Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
World Neurosurg ; 155: e460-e471, 2021 11.
Article in English | MEDLINE | ID: mdl-34454071

ABSTRACT

BACKGROUND: Endoscopic endonasal surgery has proved to offer a practical route to treat suprasellar lesions, including tumors and vascular pathologies. Understanding the different configurations of the anterior cerebral communicating artery (ACoA) complex (ACoA-C) is crucial to properly navigate the suprachiasmatic space and decrease any vascular injury while approaching this region through an endonasal approach. METHODS: An endoscopic endonasal transplanum-transtubercular approach was performed on 36 cadaveric heads (72 sides). The variations of the ACoA-C and feasibility of reaching its different components were analyzed. The surgical area exposure of the lamina terminalis was also quantified before and after mobilization of the ACoA-C. RESULTS: The typical ACoA-C configuration was found in 41.6% of specimens. The following 2 main variations were identified: accessory anterior cerebral artery segment 2 (5, 13.9%) and common trunk of anterior cerebral artery with absence of ACoA (5, 13.9%). Of 101 recurrent arteries of Heubner, 96 (95.0%) were identified within 4 mm proximal or distal to the ACoA. The mean lamina terminalis exposure area was 33.1 ± 16.7 mm2, which increased to 59.9 ± 11.9 mm2 after elevating the ACoA. CONCLUSIONS: A considerable amount of variation of the ACoA-C can be found through an endoscopic endonasal transplanum-transtubercular approach. These configurations determine the feasibility of lamina terminalis exposure and the complexity of reaching the ACoA. Assessment of ACoA morphology and its adjacent structures is crucial while approaching the suprachiasmatic through a transnasal corridor.


Subject(s)
Cerebral Arteries/surgery , Nasal Cavity/surgery , Neuroendoscopy/methods , Suprachiasmatic Nucleus/blood supply , Suprachiasmatic Nucleus/surgery , Cadaver , Cerebral Arteries/pathology , Humans , Nasal Cavity/pathology , Suprachiasmatic Nucleus/pathology
2.
Genes Dev ; 35(5-6): 329-334, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33602874

ABSTRACT

It has been assumed that the suprachiasmatic nucleus (SCN) synchronizes peripheral circadian oscillators. However, this has never been convincingly shown, since biochemical time series experiments are not feasible in behaviorally arrhythmic animals. By using long-term bioluminescence recording in freely moving mice, we show that the SCN is indeed required for maintaining synchrony between organs. Surprisingly, however, circadian oscillations persist in the livers of mice devoid of an SCN or oscillators in cells other than hepatocytes. Hence, similar to SCN neurons, hepatocytes can maintain phase coherence in the absence of Zeitgeber signals produced by other organs or environmental cycles.


Subject(s)
Circadian Clocks/physiology , Hepatocytes/physiology , Suprachiasmatic Nucleus/physiology , Animals , Circadian Clocks/genetics , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Suprachiasmatic Nucleus/surgery
3.
Curr Biol ; 29(15): 2477-2487.e6, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31353184

ABSTRACT

The cochlea possesses a robust circadian clock machinery that regulates auditory function. How the cochlear clock is influenced by the circadian system remains unknown. Here, we show that cochlear rhythms are system driven and require local Bmal1 as well as central input from the suprachiasmatic nuclei (SCN). SCN ablations disrupted the circadian expression of the core clock genes in the cochlea. Because the circadian secretion of glucocorticoids (GCs) is controlled by the SCN and GCs are known to modulate auditory function, we assessed their influence on circadian gene expression. Removal of circulating GCs by adrenalectomy (ADX) did not have a major impact on core clock gene expression in the cochlea. Rather it abolished the transcription of clock-controlled genes involved in inflammation. ADX abolished the known differential auditory sensitivity to day and night noise trauma and prevented the induction of GABA-ergic and glutamate receptors mRNA transcripts. However, these improvements were unrelated to changes at the synaptic level, suggesting other cochlear functions may be involved. Due to this circadian regulation of noise sensitivity by GCs, we evaluated the actions of the synthetic glucocorticoid dexamethasone (DEX) at different times of the day. DEX was effective in protecting from acute noise trauma only when administered during daytime, when circulating glucocorticoids are low, indicating that chronopharmacological approaches are important for obtaining optimal treatment strategies for hearing loss. GCs appear as a major regulator of the differential sensitivity to day or night noise trauma, a mechanism likely involving the circadian control of inflammatory responses.


