. [PMID: 15869528 DOI: 10.1111/j.1460-9568.2005.04040.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The suprachiasmatic nucleus-pineal system acts as a neuroendocrine transducer of seasonal changes in the photoperiod by regulating melatonin formation. In the present study, we have investigated the extent to which the photoperiod entrains the nonself-cycling oscillator in the Sprague-Dawley rat pineal. For this purpose, the 24-h expression of nine clock genes (bmal1, clock, per1, per2, per3, cry1, cry2, dec1 and dec2) and the aa-nat gene was monitored under light-dark 8 : 16 and light-dark 16 : 8 in the rat pineal by using real-time RT-PCR. The 24-h pattern of the expression of only per1, dec2 and aa-nat genes was affected by photoperiod. In comparison with the short photoperiod, the duration of elevated expression under the long photoperiod was elongated for per1 and shortened for dec2 and aa-nat. For each of the genes, photoperiod-dependent variations partly persisted under constant darkness. Therefore, the pineal clockwork appears to memorize the photoperiod of prior entrained cycles. The findings of the present study indicate that the nonself-cycling oscillator of the rat pineal is entrained by photoperiodic information and therefore that it participates in seasonal timekeeping.
Collapse
Affiliation(s)
- Lydia Engel
- Department of Anatomy, Johannes Gutenberg University, Saarstrasse 19-21, D-55099 Mainz, Germany
| | | | | | | | | |
Collapse
|
203
|
Feil R, Hofmann F, Kleppisch T. Function of cGMP-dependent protein kinases in the nervous system. Rev Neurosci 2005; 16:23-41. [PMID: 15810652 DOI: 10.1515/revneuro.2005.16.1.23] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The second messenger cyclic guanosine-3',5'-monophosphate (cGMP) mediates many effects of nitric oxide in the nervous system. cGMP may act through various intracellular receptors, among them a family of serine/threonine kinases, the cGMP-dependent protein kinases (cGKs). Hitherto, three mammalian cGKs have been identified: cGKIalpha, cGKIbeta and cGKII. Discrete functions of cGKI and cGKII are determined by their distinct expression patterns and targeting to specific substrates. This review provides an overview about the expression and functions of cGKs in the nervous system. Main emphasis is put on the discussion of phenotypes observed in cGK-deficient mouse models that lack cGKI and/or cGKII globally or selectively in brain regions of interest. Recent data demonstrate important functions of cGKI in (1) the development and sensitization of nociceptive neurons, and (2) synaptic plasticity and learning. There is also evidence suggesting that cGKII in the suprachiasmatic nucleus of the hypothalamus is involved in the regulation of circadian rhythmicity. Thus, cGKs serve key functions in the transduction of cGMP signals into cellular responses in distinct regions of the nervous system.-
Collapse
Affiliation(s)
- Robert Feil
- Institut für Pharmakologie und Toxikologie, Technische Universität München, München, Germany
| | | | | |
Collapse
|
204
|
Abstract
In mammals, circadian rhythms are driven by a pacemaker located in the suprachiasmatic nucleus (SCN) of the hypothalamus. We measured the rhythm of arginine vasopressin release in rat organotypic SCN slices following application of tetrodotoxin (TTX) or N-methyl-D-aspartate (NMDA) at various times throughout the circadian cycle. TTX resets the clock in a manner similar to dark pulses. A 4-h application of TTX starting in mid subjective day, at around circadian time (CT) 7.0, induced phase advances, while TTX treatment started in early subjective morning, at about CT 2.0, induced phase delays. On the other hand, NMDA resets the clock in a manner similar to a light pulse; that is, NMDA treatment in the early evening induced phase delays while treatment in the late night induced phase advances. The data indicate that deprivation of neuronal firing changes the circadian rhythm.
Collapse
Affiliation(s)
- Takako Noguchi
- Department of Physiology, Dokkyo University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan
| | | |
Collapse
|
205
|
Buchanan GF, Gillette MU. New light on an old paradox: site-dependent effects of carbachol on circadian rhythms. Exp Neurol 2005; 193:489-96. [PMID: 15869951 DOI: 10.1016/j.expneurol.2005.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2004] [Revised: 12/30/2004] [Accepted: 01/13/2005] [Indexed: 10/25/2022]
Abstract
Acetylcholine (ACh) was the first neurotransmitter identified as a regulator of mammalian circadian rhythms. When injected in vivo, cholinergics induced biphasic clock resetting at night, similar to nocturnal light exposure. However, the retinohypothalamic tract connecting the eye to the suprachiasmatic nucleus (SCN) uses glutamate (GLU) to transmit light signals. We here resolve this long-standing paradox. Whereas injection of the cholinergic agonist, carbachol, into the mouse ventricular system in vivo induced light-like effects, direct application to the SCN in vitro or in vivo induced a distinct response pattern: phase advance of circadian rhythms throughout the nighttime. These results indicate that a new regulatory pathway, involving an extra-SCN cholinergic synapse accessible via ventricular injection, mediates the light-like cholinergic clock resetting reported previously.
Collapse
Affiliation(s)
- Gordon F Buchanan
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | |
Collapse
|
206
|
Menger GJ, Lu K, Thomas T, Cassone VM, Earnest DJ. Circadian profiling of the transcriptome in immortalized rat SCN cells. Physiol Genomics 2005; 21:370-81. [PMID: 15769907 DOI: 10.1152/physiolgenomics.00224.2004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Endogenous oscillations in gene expression are a prevalent feature of the circadian clock in the mammalian suprachiasmatic nucleus (SCN) and similar timekeeping systems in other organisms. To determine whether immortalized cells derived from the rat SCN (SCN2.2) retain these intrinsic rhythm-generating properties, oscillatory behavior of the SCN2.2 transcriptome was analyzed and compared with that found in the rat SCN in vivo using rat U34A Affymetrix GeneChips. In SCN2.2 cells, 116 unique genes and 46 ESTs or genes of unknown function exhibited circadian fluctuations with a 1.5-fold or greater difference in their mRNA abundance for two cycles. Many (35%) of these rhythmically regulated genes in SCN2.2 cells also exhibited circadian profiles of mRNA expression in the rat SCN in vivo. Functional analyses and cartography indicate that a diverse set of cellular pathways are strategically regulated by the circadian clock in SCN2.2 cells and that the largest categories of rhythmic genes are those involved in cellular and systems-level communication or in metabolic processes like cellular respiration, fatty acid recycling, and steroid synthesis. Because many of the same genes or nodes within these functional categories were rhythmically expressed in both SCN2.2 cells and the rat SCN, the circadian regulation of these pathways may be important in modulating input to or output from the SCN clock mechanism. In summary, global expression and circadian regulation of the SCN2.2 transcriptome retain many SCN-like properties, suggesting that genes displaying rhythmic profiles in both experimental models may be integral to their function as both circadian oscillators and pacemakers.
Collapse
Affiliation(s)
- Gus J Menger
- Department of Human Anatomy and Medical Neurobiology, Texas A & M University Health Science Center, College of Medicine, Texas 77843-1114, USA
| | | | | | | | | |
Collapse
|
207
|
Regulation of prokineticin 2 expression by light and the circadian clock. BMC Neurosci 2005; 6:17. [PMID: 15762991 PMCID: PMC555564 DOI: 10.1186/1471-2202-6-17] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Accepted: 03/11/2005] [Indexed: 11/10/2022] Open
Abstract
Background The suprachiasmatic nucleus (SCN) contains the master circadian clock that regulates daily rhythms of many physiological and behavioural processes in mammals. Previously we have shown that prokineticin 2 (PK2) is a clock-controlled gene that may function as a critical SCN output molecule responsible for circadian locomotor rhythms. As light is the principal zeitgeber that entrains the circadian oscillator, and PK2 expression is responsive to nocturnal light pulses, we further investigated the effects of light on the molecular rhythm of PK2 in the SCN. In particular, we examined how PK2 responds to shifts of light/dark cycles and changes in photoperiod. We also investigated which photoreceptors are responsible for the light-induced PK2 expression in the SCN. To determine whether light requires an intact functional circadian pacemaker to regulate PK2, we examined PK2 expression in cryptochrome1,2-deficient (Cry1-/-Cry2-/-) mice that lack functional circadian clock under normal light/dark cycles and constant darkness. Results Upon abrupt shifts of the light/dark cycle, PK2 expression exhibits transients in response to phase advances but rapidly entrains to phase delays. Photoperiod studies indicate that PK2 responds differentially to changes in light period. Although the phase of PK2 expression expands as the light period increases, decreasing light period does not further condense the phase of PK2 expression. Genetic knockout studies revealed that functional melanopsin and rod-cone photoreceptive systems are required for the light-inducibility of PK2. In Cry1-/-Cry2-/- mice that lack a functional circadian clock, a low amplitude PK2 rhythm is detected under light/dark conditions, but not in constant darkness. This suggests that light can directly regulate PK2 expression in the SCN. Conclusion These data demonstrate that the molecular rhythm of PK2 in the SCN is regulated by both the circadian clock and light. PK2 is predominantly controlled by the endogenous circadian clock, while light plays a modulatory role. The Cry1-/-Cry2-/- mice studies reveal a light-driven PK2 rhythm, indicating that light can induce PK2 expression independent of the circadian oscillator. The light inducibility of PK2 suggests that in addition to its role in clock-driven rhythms of locomotor behaviour, PK2 may also participate in the photic entrainment of circadian locomotor rhythms.
Collapse
|
208
|
Sprouse J. Pharmacological modulation of circadian rhythms: a new drug target in psychotherapeutics. Expert Opin Ther Targets 2005; 8:25-38. [PMID: 14996616 DOI: 10.1517/14728222.8.1.25] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Daily variation in an organism's physiology and behaviour is regulated by the synchrony that is achieved between the internal timing mechanisms - the circadian rhythms of the biological clock - and the prevailing environmental cues. Proper synchrony constitutes an adaptive response; improper or lost synchrony may well yield maladaptation and, in the case of humans, a psychiatric disorder. On a basic level, the circadian system is comprised of three parts: a central oscillator, its various neuronal inputs and its outputs. For all three of these parts, the dissemination of new information is moving at an unprecedented pace, and the number of molecular targets for the opportunistic pharmacologist is growing in step. Monoamines, neuropeptides, kinases - sorting through all these, much less developing one into a drug discovery programme, may be the biggest challenge. However, the potential benefits in targeting a basic flaw in a fundamental biological system may be enormous.