Subject(s)
Circadian Clocks/physiology , Cochlea/physiology , Dexamethasone/administration & dosage , Glucocorticoids/administration & dosage , Noise , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Dexamethasone/metabolism , Glucocorticoids/metabolism , Male , Mice , Suprachiasmatic Nucleus/physiology , Suprachiasmatic Nucleus/surgery
4.
Vestn Oftalmol ; 132(3): 32-36, 2016.
Article in Russian | MEDLINE | ID: mdl-27456562

ABSTRACT

UNLABELLED: Studying compensatory capacity of the brain is a pressing issue. To resolve it, diversified experiments should be conducted providing an idea of the underlying mechanisms and possibilities of functional restoration. AIM: To determine the effect of unilateral electrocoagulation of supraoptic (SO) and suprachiasmatic (SCH) hypothalamic nuclei on the appearance of the electroretinogram (ERG). MATERIAL AND METHODS: We conducted chronic experiments on awake rabbits with a total duration of 30 days. RESULTS: The study revealed considerable changes in the total ERG amplitude (reduction) as well as the amplitudes of a- and b-waves. Thus, for the first 15-30 minutes after coagulation the b-wave showed an increase, while the a-wave appeared completely suppressed. Longer postcoagulation periods were associated with gradual, though incomplete, amplitude regain of the latter. CONCLUSION: The obtained data suggest that the process of ERG recovery involves compensatory mechanisms of the SO and SCH nuclei that enable partial recovery of the neurotransmitter activity of the retina.


Subject(s)
Neuronal Plasticity/physiology , Retina , Suprachiasmatic Nucleus , Supraoptic Nucleus , Animals , Electrocoagulation/methods , Electroretinography/methods , Models, Animal , Nerve Growth Factor/physiology , Rabbits , Retina/physiology , Retina/physiopathology , Suprachiasmatic Nucleus/physiology , Suprachiasmatic Nucleus/physiopathology , Suprachiasmatic Nucleus/surgery , Supraoptic Nucleus/physiology , Supraoptic Nucleus/physiopathology , Supraoptic Nucleus/surgery , Synaptic Transmission/physiology
5.
Genes Dev ; 27(13): 1526-36, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23824542

ABSTRACT

The mammalian circadian timing system consists of a master pacemaker in the suprachiasmatic nucleus (SCN) in the hypothalamus, which is thought to set the phase of slave oscillators in virtually all body cells. However, due to the lack of appropriate in vivo recording technologies, it has been difficult to study how the SCN synchronizes oscillators in peripheral tissues. Here we describe the real-time recording of bioluminescence emitted by hepatocytes expressing circadian luciferase reporter genes in freely moving mice. The technology employs a device dubbed RT-Biolumicorder, which consists of a cylindrical cage with reflecting conical walls that channel photons toward a photomultiplier tube. The monitoring of circadian liver gene expression revealed that hepatocyte oscillators of SCN-lesioned mice synchronized more rapidly to feeding cycles than hepatocyte clocks of intact mice. Hence, the SCN uses signaling pathways that counteract those of feeding rhythms when their phase is in conflict with its own phase.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm , Gene Expression Regulation , Hepatocytes/physiology , Liver/metabolism , Motor Activity/physiology , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Feeding Behavior , Liver/cytology , Luminescent Measurements , Male , Mice , Mice, Hairless , Motor Activity/genetics , Signal Transduction , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/surgery
6.
Am J Physiol Gastrointest Liver Physiol ; 303(4): G461-73, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22723262

ABSTRACT

Circadian clocks are responsible for daily rhythms in a wide array of processes, including gastrointestinal (GI) function. These are vital for normal digestive rhythms and overall health. Previous studies demonstrated circadian clocks within the cells of GI tissue. The present study examines the roles played by the suprachiasmatic nuclei (SCN), master circadian pacemaker for overt circadian rhythms, and the sympathetic nervous system in regulation of circadian GI rhythms in the mouse Mus musculus. Surgical ablation of the SCN abolishes circadian locomotor, feeding, and stool output rhythms when animals are presented with food ad libitum, while restricted feeding reestablishes these rhythms temporarily. In intact mice, chemical sympathectomy with 6-hydroxydopamine has no effect on feeding and locomotor rhythmicity in light-dark cycles or constant darkness but attenuates stool weight and stool number rhythms. Again, however, restricted feeding reestablishes rhythms in locomotor activity, feeding, and stool output rhythms. Ex vivo, intestinal tissue from PER2::LUC transgenic mice expresses circadian rhythms of luciferase bioluminescence. Chemical sympathectomy has little effect on these rhythms, but timed administration of the ß-adrenergic agonist isoproterenol causes a phase-dependent shift in PERIOD2 expression rhythms. Collectively, the data suggest that the SCN are required to maintain feeding, locomotor, and stool output rhythms during ad libitum conditions, acting at least in part through daily activation of sympathetic activity. Even so, this input is not necessary for entrainment to timed feeding, which may be the province of oscillators within the intestines themselves or other components of the GI system.


Subject(s)
Biological Clocks , Circadian Rhythm , Gastrointestinal Tract/innervation , Suprachiasmatic Nucleus/physiology , Sympathetic Nervous System/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Biological Clocks/drug effects , Biological Clocks/genetics , Blotting, Western , Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Defecation , Denervation/methods , Eating , Feeding Behavior , Immunohistochemistry , Isoproterenol/pharmacology , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity , Oxidopamine/pharmacology , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Photoperiod , Suprachiasmatic Nucleus/surgery , Sympathectomy, Chemical , Sympathetic Nervous System/drug effects , Sympatholytics/pharmacology , Time Factors
7.
Chronobiol Int ; 29(4): 454-68, 2012 May.
Article in English | MEDLINE | ID: mdl-22475541

ABSTRACT

A unique extra-suprachiasmatic nucleus (SCN) oscillator, operating independently of the light-entrainable oscillator, has been hypothesized to generate feeding and drug-related rhythms. To test the validity of this hypothesis, sham-lesioned (Sham) and SCN-lesioned (SCNx) rats were housed in constant dim-red illumination (LL(red)) and received a daily cocaine injection every 24 h for 7 d (Experiment 1). In a second experiment, rats underwent 3-h daily restricted feeding (RF) followed 12 d later by the addition of daily cocaine injections given every 25 h in combination with RF until the two schedules were in antiphase. In both experiments, body temperature and total activity were monitored continuously. Results from Experiment 1 revealed that cocaine, but not saline, injections produced anticipatory increases in temperature and activity in SCNx and Sham rats. Following withdrawal from cocaine, free-running temperature rhythms persisted for 2-10 d in SCNx rats. In Experiment 2, robust anticipatory increases in temperature and activity were associated with RF and cocaine injections; however, the feeding periodicity (23.9 h) predominated over the cocaine periodicity. During drug withdrawal, the authors observed two free-running rhythms of temperature and activity that persisted for >14 d in both Sham and SCNx rats. The periods of the free-running rhythms were similar to the feeding entrainment (period = 23.7 and 24.0 h, respectively) and drug entrainment (period = 25.7 and 26.1 h, respectively). Also during withdrawal, the normally close correlation between activity and temperature was greatly disrupted in Sham and SCNx rats. Taken together, these results do not support the existence of a single oscillator mediating the rewarding properties of both food and cocaine. Rather, they suggest that these two highly rewarding behaviors can be temporally isolated, especially during drug withdrawal. Under stable dual-entrainment conditions, food reward appears to exhibit a slightly greater circadian influence than drug reward. The ability to generate free-running temperature rhythms of different frequencies following combined food and drug exposures could reflect a state of internal desynchrony that may contribute to the addiction process and drug relapse.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Stimulants/administration & dosage , Circadian Rhythm/drug effects , Cocaine-Related Disorders/psychology , Cocaine/administration & dosage , Discrimination, Psychological/drug effects , Feeding Behavior , Reward , Animals , Behavior, Addictive/physiopathology , Behavior, Addictive/psychology , Biological Clocks/drug effects , Body Temperature Regulation/drug effects , Caloric Restriction , Cocaine-Related Disorders/physiopathology , Male , Motor Activity/drug effects , Photoperiod , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/physiopathology , Substance Withdrawal Syndrome/psychology , Suprachiasmatic Nucleus/pathology , Suprachiasmatic Nucleus/surgery , Time Factors
8.
World Neurosurg ; 77(2): 342-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22079813

ABSTRACT

BACKGROUND: The anterolateral approach is one of the main routes for accessing suprachiasmatic lesions involving the anterior communicating artery (AComA) complex. Pterional (PT) craniotomy and its alternatives, including orbitozygomatic, orbitopterional, and mini-supraorbital craniotomies, have been developed as tailored frontotemporal craniotomies. One of the main differences between PT craniotomy and its alternatives is the removal of the orbital bone along with the sphenoid wing. However, which bone part is the most important to remove has not been discussed in relation to frontal lobe retraction. We have evaluated how the removal of the supraorbital bar versus the removal of the lateral orbital wall along with the sphenoid wing affects the relationship between the levels of frontal lobe retraction and area of exposure (AOE) in the suprachiasmatic region. METHODS: We performed three types of craniotomies: PT craniotomy, PT craniotomy with the removal of the supraorbital bar (PT-SO craniotomy), and PT craniotomy with the removal of the lateral orbital wall along with the sphenoid wing, i.e., the frontal process of the zygomatic bone and the orbital and cerebral faces of the greater sphenoid wing (PT-LO-SW craniotomy). For each craniotomy, the AOE around the suprachiasmatic region was measured at four different levels of frontal lobe retraction, namely, 5, 10, 15, and 20 mm, from the cranial base. RESULTS: At 5-mm retraction, PT-LO-SW craniotomy was the only craniotomy in which the AComA complex was visible. At 10-mm retraction, PT-LO-SW craniotomy afforded the greatest AOE among the three craniotomies, and the AOE was significantly greater than that of PT craniotomy (P = 0.025). At 15- and 20-mm retraction, there were no significant differences among the three craniotomies. CONCLUSIONS: Treatment of lesions in the suprachiasmatic region via an anterolateral route involving a frontotemporal craniotomy requires sufficient removal of the lateral orbital wall along with the greater sphenoid wing so that brain retraction is minimized.