Collapse
Affiliation(s)
- Jeffrey Sprouse
- Pfizer Global Research & Development, Groton, CT 06340, USA.
| |
Collapse
|
209
|
Kim DY, Choi HJ, Kim JS, Kim YS, Jeong DU, Shin HC, Kim MJ, Han HC, Hong SK, Kim YI. Voltage-gated calcium channels play crucial roles in the glutamate-induced phase shifts of the rat suprachiasmatic circadian clock. Eur J Neurosci 2005; 21:1215-22. [PMID: 15813931 DOI: 10.1111/j.1460-9568.2005.03950.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The resetting of the circadian clock based on photic cues delivered by the glutamatergic retinohypothalamic tract is an important process helping mammals to function adaptively to the daily light-dark cycle. To see if the photic resetting relies on voltage-gated Ca(2+) channels (VGCCs), we examined the effects of VGCC blockers on the glutamate-induced phase shifts of circadian firing activity rhythms of suprachiasmatic nucleus (SCN) neurons in hypothalamic slices. First, we found that a cocktail of amiloride, nimodipine and omega-conotoxin MVIIC (T-, L- and NPQ-type VGCC antagonists, respectively) completely blocked both phase delays and advances, which were, respectively, induced by glutamate application in early and late night. Next, we discovered that: (i) amiloride and another T-type VGCC antagonist, mibefradil, completely obstructed the delays without affecting the advances; (ii) nimodipine completely blocked the advances while having less impact on delays; and (iii) omega-conotoxin MVIIC blocked largely, if not entirely, both delays and advances. Subsequent whole-cell recordings revealed that T-type Ca(2+) currents in neurons in the ventrolateral, not dorsomedial, region of the SCN were larger during early than late night, whereas L-type Ca(2+) currents did not differ from early to late night in both regions. These results indicate that VGCCs play important roles in glutamate-induced phase shifts, T-type being more important for phase delays and L-type being so for phase advances. Moreover, the results point to the possibility that a nocturnal modulation of T-type Ca(2+) current in retinorecipient neurons is related to the differential involvement of T-type VGCC in phase delays and advances.
Collapse
Affiliation(s)
- Do Young Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul, Korea 136-705.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Barakat MT, O'Hara BF, Cao VH, Larkin JE, Heller HC, Ruby NF. Light pulses do not induce c-fos or per1 in the SCN of hamsters that fail to reentrain to the photocycle. J Biol Rhythms 2005; 19:287-97. [PMID: 15245648 DOI: 10.1177/0748730404266771] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Circadian activity rhythms of most Siberian hamsters (Phodopus sungorus sungorus) fail to reentrain to a 5-h phase shift of the light-dark (LD) cycle. Instead, their rhythms free-run at periods close to 25 h despite the continued presence of the LD cycle. This lack of behavioral reentrainment necessarily means that molecular oscillators in the master circadian pacemaker, the SCN, were unable to reentrain as well. The authors tested the hypothesis that a phase shift of the LD cycle rendered the SCN incapable of responding to photic input. Animals were exposed to a 5-h phase delay of the photocycle, and activity rhythms were monitored until a lack of reentrainment was confirmed. Hamsters were then housed in constant darkness for 24 h and administered a 30-min light pulse 2 circadian hours after activity onset. Brains were then removed, and tissue sections containing the SCN were processed for in situ hybridization. Sections were probed with Siberian hamster c-fos and per1 mRNA probes because light rapidly induces these 2 genes in the SCN during subjective night but not at other circadian phases. Light pulses induced robust expression of both genes in all animals that reentrained to the LD cycle, but no expression was observed in any animal that failed to reentrain. None of the animals exhibited an intermediate response. This finding is the first report of acute shift in a photocycle eliminating photosensitivity in the SCN and suggests that a specific pattern of light exposure may desensitize the SCN to subsequent photic input.
Collapse
Affiliation(s)
- Monique T Barakat
- Department of Biological Sciences, 371 Serra Mall, Stanford University, Stanford, CA 94305-5020, USA.
| | | | | | | | | | | |
Collapse
|
211
|
Farnell YZ, West JR, Chen WJA, Allen GC, Earnest DJ. Developmental alcohol exposure alters light-induced phase shifts of the circadian activity rhythm in rats. Alcohol Clin Exp Res 2005; 28:1020-7. [PMID: 15252288 PMCID: PMC2695982 DOI: 10.1097/01.alc.0000130807.21020.1b] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Developmental alcohol (EtOH) exposure produces long-term changes in the photic regulation of rat circadian behavior. Because entrainment of circadian rhythms to 24-hr light/dark cycles is mediated by phase shifting or resetting the clock mechanism, we examined whether developmental EtOH exposure also alters the phase-shifting effects of light pulses on the rat activity rhythm. METHODS Artificially reared Sprague-Dawley rat pups were exposed to EtOH (4.5 g/kg/day) or an isocaloric milk formula (gastrostomy control; GC) on postnatal days 4 to 9. At 2 months of age, rats from the EtOH, GC, and suckle control groups were housed individually, and wheel-running behavior was continuously recorded first in a 12-hr light/12-hr dark photoperiod for 10 to 14 days and thereafter in constant darkness (DD). Once the activity rhythm was observed to stably free-run in DD for at least 14 days, animals were exposed to a 15-min light pulse at either 2 or 10 hr after the onset of activity [i.e., circadian time (CT) 14 or 22, respectively], because light exposure at these times induces maximal phase delays or advances of the rat activity rhythm. RESULTS EtOH-treated rats were distinguished by robust increases in their phase-shifting responses to light. In the suckle control and GC groups, light pulses shifted the activity rhythm as expected, inducing phase delays of approximately 2 hr at CT 14 and advances of similar amplitude at CT 22. In contrast, the same light stimulus produced phase delays at CT 14 and advances at CT 22 of longer than 3 hr in EtOH-treated rats. The mean phase delay at CT 14 and advance at CT 22 in EtOH rats were significantly greater (p < 0.05) than the light-induced shifts observed in control animals. CONCLUSIONS The data indicate that developmental EtOH exposure alters the phase-shifting responses of the rat activity rhythm to light. This finding, coupled with changes in the circadian period and light/dark entrainment observed in EtOH-treated rats, suggests that developmental EtOH exposure may permanently alter the clock mechanism in the suprachiasmatic nucleus and its regulation of circadian behavior.
Collapse
Affiliation(s)
- Yuhua Z Farnell
- Department of Human Anatomy and Medical Neurobiology, Texas A&M University System Health Science Center, College of Medicine, College Station, Texas 77843-1114, USA
| | | | | | | | | |
Collapse
|
212
|
Tischkau SA, Gillette MU. Oligodeoxynucleotide methods for analyzing the circadian clock in the suprachiasmatic nucleus. Methods Enzymol 2005; 393:593-610. [PMID: 15817314 DOI: 10.1016/s0076-6879(05)93031-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The recent identification of specific genes responsible for the generation of endogenous circadian rhythmicity in the suprachiasmatic nucleus presents a new level of investigation into endogenous rhythmicity and mechanisms of synchronization of this circadian clock with the environmental light?dark cycle. This article describes techniques that employ antisense and decoy oligodeoxynucleotides (ODN) to determine the roles of specific molecular substrates both in endogenous rhythmicity and in regulating the effects of light on the mammalian circadian clock. Application of antisense ODN technology has revealed a role for timeless (Tim) in the core clock mechanism and established that induction of period1 (Per1) is required for light responsiveness. Likewise, a decoy ODN designed to sequester activated CREB protein definitively demonstrated a requirement for CRE-mediated transcription in light signaling. Experiments designed with these molecular tools offer new insights on the interaction of cellular processes and signaling with the molecular clockworks.
Collapse
Affiliation(s)
- Shelley A Tischkau
- Department of Cell & Structural Biology and Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 60801, USA
| | | |
Collapse
|
213
|
Abstract
Circadian clock systems are composed of an input or "entrainment" pathway by which synchronization to the external environment occurs, a pacemaker responsible for generating rhythmicity, and an output or "expression" pathway through which rhythmic signals act to modulate physiology and behavior. The circadian pacemaker contains molecular feedback loops of rhythmically expressed genes and their protein products, which, through interactions, generate a circa 24-h cycle of transcription and translation of clock and clock-controlled genes. Neuronal membrane events appear to play major roles in entrainment of circadian rhythms in mollusks and mammals. In mammals, the suprachiasmatic nuclei of the hypothalamus receive photic information via the retinohypothalamic tract. Retinal signals, mediated by glutamate, induce calcium release and activate a number of intracellular cascades involved in photic gating and phase shifting. Membrane events are also involved in rhythm expression. Calcium and potassium currents influence the electrical output of pacemaker neurons by altering shape and intervals of impulse prepotentials, afterhyperpolarization periods, and interspike intervals, as well as altering membrane potentials and thereby shaping the spontaneous rhythmic spiking patterns. Unlike the involvement of membrane events in circadian entrainment and expression, it is less clear whether electrical activity, postsynaptic events, and transmembrane ion fluxes also are essential elements in rhythm generation. Studies, however, suggest that neuronal membrane activity may indeed play a crucial role in circadian rhythm generation.
Collapse
Affiliation(s)
- Gabriella B Lundkvist
- Department of Biology, Center for Biological Timing, University of Virginia, Charlottesville, Virginia 22903, USA
| | | |
Collapse
|
214
|
Pardee K, Reinking J, Krause H. Nuclear Hormone Receptors, Metabolism, and Aging: What Goes Around Comes Around. ACTA ACUST UNITED AC 2004; 2004:re8. [PMID: 15564562 DOI: 10.1126/sageke.2004.47.re8] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Previous studies have linked the mysterious and inevitable process of aging to essential processes such as metabolism, maturation, and fecundity. Each of these processes is controlled to a large extent by nuclear hormone receptors (NHRs). NHRs also play important roles in the control of periodical processes, the most recently implicated being circadian rhythm. This Review stresses the mounting evidence for tight relationships between each of these NHR-regulated processes and the processes of aging.