Subject(s)
Craniotomy/methods , Frontal Bone/anatomy & histology , Frontal Bone/surgery , Skull/anatomy & histology , Skull/surgery , Suprachiasmatic Nucleus/anatomy & histology , Suprachiasmatic Nucleus/surgery , Temporal Bone/anatomy & histology , Temporal Bone/surgery , Female , Humans , Intracranial Aneurysm/surgery , Middle Aged , Middle Cerebral Artery/anatomy & histology , Middle Cerebral Artery/surgery , Orbit/anatomy & histology , Orbit/surgery , Skull Base/anatomy & histology , Skull Base/surgery , Sphenoid Bone/anatomy & histology , Sphenoid Bone/surgery , Surgical Instruments , Tomography, X-Ray Computed , Zygoma/anatomy & histology , Zygoma/surgery
9.
J Vis Exp ; (48)2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21372784

ABSTRACT

A central circadian (~24 hr) clock coordinating daily rhythms in physiology and behavior resides in the suprachiasmatic nucleus (SCN) located in the anterior hypothalamus. The clock is directly synchronized by light via the retina and optic nerve. Circadian oscillations are generated by interacting negative feedback loops of a number of so called "clock genes" and their protein products, including the Period (Per) genes. The core clock is also dependent on membrane depolarization, calcium and cAMP. The SCN shows daily oscillations in clock gene expression, metabolic activity and spontaneous electrical activity. Remarkably, this endogenous cyclic activity persists in adult tissue slices of the SCN. In this way, the biological clock can easily be studied in vitro, allowing molecular, electrophysiological and metabolic investigations of the pacemaker function. The SCN is a small, well-defined bilateral structure located right above the optic chiasm. In the rat it contains ~8.000 neurons in each nucleus and has dimensions of approximately 947 µm (length, rostrocaudal axis) x 424 µm (width) x 390 µm (height). To dissect out the SCN it is necessary to cut a brain slice at the specific level of the brain where the SCN can be identified. Here, we describe the dissecting and slicing procedure of the SCN, which is similar for mouse and rat brains. Further, we show how to culture the dissected tissue organotypically on a membrane, a technique developed for SCN tissue culture by Yamazaki et al. Finally, we demonstrate how transgenic tissue can be used for measuring expression of clock genes/proteins using dynamic luciferase reporter technology, a method that originally was used for circadian measurements by Geusz et al. We here use SCN tissues from the transgenic knock-in PERIOD2::LUCIFERASE mice produced by Yoo et al. The mice contain a fusion protein of PERIOD (PER) 2 and the firefly enzyme LUCIFERASE. When PER2 is translated in the presence of the substrate for luciferase, i.e. luciferin, the PER2 expression can be monitored as bioluminescence when luciferase catalyzes the oxidation of luciferin. The number of emitted photons positively correlates to the amount of produced PER2 protein, and the bioluminescence rhythms match the PER2 protein rhythm in vivo. In this way the cyclic variation in PER2 expression can be continuously monitored real time during many days. The protocol we follow for tissue culturing and real-time bioluminescence recording has been thoroughly described by Yamazaki and Takahashi.


Subject(s)
Biological Clocks/genetics , Luciferases, Firefly/metabolism , Period Circadian Proteins/genetics , Suprachiasmatic Nucleus/physiology , Tissue Culture Techniques/methods , Animals , Dissection , Gene Knock-In Techniques , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Luminescent Measurements/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Period Circadian Proteins/biosynthesis , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/surgery
11.
Eur J Neurosci ; 26(10): 2731-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17973924

ABSTRACT

The mammalian hypothalamic suprachiasmatic nucleus (SCN) is the master oscillator that regulates the circadian rhythms of the peripheral oscillators. Previous studies have demonstrated that the transplantation of embryonic SCN tissues into SCN-lesioned arrhythmic mice restores the behavioral circadian rhythms of these animals. In our present study, we examined the clock gene expression profiles in a transplanted SCN and peripheral tissues, and also analysed the circadian rhythm of the locomotor activity in SCN-grafted mice. These experiments were undertaken to elucidate whether the transplanted SCN generates a dynamic circadian oscillation and maintains the phase relationships that can be detected in intact mice. The grafted SCN indeed showed dynamic circadian expression rhythms of clock genes such as mPeriod1 (mPer1) and mPeriod2 (mPer2). Furthermore, the phase differences between the expression rhythms of these genes in the grafted SCN and the locomotor activity rhythms of the transplanted animals were found to be very similar to those in intact animals. Moreover, in the liver, kidney and skeletal muscles of the transplanted animals, the phase angles between the circadian rhythm of the grafted SCN and that of the peripheral tissues were maintained as in intact animals. However, in the SCN-grafted animals, the amplitudes of the mPer1 and mPer2 rhythms were attenuated in the peripheral tissues. Our current findings therefore indicate that a transplanted SCN has the capacity to generate a dynamic intrinsic circadian oscillation, and can also lock the normal phase angles among the SCN, locomotor activity and peripheral oscillators in a similar manner as in intact control animals.


Subject(s)
Brain Tissue Transplantation , Circadian Rhythm/physiology , Gene Expression/physiology , Nuclear Proteins/metabolism , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/transplantation , Animals , Behavior, Animal , Embryo, Mammalian , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Nuclear Proteins/genetics , Suprachiasmatic Nucleus/injuries , Suprachiasmatic Nucleus/surgery , Time Factors
12.
Exp Biol Med (Maywood) ; 232(3): 390-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17327472

ABSTRACT

Adiponectin is an adipocytokine that modulates energy homeostasis and glucose metabolism. Here, we examined the effects of acute intravenous (iv) and lateral cerebral ventricular (LCV) injections of adiponectin on the renal sympathetic nerve activity (RSNA) and blood pressure (b/p) in urethane-anesthetized rats. Both iv and LCV injections of adiponectin induced dose-dependent suppressions of RSNA and b/p. Moreover, we found that bilateral lesions of the hypothalamic suprachiasmatic nucleus (SCN) abolished the effects of iv injection of adiponectin on RSNA and b/p. These findings suggest that adiponectin decreases the RSNA and b/p in a dose-dependent manner and that the SCN is implicated in mechanism of adiponectin actions on RSNA and b/p. These findings also suggest that the hypotensive-action activity of adiponectin is realized, at least partially, via changes in activities of autonomic nerves activity.


Subject(s)
Adiponectin/pharmacology , Blood Pressure/drug effects , Kidney/innervation , Sympathetic Nervous System/drug effects , Adiponectin/administration & dosage , Adiponectin/blood , Animals , Blood Glucose/metabolism , Dose-Response Relationship, Drug , Heart Rate/drug effects , Hexamethonium/pharmacology , Injections, Intravenous , Male , Rats , Rats, Sprague-Dawley , Suprachiasmatic Nucleus/physiology , Suprachiasmatic Nucleus/surgery , Sympathetic Nervous System/physiology
13.
J Biol Rhythms ; 19(3): 226-37, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15155009

ABSTRACT

Timing of daily torpor was assessed in suprachiasmatic nucleus-ablated (SCNx) and sham-ablated Siberian hamsters fed restricted amounts of food each day either in the light or dark phase of a 14:10 light-dark cycle. Eighty-five percent of sham-ablated and 45% of SCNx hamsters displayed a preferred hour for torpor onset. In each group, time of torpor onset was not random but occurred at a mean hour that differed significantly from chance. Time of food presentation almost completely accounted for the timing of torpor onset in SCNx animals and significantly affected timing of this behavior in intact hamsters. These results suggest that the circadian pacemaker in the SCN controls the time of torpor onset indirectly by affecting timing of food intake, rather than by, or in addition to, direct neural and humoral outputs to relevant target tissues.