Collapse
Affiliation(s)
- Keith Pardee
- Banting and Best Department of Medical Research, University of Toronto, Charles H. Best Institute, Toronto, Ontario, Canada
| | | | | |
Collapse
|
215
|
Rodrigo R, Montoliu C, Chatauret N, Butterworth R, Behrends S, Del Olmo JA, Serra MA, Rodrigo JM, Erceg S, Felipo V. Alterations in soluble guanylate cyclase content and modulation by nitric oxide in liver disease. Neurochem Int 2004; 45:947-53. [PMID: 15312989 DOI: 10.1016/j.neuint.2004.03.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2003] [Revised: 01/30/2004] [Accepted: 02/11/2004] [Indexed: 11/17/2022]
Abstract
Hyperammonemia is the main responsible for the neurological alterations in hepatic encephalopathy in patients with liver failure. We studied the function of the glutamate-nitric oxide (NO)-cGMP pathway in brain in animal models of hyperammonemia and liver failure and in patients died with liver cirrhosis. Activation of glutamate receptors increases intracellular calcium that binds to calmodulin and activates neuronal nitric oxide synthase, increasing nitric oxide, which activates soluble guanylate cyclase (sGC), increasing cGMP. This glutamate-NO-cGMP pathway modulates cerebral processes such as circadian rhythms, the sleep-waking cycle, and some forms of learning and memory. These processes are impaired in patients with hepatic encephalopathy. Activation of sGC by NO is significantly increased in cerebral cortex and significantly reduced in cerebellum from cirrhotic patients died in hepatic coma. Portacaval anastomosis in rats, an animal model of liver failure, reproduces the effects of liver failure on modulation of sGC by NO both in cerebral cortex and cerebellum. In vivo brain microdialisis studies showed that sGC activation by NO is also reduced in vivo in cerebellum in hyperammonemic rats with or without liver failure. The content of alpha but not beta subunits of sGC are increased both in frontal cortex and cerebellum from patients died due to liver disease and from rats with portacaval anastomosis. We assessed whether determination of activation of sGC by NO-generating agent SNAP in lymphocytes could serve as a peripheral marker for the impairment of sGC activation by NO in brain. Chronic hyperammonemia and liver failure also alter sGC activation by NO in lymphocytes from rats or patients. These findings show that the content and modulation by NO of sGC are strongly altered in brain of patients with liver disease. These alterations could be responsible for some of the neurological alterations in hepatic encephalopathy such as sleep disturbances and cognitive impairment.
Collapse
Affiliation(s)
- Regina Rodrigo
- Laboratory of Neurobiology, Fundación Valenciana de Investigaciones Biomédicas, Amadeo de Saboya 4, 46010 Valencia, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
216
|
Golombek DA, Agostino PV, Plano SA, Ferreyra GA. Signaling in the mammalian circadian clock: the NO/cGMP pathway. Neurochem Int 2004; 45:929-36. [PMID: 15312987 DOI: 10.1016/j.neuint.2004.03.023] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mammalian circadian rhythms are generated by a hypothalamic suprachiasmatic nuclei (SCN) clock. Light pulses synchronize body rhythms by inducing phase delays during the early night and phase advances during the late night. Phosphorylation events are known to be involved in circadian phase shifting, both for delays and advances. Pharmacological inhibition of the cGMP-dependent kinase (cGK) or Ca2+/calmodulin-dependent kinase (CaMK), or of neuronal nitric oxide synthase (nNOS) blocks the circadian responses to light in vivo. Light pulses administered during the subjective night, but not during the day, induce rapid phosphorylation of both p-CAMKII and p-nNOS (specifically phosphorylated by CaMKII). CaMKII inhibitors block light-induced nNOS activity and phosphorylation, suggesting a direct pathway between both enzymes. Furthermore, SCN cGMP exhibits diurnal and circadian rhythms with maximal values during the day or subjective day. This variation of cGMP levels appears to be related to temporal changes in phosphodiesterase (PDE) activity and not to guanylyl cyclase (GC) activity. Light pulses increase SCN cGMP levels at circadian time (CT) 18 (when light causes phase advances of rhythms) but not at CT 14 (the time for light-induced phase delays). cGK II is expressed in the hamster SCN and also exhibits circadian changes in its levels, peaking during the day. Light pulses increase cGK activity at CT 18 but not at CT 14. In addition, cGK and GC inhibition by KT-5823 and ODQ significantly attenuated light-induced phase shifts at CT 18. This inhibition did not change c-Fos expression SCN but affected the expression of the clock gene per in the SCN. These results suggest a signal transduction pathway responsible for light-induced phase advances of the circadian clock which could be summarized as follows: Glu-Ca2+-CaMKII-nNOS-GC-cGMP-cGK-->-->clock genes. This pathway offers a signaling window that allows peering into the circadian clock machinery in order to decipher its temporal cogs and wheels.
Collapse
Affiliation(s)
- Diego A Golombek
- Laboratory of Chronobiology, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Saenz Peña 180, Bernal (1876), Buenos Aires, Argentina.
| | | | | | | |
Collapse
|
217
|
Tischkau SA, Mitchell JW, Pace LA, Barnes JW, Barnes JA, Gillette MU. Protein kinase G type II is required for night-to-day progression of the mammalian circadian clock. Neuron 2004; 43:539-49. [PMID: 15312652 DOI: 10.1016/j.neuron.2004.07.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2003] [Revised: 04/12/2004] [Accepted: 07/22/2004] [Indexed: 10/26/2022]
Abstract
Circadian clocks comprise a cyclic series of dynamic cellular states, characterized by the changing availability of substrates that alter clock time when activated. To determine whether circadian clocks, like the cell cycle, exhibit regulation by key phosphorylation events, we examined endogenous kinase regulation of timekeeping in the mammalian suprachiasmatic nucleus (SCN). Short-term inhibition of PKG-II but not PKG-Ibeta using antisense oligodeoxynucleotides delayed rhythms of electrical activity and Bmal1 mRNA. Phase resetting was rapid and dynamic; inhibition of PKG-II forced repetition of the last 3.5 hr of the cycle. Chronic inhibition of PKG-II disrupted electrical activity rhythms and tonically increased Bmal1 mRNA. PKG-II-like immunoreactivity was detected after coimmunoprecipitation with CLOCK, and CLOCK was phosphorylated in the presence of active PKG-II. PKG-II activation may define a critical control point for temporal progression into the daytime domain by acting on the positive arm of the transcriptional/translational feedback loop.
Collapse
Affiliation(s)
- Shelley A Tischkau
- Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, B107 CLSL, 601 South Goodwin Avenue, Urbana, Illinois 61801, USA
| | | | | | | | | | | |
Collapse
|
218
|
Cheng HYM, Obrietan K, Cain SW, Lee BY, Agostino PV, Joza NA, Harrington ME, Ralph MR, Penninger JM. Dexras1 Potentiates Photic and Suppresses Nonphotic Responses of the Circadian Clock. Neuron 2004; 43:715-28. [PMID: 15339652 DOI: 10.1016/j.neuron.2004.08.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2004] [Revised: 04/01/2004] [Accepted: 08/10/2004] [Indexed: 10/25/2022]
Abstract
Circadian rhythms of physiology and behavior are generated by biological clocks that are synchronized to the cyclic environment by photic or nonphotic cues. The interactions and integration of various entrainment pathways to the clock are poorly understood. Here, we show that the Ras-like G protein Dexras1 is a critical modulator of the responsiveness of the master clock to photic and nonphotic inputs. Genetic deletion of Dexras1 reduces photic entrainment by eliminating a pertussis-sensitive circadian response to NMDA. Mechanistically, Dexras1 couples NMDA and light input to Gi/o and ERK activation. In addition, the mutation greatly potentiates nonphotic responses to neuropeptide Y and unmasks a nonphotic response to arousal. Thus, Dexras1 modulates the responses of the master clock to photic and nonphotic stimuli in opposite directions. These results identify a signaling molecule that serves as a differential modulator of the gated photic and nonphotic input pathways to the circadian timekeeping system.
Collapse
Affiliation(s)
- Hai-Ying M Cheng
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna.
| | | | | | | | | | | | | | | | | |
Collapse
|
219
|
Agostino PV, Ferreyra GA, Murad AD, Watanabe Y, Golombek DA. Diurnal, circadian and photic regulation of calcium/calmodulin-dependent kinase II and neuronal nitric oxide synthase in the hamster suprachiasmatic nuclei. Neurochem Int 2004; 44:617-25. [PMID: 15016477 DOI: 10.1016/j.neuint.2003.09.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2003] [Revised: 06/18/2003] [Accepted: 09/19/2003] [Indexed: 11/28/2022]
Abstract
Mammalian circadian rhythms are entrained by light pulses that induce phosphorylation events in the suprachiasmatic nuclei (SCN). Ca(2+)-dependent enzymes are known to be involved in circadian phase shifting. In this paper, we show that calcium/calmodulin-dependent kinase II (CaMKII) is rhythmically phosphorylated in the SCN both under entrained and free-running (constant dark) conditions while neuronal nitric oxide synthase (nNOS) is rhythmically phosphorylated in the SCN only under entrained conditions. Both p-CaMKII and p-NOS (specifically phosphorylated by CaMKII) levels peak during the day or subjective day. Light pulses administered during the subjective night, but not during the day, induced rapid phosphorylation of both enzymes. Moreover, we found an inhibitory effect of KN-62 and KN-93, both CaMKII inhibitors, on light-induced nNOS activity and nNOS phosphorylation respectively, suggesting a direct pathway between both enzymes which is at least partially responsible of photic circadian entrainment.
Collapse
Affiliation(s)
- Patricia V Agostino
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Saenz Peña 180, Bernal 1876, Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
220
|
Yannielli PC, Brewer JM, Harrington ME. Blockade of the NPY Y5 receptor potentiates circadian responses to light: complementary in vivo and in vitro studies. Eur J Neurosci 2004; 19:891-7. [PMID: 15009136 DOI: 10.1111/j.0953-816x.2004.03098.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Neuropeptide Y (NPY) is delivered to the suprachiasmatic nuclei (SCN) circadian pacemaker via an input from the thalamic intergeniculate leaflet. NPY can inhibit light-induced responses of the circadian system of Syrian hamsters. Here we studied whether an antagonist to NPY receptors can be used to potentiate photic phase shifts late in the subjective night. First we determined by in situ hybridization that both NPY Y1 and Y5 receptor mRNA are expressed in the SCN of Syrian hamsters. Second, similar to our previous findings at Zeitgeber time 14 (ZT 14, where ZT 12 was the time of lights off), we found that NPY applied at ZT 18.5 onto the SCN region of brain slices maintained in vitro could block NMDA-induced phase advances of the spontaneous firing rate rhythm, and this blocking effect was probably mediated by the Y5 receptor, since co-application of Y5 receptor antagonists completely reversed the effect of NPY, while application of a Y1 receptor antagonist had no effect under the same conditions. Third, we found that co-treatment with a Y5 receptor antagonist in vivo (s.c., 10 mg/kg) not only reversed the effect of NPY applied to the SCN in vivo through a cannula but also significantly potentiated the light-induced phase advance in the absence of NPY. This is the first report of a NPY receptor antagonist having such an effect, and indicates that NPY Y5 receptor antagonists could be clinically useful for potentiating circadian system responses to light.
Collapse
Affiliation(s)
- P C Yannielli
- Department of Psychology and Neuroscience Program, Smith College, Northampton, MA 01063, USA
| | | | | |
Collapse
|
221
|
Honma S, Nakamura W, Shirakawa T, Honma KI. Diversity in the circadian periods of single neurons of the rat suprachiasmatic nucleus depends on nuclear structure and intrinsic period. Neurosci Lett 2004; 358:173-6. [PMID: 15039109 DOI: 10.1016/j.neulet.2004.01.022] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2003] [Revised: 01/08/2004] [Accepted: 01/10/2004] [Indexed: 11/21/2022]
Abstract
The circadian periods of single cultured neurons of the hypothalamic suprachiasmatic nucleus (SCN) in rats were assessed by means of multi-electrode array dish. Although the mean circadian period was not different between the dispersed cell culture and organotypic slice culture, the periods distributed in a wide range from 20.0 to 30.9 h in the former whereas concentrated in a narrow range in the latter. The same difference was also detected within each culture dish. There is a significant correlation between the period length and variation of circadian rhythm, where the more the mean circadian period in a culture dish deviates from the overall mean, the larger the standard deviation of period in a dish becomes. Such a correlation was not observed in the organotypic slice culture. These findings indicate that the diversity of circadian periods in the individual SCN neurons depends on the maintenance of SCN structure and the circadian period, suggesting that not only cell-to-cell communication but also the intrinsic circadian period plays a significant role in synchronizing the constitutional oscillators in the SCN.
Collapse
Affiliation(s)
- Sato Honma
- Department of Physiology, Hokkaido University, Graduate School of Medicine, North-15, West-7, Kita-ku, Sapporo 060-8638, Japan.
| | | | | | | |
Collapse
|
222
|
Meyer-Spasche A, Piggins HD. Vasoactive intestinal polypeptide phase-advances the rat suprachiasmatic nuclei circadian pacemaker in vitro via protein kinase A and mitogen-activated protein kinase. Neurosci Lett 2004; 358:91-4. [PMID: 15026156 DOI: 10.1016/j.neulet.2003.12.114] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2003] [Accepted: 12/30/2003] [Indexed: 10/26/2022]
Abstract
The firing activity of suprachiasmatic nuclei (SCN) neurones in vitro shows a circadian rhythm with the peak in average firing frequency during the middle of the projected day. Vasoactive intestinal polypeptide (VIP), which is abundant in the rat SCN, phase-advances the time of peak in SCN neuronal activity when applied in vitro during the late projected night; an effect most likely mediated via the VPAC(2) receptor. Employing extracellular single-unit electrophysiology, we investigated the influence of protein kinase A (PKA) inhibitor KT5720 or mitogen-activated protein kinase kinase (MAPKK) inhibitor U0126 on phase advances evoked during the late projected night by VIP or Ro 25-1553, a VPAC(2) receptor agonist. Both inhibitors blocked VIP or Ro 25-1553-induced phase advances, indicating that the late-night resetting actions of VIP via the VPAC(2) receptor require PKA and MEK.
Collapse
Affiliation(s)
- Alert Meyer-Spasche
- School of Biological Sciences, University of Manchester, 3.614 Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | | |
Collapse
|
223
|
Erren TC, Reiter RJ, Pinger A, Piekarski C, Erren M. The chronosense – what light tells man about biological time. Med Hypotheses 2004; 63:1074-80. [PMID: 15504578 DOI: 10.1016/j.mehy.2004.04.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2004] [Accepted: 04/06/2004] [Indexed: 11/19/2022]
Abstract
In the past 10 years, experimental studies have provided further evidence for the suggestion that the eye serves man as a dual sense organ, viz as a sense organ for sight but also for time and the regulation of biological rhythms. A small group of scientists interested in the adjustment of biological rhythms to the key Zeitgeber light wanted to answer the question whether rods and/or cones and/or other uncharacterized retinal photoreceptors contribute to this function in mammals. Intriguingly, in the course of elegant research, a number of laboratories around the world have been zeroing in on a novel non-rod, non-cone ocular photopigment which serves a number of responses to non-image-forming (NIF) photoreception in mammals. This paper intends to draw attention to possible implications of photoreception and phototransduction research for other scientific disciplines which study health and diesase effects in man. We therefore review the pivotal role of the photoreceptors -- old and new -- for the light-related timing and coordination of the interplay of otherwise less efficient biological rhythms. To distinguish our focus on time- and timing-related effects from classic image-forming (IF) and other NIF responses to ambient light, we refer informatively to chronoreceptors which mediate the sense of time, or chronosense. We conclude that syndisciplinary research into the physiology and pathophysiological implications of the chronosense is warranted and summarize a series of research questions.
Collapse
Affiliation(s)
- Thomas C Erren
- Institute and Polyclinic for Occupational and Social Medicine, School of Medicine and Dentistry, University of Cologne, Joseph-Stelzmann-Str. 9, 50924 Köln, Lindenthal, Germany.
| | | | | | | | | |
Collapse
|
224
|
Madeira MD, Pereira PA, Silva SM, Cadete-Leite A, Paula-Barbosa MM. Basal forebrain neurons modulate the synthesis and expression of neuropeptides in the rat suprachiasmatic nucleus. Neuroscience 2004; 125:889-901. [PMID: 15120850 DOI: 10.1016/j.neuroscience.2004.03.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2004] [Indexed: 11/22/2022]
Abstract
We tested the hypothesis that efferents from the nucleus basalis magnocellularis (NBM) play a direct role in the regulation of neuropeptide synthesis and expression by neurons of the rat suprachiasmatic nucleus (SCN). Adult male rats in which the NBM was destroyed with quinolinic acid, either unilaterally or bilaterally, were compared with rats injected with physiological saline and with control rats. The estimators used to assess the effects of cholinergic deafferentation on the neuroanatomy and neurochemistry of the SCN were the total number of SCN neurons, the total number and somatic size of SCN neurons producing vasopressin (VP) and vasoactive intestinal polypeptide (VIP), and the respective mRNA levels. Bilateral destruction of the NBM did not produce cell death in the SCN, but caused a marked reduction in the number and somatic size of SCN neurons expressing VP and VIP, and in the mRNA levels of these peptides. The decrease in the number of VP- and VIP-producing neurons provoked by unilateral lesions was less striking than that resulting from bilateral lesions. It was, however, statistically significant in the ipsilateral hemisphere, but not in the contralateral hemisphere. The results show that the reduction of cholinergic inputs to the SCN impairs the synthesis, and thereby decreases the expression of neuropeptides by SCN neurons, and that the extent of the decline correlates with the amount of cholinergic afferents destroyed. This supports the notion that acetylcholine plays an important, and direct role in the regulation of the metabolic activity of SCN neurons.
Collapse
Affiliation(s)
- M D Madeira
- Department of Anatomy, Porto Medical School, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| | | | | | | | | |
Collapse
|
225
|
Schaap J, Albus H, VanderLeest HT, Eilers PHC, Détári L, Meijer JH. Heterogeneity of rhythmic suprachiasmatic nucleus neurons: Implications for circadian waveform and photoperiodic encoding. Proc Natl Acad Sci U S A 2003; 100:15994-9. [PMID: 14671328 PMCID: PMC307681 DOI: 10.1073/pnas.2436298100] [Citation(s) in RCA: 170] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2003] [Indexed: 11/18/2022] Open
Abstract
Circadian rhythms in neuronal ensemble, subpopulations, and single unit activity were recorded in the suprachiasmatic nuclei (SCN) of rat hypothalamic slices. Decomposition of the ensemble pattern revealed that neuronal subpopulations and single units within the SCN show surprisingly short periods of enhanced electrical activity of approximately 5 h and show maximal activity at different phases of the circadian cycle. The summed activity accounts for the neuronal ensemble pattern of the SCN, indicating that circadian waveform of electrical activity is a composed tissue property. The recorded single unit activity pattern was used to simulate the responsiveness of SCN neurons to different photoperiods. We inferred predictions on changes in peak width, amplitude, and peak time in the multiunit activity pattern and confirmed these predictions with hypothalamic slices from animals that had been kept in a short or long photoperiod. We propose that the animals' ability to code for day length derives from plasticity in the neuronal network of oscillating SCN neurons.