Subject(s)
Circadian Rhythm/physiology , Eating , Feeding Behavior/physiology , Motor Activity/physiology , Suprachiasmatic Nucleus , Animals , Biological Clocks/physiology , Caloric Restriction , Cricetinae , Female , Phodopus , Photoperiod , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/pathology , Suprachiasmatic Nucleus/surgery
14.
J Neurosci ; 24(10): 2449-57, 2004 Mar 10.
Article in English | MEDLINE | ID: mdl-15014120

ABSTRACT

The role of the suprachiasmatic nuclei (SCN) in generating circadian rhythms in physiology and behavior is well established. Recent evidence based on clock gene expression indicates that the rodent SCN are composed of at least two functional subdivisions. In Syrian hamsters (Mesocricetus auratus), cells in a subregion of the caudal SCN marked by calbindin-D(28K) (CalB) express light-induced, but not rhythmic, clock genes (Per1, Per2, and Per3). In the SCN region marked by vasopressinergic cells and fibers, clock gene expression is rhythmic. Importantly, lesions of the CalB subregion that spare a significant portion of the SCN abolish rhythms in locomotor behavior. One possibility is that the CalB subregion is required to maintain SCN function necessary to support all behavioral and physiological rhythms. Alternatively, this subregion may control circadian rhythms in locomotor behavior, whereas other circadian responses in physiology and behavior are sustained by different SCN compartments. The present study sought to distinguish between these possibilities by examining the role of the CalB subregion in a battery of rhythms within an individual animal. The results indicate that lesions of the CalB subregion of the SCN abolish circadian rhythms in behavior (locomotion, drinking, gnawing), physiology (body temperature, heart rate), and hormone secretion (melatonin, cortisol), even when other SCN compartments are spared. Together, these findings suggest a novel fundamental property of SCN organization, with a subset of cells being critical for the maintenance of SCN function manifest in circadian rhythms in physiology and behavior.


Subject(s)
Mesocricetus/physiology , Periodicity , Suprachiasmatic Nucleus/anatomy & histology , Suprachiasmatic Nucleus/physiology , Animals , Behavior, Animal/physiology , Body Temperature/physiology , Calbindins , Circadian Rhythm/physiology , Cricetinae , Darkness , Gonads/physiology , Heart Rate/physiology , Hydrocortisone/blood , Male , Melatonin/blood , Motor Activity/physiology , Organ Size , S100 Calcium Binding Protein G/biosynthesis , Suprachiasmatic Nucleus/surgery
15.
J Neurosci ; 24(3): 615-9, 2004 Jan 21.
Article in English | MEDLINE | ID: mdl-14736846

ABSTRACT

The suprachiasmatic nucleus (SCN) of the hypothalamus has been termed the master circadian pacemaker of mammals. Recent discoveries of damped circadian oscillators in other tissues have led to the hypothesis that the SCN synchronizes and sustains daily rhythms in these tissues. We studied the effects of constant lighting (LL) and of SCN lesions on behavioral rhythmicity and Period 1 (Per1) gene activity in the SCN and olfactory bulb (OB). We found that LL had similar effects on cyclic locomotor and feeding behaviors and Per1 expression in the SCN but had no effect on rhythmic Period 1 expression in the OB. LL lengthened the period of locomotor and SCN rhythms by approximately 1.6 hr. After 2 weeks in LL, nearly 35% of rats lost behavioral rhythmicity. Of these, 90% showed no rhythm in Per1-driven expression in their SCN. Returning the animals to constant darkness rapidly restored their daily cycles of running wheel activity and gene expression in the SCN. In contrast, the OB remained rhythmic with no significant change in period, even when cultured from animals that had been behaviorally arrhythmic for 1 month. Similarly, we found that lesions of the SCN abolished circadian rhythms in behavior but not in the OB. Together, these results suggest that LL causes the SCN to lose circadian rhythmicity and its ability to coordinate daily locomotor and feeding rhythms. The SCN, however, is not required to sustain all rhythms because the OB continues to oscillate in vivo when the SCN is arrhythmic or ablated.


Subject(s)
Circadian Rhythm/physiology , Olfactory Bulb/physiology , Suprachiasmatic Nucleus/physiology , Animals , Behavior, Animal/physiology , Biological Clocks/physiology , Cell Cycle Proteins , Eating/physiology , Feeding Behavior/physiology , Light , Luciferases/biosynthesis , Luciferases/genetics , Male , Motor Activity/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Olfactory Bulb/metabolism , Period Circadian Proteins , Photic Stimulation , Photoperiod , Promoter Regions, Genetic , Rats , Suprachiasmatic Nucleus/surgery
16.
J Natl Cancer Inst ; 94(9): 690-7, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11983758