Collapse
Affiliation(s)
- Jeroen Schaap
- Departments of Neurophysiology and Medical Statistics, Leiden University Medical Center, Wassenaarseweg 62, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
226
|
Novak CM, Albers HE. Novel phase-shifting effects of GABAA receptor activation in the suprachiasmatic nucleus of a diurnal rodent. Am J Physiol Regul Integr Comp Physiol 2003; 286:R820-5. [PMID: 14656771 DOI: 10.1152/ajpregu.00575.2003] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The vast majority of neurons in the suprachiasmatic nucleus (SCN), the primary circadian pacemaker in mammals, contain the inhibitory neurotransmitter GABA. Most studies investigating the role of GABA in the SCN have been performed using nocturnal rodents. Activation of GABA(A) receptors by microinjection of muscimol into the SCN phase advances the circadian activity rhythm of nocturnal rodents, but only during the subjective day. Nonphotic stimuli that reset the circadian pacemaker of nocturnal rodents also produce phase advances during the subjective day. The role of GABA in the SCN of diurnal animals and how it may differ from nocturnal animals is not known. In the studies described here, the GABA(A) agonist muscimol was microinjected directly into the SCN region of diurnal unstriped Nile grass rats (Arvicanthis niloticus) at various times in their circadian cycle. The results demonstrate that GABA(A) receptor activation produces large phase delays during the subjective day in grass rats. Treatment with TTX did not affect the ability of muscimol to induce phase delays, suggesting that muscimol acts directly on pacemaker cells within the SCN. These data suggest that the circadian pacemakers of nocturnal and diurnal animals respond to the most abundant neurochemical signal found in SCN neurons in opposite ways. These findings are the first to demonstrate a fundamental difference in the functioning of circadian pacemaker cells in diurnal and nocturnal animals.
Collapse
Affiliation(s)
- C M Novak
- Center for Behavioral Neuroscience, Department of Biology, Georgia State University, PO Box 4010, Atlanta, GA 30302-4010, USA.
| | | |
Collapse
|
227
|
Bergström AL, Hannibal J, Hindersson P, Fahrenkrug J. Light-induced phase shift in the Syrian hamster (Mesocricetus auratus) is attenuated by the PACAP receptor antagonist PACAP6-38 or PACAP immunoneutralization. Eur J Neurosci 2003; 18:2552-62. [PMID: 14622156 DOI: 10.1046/j.1460-9568.2003.03000.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Circadian rhythms generated by the suprachiasmatic nucleus (SCN) are daily adjusted (entrained) by light via the retinohypothalamic tract (RHT). The RHT contains two neurotransmitters, glutamate and pituitary adenylate cyclase-activating polypeptide (PACAP), which are believed to mediate the phase-shifting effects of light on the clock. In the present study we have elucidated the role of PACAP in light-induced phase shifting at early night in hamsters and shown that (i) light-induced phase delay of running-wheel activity was significantly attenuated by a specific PAC1 receptor antagonist (PACAP6-38) or by immunoblockade with a specific anti-PACAP antibody injected intracerebroventricularly before light stimulation; (ii) PACAP administered close to the SCN was able to phase-delay the circadian rhythm of running-wheel activity in a similar way to light; (iii) PACAP was present in the hamster RHT, colocalized with melanopsin, a recently identified opsin which has been suggested to be a circadian photopigment. The findings indicate that PACAP is a neurotransmitter of the RHT mediating photic information to the clock, possibly via melanopsin located exclusively on the PACAP-expressing cells of the RHT.
Collapse
Affiliation(s)
- A L Bergström
- Department of Clinical Biochemistry, Bispebjerg Hospital, University of Copenhagen, Bispebjerg Bakke 23, DK-2400 Copenhagen NV, Denmark
| | | | | | | |
Collapse
|
228
|
Abstract
The suprachiasmatic nucleus (SCN) circadian clock exhibits a recurrent series of dynamic cellular states, characterized by the ability of exogenous signals to activate defined kinases that alter clock time. To explore potential relationships between kinase activation by exogenous signals and endogenous control mechanisms, we examined clock-controlled protein kinase G (PKG) regulation in the mammalian SCN. Signaling via the cGMP-PKG pathway is required for light- or glutamate (GLU)-induced phase advance in late night. Spontaneous cGMP-PKG activation occurred at the end of subjective night in free-running SCN in vitro. Phasing of the SCN rhythm in vitro was delayed by approximately 3 hr after treatment with guanylyl cyclase (GC) inhibitors, PKG inhibition, or antisense oligodeoxynucleotide (alphaODN) specific for PKG, but not PKA inhibitor or mismatched ODN. This sensitivity to GC-PKG inhibition was limited to the same 2 hr time window demarcated by clock-controlled activation of cGMP-PKG. Inhibition of the cGMP-PKG pathway at this time caused delays in the phasing of four endogenous rhythms: wheel-running activity, neuronal activity, cGMP, and Per1. Timing of the cGMP-PKG-necessary window in both rat and mouse depended on clock phase, established by the antecedent light/dark cycle rather than solar time. Because behavioral, neurophysiological, biochemical, and molecular rhythms showed the same temporal sensitivities and qualitative responses, we predict that clock-regulated GC-cGMP-PKG activation may provide a necessary cue as to clock state at the end of the nocturnal domain. Because sensitivity to phase advance by light-GLU-activated GC-cGMP-PKG occurs in juxtaposition, these signals may induce a premature shift to this PKG-necessary clock state.
Collapse
|
229
|
Abstract
The chick pineal organ is recognized to contain an endogenous circadian oscillator as well as having direct photic input pathways and the capability of synthesizing melatonin. Despite its interesting circadian cell biology, far less is known about the chick pineal as compared to mammalian pineal glands. The goals of our research were to identify and characterize novel components of the circadian system in this photoneuroendocrine organ. Using a subtractive screening strategy of a nocturnal chick pineal cDNA library, we identified numerous genes whose expression in the chick pineal has never been reported. Among these, we focused our attention on a homologue to the regulatory subunit of the mammalian serine/threonine protein phosphatase (STPP) 2A. The expression of this gene in the chick pineal is highly circadian both in vivo and in vitro. Analysis of the PP2A enzyme in this tissue revealed that it is predominantly cytosolic in localization, sensitive to classical PP2A inhibitors, and far more active during the subjective night. Interestingly, the acute pharmacological inhibition of PP2A leads to elevated phosphoCREB levels and concomitant melatonin secretion, indicating that this enzyme participates at some level in the control of nocturnal pineal melatonin synthesis. In a second aspect of our research, we examined the mechanisms underlying the circadian rhythmicity of cyclic GMP in the chick pineal. This signaling molecule is poorly understood, despite its well-known, high-amplitude circadian rhythms and the presence of many cGMP-dependent targets in this tissue. Our work has shown that although both soluble (sGC) and membrane-bound (mGC) forms of guanylyl cyclase are present, the primary contributor to the circadian rhythms of cGMP is the mGC-B enzyme, which is activated only by the natriuretic peptide CNP. As pharmacological blockade of mGC-B (but not sGC) suppresses nocturnal cGMP levels, we conclude that CNP-dependent mechanisms are involved. Hence, the circadian clock in the chick pineal appears to drive either CNP secretion or mGC-B expression (or synthetic efficiency) in order to elevate nocturnal cGMP. Conversely, light may inhibit cGMP by uncoupling this drive. These data provide new strategies for understanding both photic input pathways (presumed to depend on cGMP) and cGMP-dependent cellular function in the chick pineal organ.
Collapse
Affiliation(s)
- James Olcese
- Institute for Hormone and Fertility Research, University of Hamburg, Hamburg, Germany.
| |
Collapse
|
230
|
Meijer JH, Schwartz WJ. In search of the pathways for light-induced pacemaker resetting in the suprachiasmatic nucleus. J Biol Rhythms 2003; 18:235-49. [PMID: 12828281 DOI: 10.1177/0748730403018003006] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Within the suprachiasmatic nucleus (SCN) of the mammalian hypothalamus is a circadian pacemaker that functions as a clock. Its endogenous period is adjusted to the external 24-h light-dark cycle, primarily by light-induced phase shifts that reset the pacemaker's oscillation. Evidence using a wide variety of neurobiological and molecular genetic tools has elucidated key elements that comprise the visual input pathway for SCN photoentrainment in rodents. Important questions remain regarding the intracellular signals that reset the autoregulatory molecular loop within photoresponsive cells in the SCN's retino-recipient subdivision, as well as the intercellular coupling mechanisms that enable SCN tissue to generate phase shifts of overt behavioral and physiological circadian rhythms such as locomotion and SCN neuronal firing rate. Multiple neurotransmitters, protein kinases, and photoinducible genes add to system complexity, and we still do not fully understand how dawn and dusk light pulses ultimately produce bidirectional, advancing and delaying phase shifts for pacemaker entrainment.
Collapse
Affiliation(s)
- Johanna H Meijer
- Department of Physiology, Leiden University Medical Centre, 2300 RC Leiden, the Netherlands
| | | |
Collapse
|
231
|
Circadian and photic regulation of phosphorylation of ERK1/2 and Elk-1 in the suprachiasmatic nuclei of the Syrian hamster. J Neurosci 2003. [PMID: 12684495 DOI: 10.1523/jneurosci.23-07-03085.2003] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In this study we investigated the circadian and photic regulation of phosphorylation of the extracellular signal-related kinase (ERK) 1/2, and the transcription factor Elk-1 in the suprachiasmatic nuclei of the Syrian hamster. We report that levels of phosphorylated ERK (P-ERK) are rhythmic, peaking during the mid subjective day, whereas phosphorylated Elk-1 (P-Elk-1) shows no distinct rhythm. Light pulses during the subjective night rapidly, but transiently, induce P-ERK, whereas P-Elk-1 is also induced, albeit with a slower time course. Application of the ERK pathway inhibitor U0126 attenuates photic induction of both P-ERK and P-Elk-1 and phase advances of wheel-running behavior. The NMDA receptor channel blocker, MK-801, also significantly attenuates photic induction of P-ERK and P-Elk-1. Taken together, these results indicate a role of the ERK cascade in the regulation of free-running circadian rhythms and of photic-resetting of these rhythms and suggest that in the mammalian suprachiasmatic nuclei, Elk-1 represents a novel molecular component of the photic-induction pathway.