ABSTRACT

BACKGROUND: The circadian timing system controlled by the suprachiasmatic nuclei (SCN) of the hypothalamus regulates daily rhythms of motor activity and adrenocortical secretion. An alteration in these rhythms is associated with poor survival of patients with metastatic colorectal or breast cancer. We developed a mouse model to investigate the consequences of severe circadian dysfunction upon tumor growth. METHODS: The SCN of mice were destroyed by bilateral electrolytic lesions, and body activity and body temperature were recorded with a radio transmitter implanted into the peritoneal cavity. Plasma corticosterone levels and circulating lymphocyte counts were measured (n = 75 with SCN lesions, n = 64 sham-operated). Complete SCN destruction was ascertained postmortem. Mice were inoculated with implants of Glasgow osteosarcoma (n = 16 with SCN lesions, n = 12 sham-operated) or pancreatic adenocarcinoma (n = 13 with SCN lesions, n = 13 sham-operated) tumors to determine the effects of altered circadian rhythms on tumor progression. Time series for body temperature and rest-activity patterns were analyzed by spectral analysis and cosinor analysis. Parametric data were compared by the use of analysis of variance (ANOVA) and survival curves with the log-rank test. All statistical tests were two-sided. RESULTS: The 24-hour rest-activity cycle was ablated and the daily rhythms of serum corticosterone level and lymphocyte count were markedly altered in 75 mice with complete SCN destruction as compared with 64 sham-operated mice (two-way ANOVA for corticosterone: sampling time effect P<.001, lesion effect P =.001, and time x lesion interaction P<.001; for lymphocytes P =.001,.002, and.002 respectively). Body temperature rhythm was suppressed in 60 of the 75 mice with SCN lesions (P<.001). Both types of tumors grew two to three times faster in mice with SCN lesions than in sham-operated mice (two-way ANOVA: P<.001 for lesion and for tumor effects; P =.21 for lesion x tumor effect interaction). Survival of mice with SCN lesions was statistically significantly shorter compared with that of sham-operated mice (log-rank P =.0062). CONCLUSIONS: Disruption of circadian rhythms in mice was associated with accelerated growth of malignant tumors of two types, suggesting that the host circadian clock may play an important role in endogenous control of tumor progression.


Subject(s)
Adenocarcinoma/physiopathology , Circadian Rhythm/physiology , Osteosarcoma/physiopathology , Skin Neoplasms/physiopathology , Animals , Body Temperature , Body Weight , Corticosterone/blood , Disease Models, Animal , Female , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Organ Size , Suprachiasmatic Nucleus/physiology , Suprachiasmatic Nucleus/surgery
17.
Brain Res ; 929(1): 26-36, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11852028

ABSTRACT

Recently we found that food-deprived rats kept under a light-dark cycle showed a progressive reduction in body temperature during the light phase on each subsequent day while body temperature in the dark phase did not differ from baseline values. In this study, we investigated the effect of lesioning the hypothalamic suprachiasmatic nucleus (SCN) on body temperature modulation by food deprivation. In the SCN-lesioned rats in which daily rhythms of body temperature and activity were abolished, body temperature was unchanged by food deprivation. We also examined the effect of food deprivation on the daily changes in Fos expression in the SCN. Under normal fed conditions the number of SCN cells expressing Fos is high during the day and low at night. Food deprivation attenuated the amplitude of this daily change in Fos expression in the SCN. This tendency was prominent in the dorsal part of the SCN, while the ventral part showed no effect of food deprivation. These findings suggest that the SCN plays some role in body temperature modulation due to food deprivation.


Subject(s)
Body Temperature Regulation/physiology , Circadian Rhythm/physiology , Food Deprivation/physiology , Neurons/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Down-Regulation/physiology , Male , Photic Stimulation , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/surgery , Up-Regulation/physiology
18.
J Neurol Neurosurg Psychiatry ; 71(4): 505-14, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11561035

ABSTRACT

OBJECTIVE: To compare the results of visual functional MRI with those of perimetric evaluation in patients with visual field defects and retrochiasmastic tumours and in normal subjects without visual field defect. The potential clinical usefulness of visual functional MRI data during resective surgery was evaluated in patients with occipital lobe tumours. METHODS: Eleven patients with various tumours and visual field defects and 12 normal subjects were studied by fMRI using bimonocular or monocular repetitive photic stimulation (8 Hz). The data obtained were analyzed with the statistical parametric maps software (p<10(-8)) and were compared with the results of Goldmann visual field perimetric evaluation. In patients with occipital brain tumours undergoing surgery, the functional data were registered in a frameless stereotactic device and the images fused into anatomical three standard planes and three dimensional reconstructions of the brain surface. RESULTS: Two studies of patients were discarded, one because of head motion and the other because of badly followed instructions. On the remaining patients the functional activations found in the visual cortex were consistent with the results of perimetric evaluation in all but one of the patients and all the normal subjects although the results of fMRI were highly dependent on the choices of the analysis thresholds. Visual functional MRI image guided data were used in five patients with occipital brain tumours. No added postoperative functional field defect was detected. CONCLUSIONS: There was a good correspondence between fMRI data and the results of perimetric evaluation although dependent on the analysis thresholds. Visual fMRI data registered into a frameless stereotactic device may be useful in surgical planning and tumour removal.