Collapse
|
232
|
Akiyama M, Minami Y, Kuriyama K, Shibata S. MAP kinase-dependent induction of clock gene expression by alpha 1-adrenergic receptor activation. FEBS Lett 2003; 542:109-14. [PMID: 12729908 DOI: 10.1016/s0014-5793(03)00360-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
While peripheral oscillators can be reset by humoral factors such as glucocorticoid hormones, indirect neural communications involving sympathetic and parasympathetic neurons from the suprachiasmatic nucleus to various peripheral tissues suggest that autonomic nerve innervations also function in the resetting and synchronization of peripheral tissues. To study the role of sympathetic adrenergic signaling on clock gene expression, we constructed NIH3T3 cells that stably expressed each of three alpha(1)-adrenergic receptor subtypes (alpha(1A), alpha(1B) and alpha(1D)). We found that noradrenaline transiently induced the expression of mPer1, mPer2, and mE4bp4 1-2 h after alpha(1)-receptor activation. The extent and time course of clock gene mRNA induction by noradrenaline or the alpha(1)-receptor agonist phenylephrine (PE) was similar to that seen by 50% horse serum shock. Clock gene mRNA induction by PE was inhibited by U0126, a MEK inhibitor, suggesting involvement of the mitogen-activated protein kinase signaling pathway. We also found that both mPer1 and mPer2 mRNAs were induced in the mouse liver 60 min after PE injection. These results suggest that although humoral factors are important for entrainment of the peripheral clock, the autonomic nervous system may also be involved in the process.
Collapse
Affiliation(s)
- Masashi Akiyama
- Department of Pharmacology and Brain Science, School of Human Sciences, Waseda University, Tokorozawa, Saitama 359-1192, Japan
| | | | | | | |
Collapse
|
233
|
Nishimura M, Yamagata K, Sugiura H, Okamura H. The activity-regulated cytoskeleton-associated (Arc) gene is a new light-inducible early gene in the mouse suprachiasmatic nucleus. Neuroscience 2003; 116:1141-7. [PMID: 12617955 DOI: 10.1016/s0306-4522(02)00786-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The mammalian circadian system is entrained to the environmental light/dark cycle by shifting the phase of the master clock in the suprachiasmatic nucleus. Accompanying the light-induced phase-shift, a variety of immediate-early genes appears in suprachiasmatic nucleus clock cells, and here, we report the expression of a new immediate-early gene Arc (activity-regulated cytoskeleton-associated gene) in mice. Arc messenger RNAs were strongly induced at 30-120 min after the light exposure at subjective night (CT12-CT20) in neurons of the retinorecipient area of the suprachiasmatic nucleus, although their spontaneous expression was absent in usual light-dark cycles and in constant dark conditions. At protein level, ARC appeared not only in the nucleus but also in the perikarya and their processes of the suprachiasmatic nucleus neurons. These findings indicate that Arc is an activity-regulated cytoskeletal gene possibly involved in the light-induced phase-shift of the circadian rhythm.
Collapse
Affiliation(s)
- M Nishimura
- Department of Brain Sciences, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | | | | | | |
Collapse
|
234
|
Sharma VK. Period responses to Zeitgeber signals stabilize circadian clocks during entrainment. Chronobiol Int 2003; 20:389-404. [PMID: 12868536 DOI: 10.1081/cbi-120020422] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Circadian clocks with characteristic period (tau) can be entrained to light/dark (LD) cycles by means of(i) phase shifts which are due to D/L "dawn" and/or L/D "dusk" transitions, (ii) period changes associated with long-term light exposure, or (iii) by combinations of the above possibilities. Based on stability analysis of a model circadian clock it was predicted that nocturnal burrowing mammals would benefit less from period responses than their diurnal counterparts. The model further predicted that maximal stability of circadian clock is reached when the clock slightly changes both its phase and period in response to light stimuli. Analyses of empirical phase response curve (PRC) and period response curve (tauRC) of some diurnal and nocturnal mammals revealed that PRCs of both diurnal and nocturnal mammals have similar waveform while tauRCs of nocturnal mammals are of smaller amplitude than those of diurnal mammals. The shape of the tauRC also changes with age and with increasing strength of light stimuli. During erratic fluctuations in light intensity under different weather conditions, the stability of phase of entrainment of circadian clocks appears to be achieved by an interplay between phase and period responses and the strength of light stimuli.
Collapse
Affiliation(s)
- Vijay Kumar Sharma
- Chronobiology Laboratory, Evolutionary and Organismal Biology Unit, Jawaharlal Nehru Center for Advanced Scientific Research, Jakkur, Bangalore, Karnataka, India.
| |
Collapse
|
235
|
Scheer FA, Kalsbeek A, Buijs RM. Cardiovascular control by the suprachiasmatic nucleus: neural and neuroendocrine mechanisms in human and rat. Biol Chem 2003; 384:697-709. [PMID: 12817466 DOI: 10.1515/bc.2003.078] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The risk for cardiovascular incidents is highest in the early morning, which seems partially due to endogenous factors. Endogenous circadian rhythms in mammalian physiology and behavior are regulated by the suprachiasmatic nucleus (SCN). Recently, anatomical evidence has been provided that SCN functioning is disturbed in patients with essential hypertension. Here we review neural and neuroendocrine mechanisms by which the SCN regulates the cardiovascular system. First, we discuss evidence for an endogenous circadian rhythm in cardiac activity, both in humans and rats, which is abolished after SCN lesioning in rats. The immediate impact of retinal light exposure at night on SCN-output to the cardiovascular system, which signals 'day' in both diurnal (human) and nocturnal (rat) mammals with opposite effects on physiology, is discussed. Furthermore, we discuss the impact of melatonin treatment on the SCN and its potential medical relevance in patients with essential hypertension. Finally, we argue that regional differentiation of the SCN and autonomous nervous system is required to explain the multitude of circadian rhythms. Insights into the mechanisms by which the SCN affects the cardiovascular system may provide new strategies for the treatment of disease conditions known to coincide with circadian rhythm disturbances, as is presented for essential hypertension.
Collapse
Affiliation(s)
- Frank A Scheer
- Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Brain Research, NL-1105 Amsterdam AZ, The Netherlands
| | | | | |
Collapse
|
236
|
Oster H, Werner C, Magnone MC, Mayser H, Feil R, Seeliger MW, Hofmann F, Albrecht U. cGMP-dependent protein kinase II modulates mPer1 and mPer2 gene induction and influences phase shifts of the circadian clock. Curr Biol 2003; 13:725-33. [PMID: 12725729 DOI: 10.1016/s0960-9822(03)00252-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND In mammals, the master circadian clock that drives many biochemical, physiological, and behavioral rhythms is located in the suprachiasmatic nuclei (SCN) of the hypothalamus. Generation and maintenance of circadian rhythmicity rely on complex interlocked transcriptional/translational feedback loops involving a set of clock genes. Among the molecular components driving the mammalian circadian clock are the Period 1 and 2 (mPer1 and mPer2) genes. Because the periodicity of the clock is not exactly 24 hr, it has to be adjusted periodically. The major stimulus for adjustment (resetting) of the clock is nocturnal light. It evokes activation of signaling pathways in the SCN that ultimately lead to expression of mPer1 and mPer2 genes conveying adjustment of the clock. RESULTS We show that mice deficient in cGMP-dependent protein kinase II (cGKII, also known as PKGII), despite regular retinal function, are defective in resetting the circadian clock, as assessed by changes in the onset of wheel running activity after a light pulse. At the molecular level, light induction of mPer2 in the SCN is strongly reduced in the early period of the night, whereas mPer1 induction is elevated in cGKII-deficient mice. Additionally, we show that light induction of cfos and light-dependent phosphorylation of CREB at serine 133 are not affected in these animals. CONCLUSIONS cGKII plays a role in the clock-resetting mechanism. In particular, the ability to delay clock phase is affected in cGKII-deficient mice. It seems that the signaling pathway involving cGKII influences in an opposite manner the light-induced induction of mPer1 and mPer2 genes and thereby influences the direction of a phase shift of the circadian clock.
Collapse
Affiliation(s)
- Henrik Oster
- Department of Medicine, Division of Biochemistry, University of Fribourg, 1700 Fribourg, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
237
|
Ikeda M, Sugiyama T, Wallace CS, Gompf HS, Yoshioka T, Miyawaki A, Allen CN. Circadian Dynamics of Cytosolic and Nuclear Ca2+ in Single Suprachiasmatic Nucleus Neurons. Neuron 2003; 38:253-63. [PMID: 12718859 DOI: 10.1016/s0896-6273(03)00164-8] [Citation(s) in RCA: 223] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Intracellular free Ca(2+) regulates diverse cellular processes, including membrane potential, neurotransmitter release, and gene expression. To examine the cellular mechanisms underlying the generation of circadian rhythms, nucleus-targeted and untargeted cDNAs encoding a Ca(2+)-sensitive fluorescent protein (cameleon) were transfected into organotypic cultures of mouse suprachiasmatic nucleus (SCN), the primary circadian pacemaker. Circadian rhythms in cytosolic but not nuclear Ca(2+) concentration were observed in SCN neurons. The cytosolic Ca(2+) rhythm period matched the circadian multiple-unit-activity (MUA)-rhythm period monitored using a multiple-electrode array, with a mean advance in phase of 4 hr. Tetrodotoxin blocked MUA, but not Ca(2+) rhythms, while ryanodine damped both Ca(2+) and MUA rhythms. These results demonstrate cytosolic Ca(2+) rhythms regulated by the release of Ca(2+) from ryanodine-sensitive stores in SCN neurons.
Collapse
Affiliation(s)
- Masayuki Ikeda
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, 6-2-4 Furuedai, Suita, 565-0874, Osaka, Japan.
| | | | | | | | | | | | | |
Collapse
|
238
|
Sanggaard KM, Hannibal J, Fahrenkrug J. Serotonin inhibits glutamate- but not PACAP-induced per gene expression in the rat suprachiasmatic nucleus at night. Eur J Neurosci 2003; 17:1245-52. [PMID: 12670312 DOI: 10.1046/j.1460-9568.2003.02562.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Circadian rhythms of physiology and behaviour generated by the brain's biological clock located in the suprachiasmatic nucleus are entrained by light via the retinohypothalamic tract. Two neurotransmitters, glutamate and pituitary adenylate cyclase-activating polypeptide (PACAP), found in this monosynaptic pathway mediate the effects of light to the clock. It is well known that not only light entrains the clock. Nonphotic cues mediated by neurotransmitters such as serotonin reaching the suprachiasmatic nucleus from the midbrain raphe nucleus modulate light-induced phase shifts at night. Two clock genes, per1 and per2, have been attributed a role in light-induced phase shift. In the present study, using an in vitro brain slice model and quantitative in situ hybridization for per1 and per2, we have shown that serotonin induces per1 gene expression at late subjective night but not at early night. Furthermore, serotonin application before glutamate or PACAP blocked glutamate-induced per1 expression at early night and per2 gene expression at late night. In contrast, serotonin did not influence PACAP-induced per gene expression at late night. Triple antigen immunohistochemistry and confocal microscopy supported both a pre- and post-synaptic interaction of retinohypothalamic tract (PACAP-immunoreactive) and serotonin projections on vasoactive intestinal peptide- and gastrin-releasing peptide-containing cell bodies in the ventro-lateral suprachiasmatic nucleus. Our findings suggest that the per genes could be the molecular target for the modulatory effects of serotonin on light signalling to the clock.
Collapse
Affiliation(s)
- K M Sanggaard
- Department of Clinical Biochemistry, Bispebjerg Hospital, University of Copenhagen, DK-2400 Copenhagen NV, Denmark
| | | | | |
Collapse
|
239
|
Abstract
The neurons of the mammalian suprachiasmatic nuclei (SCN) control circadian rhythms in molecular, physiological, endocrine, and behavioral functions. In the SCN, circadian rhythms are generated at the level of individual neurons. The last decade has provided a wealth of information on the genetic basis for circadian rhythm generation. In comparison, a modest but growing number of studies have investigated how the molecular rhythm is translated into neuronal function. Neuronal attributes have been measured at the cellular and tissue level with a variety of electrophysiological techniques. We have summarized electrophysiological research on neurons that constitute the SCN in an attempt to provide a comprehensive view on the current state of the art.
Collapse
Affiliation(s)
- Jeroen Schaap
- Department of Neurophysiology, Leiden University Medical School, RC Leiden, The Netherlands
| | | | | |
Collapse
|
240
|
Takahashi H, Umeda N, Tsutsumi Y, Fukumura R, Ohkaze H, Sujino M, van der Horst G, Yasui A, Inouye SIT, Fujimori A, Ohhata T, Araki R, Abe M. Mouse dexamethasone-induced RAS protein 1 gene is expressed in a circadian rhythmic manner in the suprachiasmatic nucleus. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 110:1-6. [PMID: 12573527 DOI: 10.1016/s0169-328x(02)00543-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We identified the Dexamethasone-induced RAS protein 1 (Dexras1) gene as a cycling gene in the suprachiasmatic nucleus (SCN). Investigation of the whole brain using in situ hybridization demonstrated the localization of the expression of the gene in the SCN, thalamus, piriform cortex and hippocampus. However, rhythmic expression of the gene was observed only in the SCN. The rhythmic change in gene expression during 1 day was approximately five-fold, and the maximum expression was observed during subjective night. Real-time PCR using the SCN, paraventricular nucleus and cortex confirmed these results. Next, we analyzed the expression of the Dexras1 gene in the SCN of cryptochrome (Cry) 1 and 2 double knockout mice. We found that the rhythmic expression disappeared. The results indicate that Dexras1 rhythmicity and levels are dependent upon CRYs. This is the first time that the G protein, which may be involved in the input pathway, has been isolated as a cycling gene in the SCN.
Collapse
Affiliation(s)
- Hirokazu Takahashi
- Transcriptome Profiling Group, National Institute of Radiological Sciences, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Intrinsic role of polysialylated neural cell adhesion molecule in photic phase resetting of the Mammalian circadian clock. J Neurosci 2003. [PMID: 12533624 DOI: 10.1523/jneurosci.23-02-00652.2003] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The suprachiasmatic nuclei (SCN), the location of the mammalian circadian clock, are one of the few adult brain regions that express the highly polysialylated form of neural cell adhesion molecule (PSA-NCAM). A role for the polysialic acid (PSA) component of PSA-NCAM, which is known to promote tissue plasticity, has been reported for photic entrainment of circadian rhythmicity in vivo. The in vivo results, however, do not discriminate between PSA acting upstream or downstream of the glutamatergic synapses that convey photic information to the SCN. To address this key issue, we exploited an in vitro rat brain slice preparation that retains robust circadian function. As in the intact SCN, PSA levels in the isolated SCN are rhythmic, with higher levels during the early subjective day and lower levels during subjective night. Importantly, bath application of glutamate to SCN slices rapidly and transiently increases PSA levels during both the subjective day and night. Pretreating the slices with endoneuraminidase, which selectively removes PSA from NCAM and thereby prevents this increase, abolishes glutamate- and optic chiasm stimulation-induced phase delays of the SCN circadian neuronal activity rhythm. These results support the hypothesis that PSA expression in the SCN is controlled by both the circadian clock and photic input to the clock and that expression of PSA in the SCN is critical for photic-like phase shifts of the clock. Together, these results establish that such actions of PSA are manifested downstream from presynaptic retinohypothalamic terminals and therefore are intrinsic to the SCN itself.
Collapse
|
242
|
Tischkau SA, Mitchell JW, Tyan SH, Buchanan GF, Gillette MU. Ca2+/cAMP response element-binding protein (CREB)-dependent activation of Per1 is required for light-induced signaling in the suprachiasmatic nucleus circadian clock. J Biol Chem 2003; 278:718-23. [PMID: 12409294 DOI: 10.1074/jbc.m209241200] [Citation(s) in RCA: 223] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Light is a prominent stimulus that synchronizes endogenous circadian rhythmicity to environmental light/dark cycles. Nocturnal light elevates mRNA of the Period1 (Per1) gene and induces long term state changes, expressed as phase shifts of circadian rhythms. The cellular mechanism for Per1 elevation and light-induced phase advance in the suprachiasmatic nucleus (SCN), a process initiated primarily by glutamatergic neurotransmission from the retinohypothalamic tract, was examined. Glutamate (GLU)-induced phase advances in the rat SCN were blocked by antisense oligodeoxynucleotide (ODN) against Per1 and Ca(2+)/cAMP response element (CRE)-decoy ODN. CRE-decoy ODN also blocked light-induced phase advances in vivo. Furthermore, the CRE-decoy blocked GLU-induced accumulation of Per1 mRNA. Thus, Ca(2+)/cAMP response element-binding protein (CREB) and Per1 are integral components of the pathway transducing light-stimulated GLU neurotransmission into phase advance of the circadian clock.
Collapse
Affiliation(s)
- Shelley A Tischkau
- Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, B107 CLSL, 61801, USA
| | | | | | | | | |
Collapse
|
243
|
Ikeda M, Yoshioka T, Allen CN. Developmental and circadian changes in Ca2+ mobilization mediated by GABAA and NMDA receptors in the suprachiasmatic nucleus. Eur J Neurosci 2003; 17:58-70. [PMID: 12534969 DOI: 10.1046/j.1460-9568.2003.02427.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The hypothalamic suprachiasmatic nucleus (SCN) develops as the circadian pacemaker during postnatal life. Although both GABAA and NMDA receptors are expressed in the majority of SCN neurons, postnatal development of their functions has not been analysed. Thus, we studied the receptor-mediated Ca2+ responses in mouse hypothalamic slices prepared on postnatal days (P) 6-16. The NMDA-induced Ca2+ flux was prominent in the SCN and maximal Ca2+ responses in Mg2+-free conditions had no day-night variations in P14-16 mice. At P6-7, extracellular Mg2+ reduced the NMDA-induced Ca2+ flux irrespective of the circadian time whereas, after P9-10, Mg2+ produced a larger reduction at night than during the daytime. Muscimol also significantly increased Ca2+ in the developing SCN. Voltage-sensitive Ca2+ channel blockers inhibited the muscimol-induced Ca2+ increase whereas tetrodotoxin had no effect, suggesting that stimulation of postsynaptic GABAA receptors depolarizes SCN neurons to increase Ca2+. Macroscopic imaging analysis demonstrated a developmental reduction in the muscimol-induced Ca2+ increase preferentially in the nighttime group older than P9-10. The day-night variation in the magnitude of the Ca2+ response was due to two cell populations, one of which exhibited an increase and the other a decrease in Ca2+ in response to muscimol. Because the critical developmental stages for exhibiting day-night variations in the receptor-mediated Ca2+ responses overlapped the maturation of firing rhythms in SCN neurons, the Ca2+ signalling may be necessary for or regulated by the mature circadian clock.
Collapse
Affiliation(s)
- Masayuki Ikeda
- Department of Molecular Behavioural Biology, Osaka Bioscience Institute, 6-2-4 Furuedai, Suita, Osaka 565-0874, Japan.
| | | | | |
Collapse
|
244
|
Dardente H, Poirel VJ, Klosen P, Pévet P, Masson-Pévet M. Per and neuropeptide expression in the rat suprachiasmatic nuclei: compartmentalization and differential cellular induction by light. Brain Res 2002; 958:261-71. [PMID: 12470861 DOI: 10.1016/s0006-8993(02)03563-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Per1 and Per2, two clock genes rhythmically expressed in the suprachiasmatic nucleus (SCN), are implicated in the molecular mechanism of the circadian pacemaker and play a major role in its entrainment by light. To date, it is not known if every cell of the SCN, a heterogeneous structure in respect of neuropeptide content, expresses clock genes equally. The aim of this study was to identify, by single and double non-radioactive and/or radioactive hybridizations, the cell types (AVP, VIP and GRP) expressing Per1 or Per2 in the SCN of rats, (1) when Per are highly expressed during the daytime, and (2) after induction of Per expression by a light pulse at night. Our results indicate that, during the daytime, Per1 and Per2 genes are both mainly expressed in the AVP cells of the dorso-median part of the SCN, whereas only a few VIP cells in the ventral part of the SCN exhibit Per gene expression. In contrast, following a light pulse at night, there is differential induction of the two Per genes. Per1 expression essentially occurs in the ventro-lateral GRP cells, while Per2 expression is not restricted to the retinorecipient part of the SCN as it also occurs in AVP cells. Altogether, our results suggest that Per1 and Per2 are mainly expressed in AVP cells during the daytime and suggest that GRP cells play an important role in resetting of the clock by light.