Subject(s)
Brain Neoplasms/surgery , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Occipital Lobe/surgery , Stereotaxic Techniques , Visual Field Tests , Visual Fields/physiology , Adult , Aged , Brain Neoplasms/physiopathology , Female , Humans , Male , Middle Aged , Occipital Lobe/physiopathology , Patient Care Planning , Suprachiasmatic Nucleus/physiopathology , Suprachiasmatic Nucleus/surgery , User-Computer Interface , Visual Cortex/physiopathology , Visual Cortex/surgery
19.
Brain Res ; 909(1-2): 81-91, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11478924

ABSTRACT

The circadian pacemaker in the suprachiasmatic nuclei is primarily synchronized to the daily light-dark cycle. The phase-shifting and synchronizing effects of light can be modulated by non-photic factors, such as behavioral, metabolic or serotonergic cues. The present experiments examine the effects of sleep deprivation on the response of the circadian pacemaker to light and test the possible involvement of serotonergic and/or metabolic cues in mediating the effects of sleep deprivation. Photic phase-shifting of the locomotor activity rhythm was analyzed in mice transferred from a light-dark cycle to constant darkness, and sleep-deprived for 8 h from Zeitgeber Time 6 to Zeitgeber Time 14. Phase-delays in response to a 10-min light pulse at Zeitgeber Time 14 were reduced by 30% in sleep-deprived mice compared to control mice, while sleep deprivation without light exposure induced no significant phase-shifts. Stimulation of serotonin neurotransmission by fluoxetine (10 mg/kg), a serotonin reuptake inhibitor that decreases light-induced phase-delays in non-deprived mice, did not further reduce light-induced phase-delays in sleep-deprived mice. Impairment of serotonin neurotransmission with p-chloroamphetamine (three injections of 10 mg/kg), which did not increase light-induced phase-delays in non-deprived mice significantly, partially normalized light-induced phase-delays in sleep-deprived mice. Injections of glucose increased light-induced phase-delays in control and sleep-deprived mice. Chemical damage of the ventromedial hypothalamus by gold-thioglucose (600 mg/kg) prevented the reduction of light-induced phase-delays in sleep-deprived mice, without altering phase-delays in control mice. Taken together, the present results indicate that sleep deprivation can reduce the light-induced phase-shifts of the mouse suprachiasmatic pacemaker, due to serotonergic and metabolic changes associated with the loss of sleep.


Subject(s)
Circadian Rhythm/physiology , Energy Metabolism/physiology , Neurons/metabolism , Serotonin/metabolism , Sleep Deprivation/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Aurothioglucose/pharmacokinetics , Body Weight/drug effects , Body Weight/physiology , Circadian Rhythm/drug effects , Denervation , Energy Metabolism/drug effects , Gliosis/chemically induced , Gliosis/pathology , Gliosis/physiopathology , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurons/drug effects , Photic Stimulation , Sleep Deprivation/physiopathology , Stress, Physiological/metabolism , Stress, Physiological/physiopathology , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/surgery
20.
Endocrinology ; 142(6): 2677-85, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356719

ABSTRACT

At present it is not clear which factors are responsible for the diurnal pattern of plasma leptin levels, although the timing of food intake and circulating hormones such as glucocorticoids and insulin have both been proposed as independent determinants. In this study we show that ablation of the biological clock by thermal lesions of the hypothalamic suprachiasmatic nucleus (SCN) completely eliminates the diurnal pattern of plasma leptin levels. By contrast, removal of the diurnal corticosterone signal by adrenalectomy and corticosterone replacement did not affect diurnal plasma leptin levels. More importantly, removal of the nocturnal feeding signal by submitting the animals to a regular feeding schedule of six meals per day did not abolish the diurnal plasma leptin levels. However, both SCN lesions and the regular feeding schedule did cause an increase in the 24-h mean plasma leptin levels. As neither rhythmic feeding, insulin, or corticosterone signals can completely explain the diurnal plasma leptin rhythm, we conclude that biological clock control of the sympathetic input to the adipocyte is essential for regulation of the daily rhythm in leptin release.


Subject(s)
Circadian Rhythm , Leptin/metabolism , Suprachiasmatic Nucleus/physiology , Adipocytes/chemistry , Adipose Tissue/innervation , Adipose Tissue/metabolism , Adrenalectomy , Animals , Corticosterone/blood , Corticosterone/pharmacology , Eating , Leptin/genetics , Male , RNA, Messenger/analysis , Rats , Rats, Wistar , Suprachiasmatic Nucleus/surgery , Sympathetic Nervous System/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...