Collapse
Affiliation(s)
- Hugues Dardente
- Neurobiologie des Rythmes, UMR 7518 CNRS/ULP, IFR 37, 12 Rue de l'Université, 67000 Strasbourg, France
| | | | | | | | | |
Collapse
|
245
|
Amir S, Beaulé C, Arvanitogiannis A, Stewart J. Modes of plasticity within the mammalian circadian system. PROGRESS IN BRAIN RESEARCH 2002; 138:191-203. [PMID: 12432771 DOI: 10.1016/s0079-6123(02)38079-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Affiliation(s)
- Shimon Amir
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, 1455 de Maisonneuve Boulevard, West, Montreal, QC, H3G 1M8, Canada.
| | | | | | | |
Collapse
|
246
|
Fukunaga K, Horikawa K, Shibata S, Takeuchi Y, Miyamoto E. Ca2+/calmodulin-dependent protein kinase II-dependent long-term potentiation in the rat suprachiasmatic nucleus and its inhibition by melatonin. J Neurosci Res 2002; 70:799-807. [PMID: 12444602 DOI: 10.1002/jnr.10400] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We recently reported that Ca(2+)/calmodulin-dependent protein (CaM) kinase II is involved in light-induced phase delays and Per gene induction in the suprachiasmatic nucleus (SCN). To clarify the activation mechanisms of CaM kinase II by glutamate receptor stimulation in the SCN, we documented CaM kinase II activation following induction of long-term potentiation (LTP) in the rat SCN. High-frequency stimulation (100 Hz, 1 sec) applied to the optic nerve resulted in LTP of a postsynaptic field potential in the rat SCN. Unlike LTP in the hippocampal CA1 region, LTP onset in the SCN was slow and partly dependent on N-methyl-D-aspartate receptor activation. LTP induction in the SCN was completely inhibited by treatment with a nitric oxide synthase inhibitor or with a specific CaM kinase II inhibitor. Immunoblotting analysis using phosphospecific antibodies against autophosphorylated CaM kinase II revealed that LTP induction was accompanied by an increase in autophosphorylation. After high-frequency stimulation, we could visualize activation of CaM kinase II in vasoactive intestinal polypeptide-positive neurons in the SCN by immunohistochemistry. Treatment with cyclosporin A, a calcineurin inhibitor, potentiated LTP induction in the rat SCN. Interestingly, treatment with melatonin totally prevented LTP induction, without changes in basal synaptic transmission. Analyses of phosphorylation of CaM kinase II, mitogen-activated protein kinase, and cAMP-responsive element binding protein revealed that stimulatory and inhibitory effects on CaM kinase II autophosphorylation underlie the effects of cyclosporin A and melatonin, respectively. These results suggest that CaM kinase II plays critical roles in LTP induction in the SCN and that melatonin has inhibitory effects on synaptic plasticity through CaM kinase II.
Collapse
Affiliation(s)
- Kohji Fukunaga
- Department of Pharmacology, Kumamoto University School of Medicine, Kumamoto, Japan.
| | | | | | | | | |
Collapse
|
247
|
Scholz NL, Labenia JS, de Vente J, Graubard K, Goy MF. Expression of nitric oxide synthase and nitric oxide-sensitive guanylate cyclase in the crustacean cardiac ganglion. J Comp Neurol 2002; 454:158-67. [PMID: 12412140 DOI: 10.1002/cne.10442] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The cardiac ganglion is a simple central pattern-generating network that controls the rhythmic contractions of the crustacean heart. Enzyme assays and Western blots show that whole heart homogenates from the crab Cancer productus contain high levels of nitric oxide synthase (NOS), an enzyme that catalyzes the conversion of arginine to citrulline with concomitant production of the transmitter nitric oxide (NO). Crab heart NOS is calcium-dependent and has an apparent molecular weight of 110 kDa. In the cardiac ganglion, antibodies to NOS and citrulline indicate the presence of a NOS-like protein and NOS enzymatic activity in the four small pacemaker neurons and the five large motor neurons of the cardiac network. In addition, all cardiac neurons label positively with an antibody to cyclic guanosine monophosphate (cGMP). The NO donor sodium nitroprusside (SNP, 10 mM) stimulates additional cGMP production in the isolated ganglion. This increase is blocked by [(1)H](1,2,4)oxadiazole(4,3-a)quinoxalin-1-one (ODQ, 50 microM), an inhibitor of the NO-sensitive soluble guanylate cyclase (sGC). Taken together, our data indicate that NO- and cGMP-mediated signaling pathways are enriched in the cardiac system relative to other crab tissues and that the cardiac network may be a target for extrinsic and intrinsic neuromodulation via NO produced from the heart musculature and individual cardiac neurons, respectively. The crustacean cardiac ganglion is therefore a promising system for studying cellular and synaptic mechanisms of nitrergic neuromodulation in a simple pattern-generating network.
Collapse
Affiliation(s)
- Nathaniel L Scholz
- Department of Zoology, University of Washington, Seattle, Washington 98112, USA
| | | | | | | | | |
Collapse
|
248
|
Corbalán R, Chatauret N, Behrends S, Butterworth RF, Felipo V. Region selective alterations of soluble guanylate cyclase content and modulation in brain of cirrhotic patients. Hepatology 2002; 36:1155-62. [PMID: 12395325 DOI: 10.1053/jhep.2002.36365] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Modulation of soluble guanylate cyclase (sGC) by nitric oxide (NO) is altered in brain from experimental animals with hyperammonemia with or without liver failure. The aim of this work was to assess the content and modulation of sGC in brain in chronic liver failure in humans. Expression of the alpha-1, alpha-2, and beta-1 subunits of sGC was measured by immunoblotting in autopsied frontal cortex and cerebellum from cirrhotic patients and controls. The contents of alpha-1 and alpha-2 subunits of guanylate cyclase was increased both in cortex and cerebellum, whereas the beta-1 subunit was not affected. Addition of the NO-generating agent S-nitroso-N-acetyl-penicillamine (SNAP) to homogenates of frontal cortex from controls increased the activity of sGC 87-fold, whereas, in homogenates from cirrhotic patients, the increase was significantly higher (183-fold). In contrast, in cerebellum, activation of guanylate cyclase by NO was significantly lower in patients (156-fold) than in controls (248-fold). A similar regional difference was found in rats with portacaval anastomosis. In conclusion, these findings show that the NO-guanylate cyclase signal transduction pathway is strongly altered in brain in patients with chronic liver failure and that the effects are different in different brain areas. Given that activation of sGC by NO in brain is involved in the modulation of important cerebral processes such as intercellular communication, learning and memory, and the sleep-wake cycle, these changes could be implicated in the pathogenesis of hepatic encephalopathy in these patients.
Collapse
Affiliation(s)
- Regina Corbalán
- Laboratory of Neurobiology, Instituto de Investigaciones Citológicas, Fundación Valenciana de Investigaciones Biomédicas, Spain
| | | | | | | | | |
Collapse
|
249
|
Hurst WJ, Mitchell JW, Gillette MU. Synchronization and phase-resetting by glutamate of an immortalized SCN cell line. Biochem Biophys Res Commun 2002; 298:133-43. [PMID: 12379231 DOI: 10.1016/s0006-291x(02)02346-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
SCN 2.2 cultures were stably transfected with luciferase reporter constructs driven by Ca(2+)/cAMP response element, E-box, or vasoactive intestinal peptide promoter to probe the circadian properties of this clock cell line. SCN 2.2 reporter lines displayed approximately 24-h rhythms of transcriptional activation after serum-shock. Serum-shocked cultures pulsed with glutamate exhibited phase-gated induction of phospho-CREB and of VIP, CRE, and E-box promoter activity. Glutamate-induced CRE promoter activity displayed restricted sensitivity to inhibitors of nitric oxide synthase and cGMP-dependent protein kinase. The temporal pattern of these sensitivities paralleled those of the SCN to light and glutamate during the night. Taken together, our data indicate that serum-shock can synchronize the circadian clock of SCN 2.2 cells to a state consistent with the day/night transition and, thus, establishes a temporal context for this cell line.
Collapse
Affiliation(s)
- William J Hurst
- Departments of Cell and Structural Biology, University of Illinois at Urbana-Champaign, 601 South Goodwin Avenue, Urbana, IL 61801, USA
| | | | | |
Collapse
|
250
|
Lundkvist GB, Hill RH, Kristensson K. Disruption of circadian rhythms in synaptic activity of the suprachiasmatic nuclei by African trypanosomes and cytokines. Neurobiol Dis 2002; 11:20-7. [PMID: 12460543 DOI: 10.1006/nbdi.2002.0536] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Disturbances in biological rhythms pose a major disease problem, not the least in the aging population. Experimental sleeping sickness, caused by Trypanosoma brucei brucei, in rats constitutes a unique and robust chronic model for studying mechanisms of such disturbances. The spontaneous postsynaptic activity was recorded in slice preparations of the suprachiasmatic nuclei (SCN), which contain the master pacemaker for circadian rhythms in mammals, from trypanosome-infected rats. The excitatory synaptic events, which in normal rats show a daily variation, were reduced in frequency, while the inhibitory synaptic events did not significantly differ. This indicates selective disturbances in glutamate receptor-mediated neurotransmission in the SCN. Treatment with interferon-gamma in combination with lipopolysaccharide, which has synergistic actions with cytokines, and tumor necrosis factor-alpha similarly caused a reduction in excitatory synaptic SCN activity. We suggest that changes in the synaptic machinery of SCN neurons play an important pathogenetic role in sleeping sickness, and that proinflammatory cytokines can mimic these changes.
Collapse
Affiliation(s)
- G B Lundkvist
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | | |
Collapse
|