Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
1.
Eur J Neurosci ; 46(3): 1875-1886, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28661071

ABSTRACT

Travelling across several time zones requires a fast adjustment of the circadian system and the differential adjustment speeds of organs and systems results in what is commonly referred as jet lag. During this transitory state of circadian disruption, individuals feel discomfort, appetite loss, fatigue, disturbed sleep and deficient performance of multiple tasks. We have demonstrated that after a 6-h phase advance of the light-dark cycle (LD) scheduled food in phase with the new night onset can speed up re-entrainment. In this study, we explored the possible mechanisms underlying the fast re-entrainment due to the feeding schedule. We focused on first- and second-order structures that provide metabolic information to the suprachiasmatic nucleus (SCN). We compared (i) control rats without change in LD cycle; (ii) rats exposed to a 6-h phase advance of the LD cycle with food ad libitum; and (iii) rats exposed to the 6-h phase advance combined with food access in phase with the new night. We found an immediate synchronizing effect of food on stomach distention and on c-Fos expression in the nucleus of the solitary tract, arcuate nucleus of the hypothalamus, dorsomedial hypothalamic nucleus and paraventricular nucleus. These observations indicate that in a model of jet lag, scheduled feeding can favour an immediate shift in first- and second-order relays to the SCN and that by keeping feeding schedules coupled to the new night, a fast re-entrainment may be achieved by shifting peripheral and extra-SCN oscillations.


Subject(s)
Circadian Rhythm , Hypothalamus/physiology , Jet Lag Syndrome/physiopathology , Meals/physiology , Photoperiod , Animals , Feeding Behavior , Hypothalamus/physiopathology , Male , Rats , Rats, Wistar
2.
Endocrinology ; 157(9): 3439-51, 2016 09.
Article in English | MEDLINE | ID: mdl-27429160

ABSTRACT

The suprachiasmatic nucleus (SCN) and arcuate nucleus (ARC) have reciprocal connections; catabolic metabolic information activates the ARC and inhibits SCN neuronal activity. Little is known about the influence of the SCN on the ARC. Here, we investigated whether the SCN modulated the sensitivity of the ARC to catabolic metabolic conditions. ARC neuronal activity, as determined by c-Fos immunoreactivity, was increased after a hypoglycemic stimulus by 2-deoxyglucose (2DG). The highest ARC neuronal activity after 2DG was found at the end of the light period (zeitgeber 11, ZT11) with a lower activity in the beginning of the light period (zeitgeber 2, ZT2), suggesting the involvement of the SCN. The higher activation of ARC neurons after 2DG at ZT11 was associated with higher 2DG induced blood glucose levels as compared with ZT2. Unilateral SCN-lesioned animals, gave a mainly ipsilateral activation of ARC neurons at the lesioned side, suggesting an inhibitory role of the SCN on ARC neurons. The 2DG-induced counterregulatory glucose response correlated with increased ARC neuronal activity and was significantly higher in unilateral SCN-lesioned animals. Finally, the ARC as site where 2DG may, at least partly, induce a counterregulatory response was confirmed by local microdialysis of 2DG. 2DG administration in the ARC produced a higher increase in circulating glucose compared with 2DG administration in surrounding areas such as the ventromedial nucleus of the hypothalamus (VMH). We conclude that the SCN uses neuronal pathways to the ARC to gate sensory metabolic information to the brain, regulating ARC glucose sensitivity and counterregulatory responses to hypoglycemic conditions.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Hypoglycemia/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Arcuate Nucleus of Hypothalamus/anatomy & histology , Fasting/metabolism , Male , Melanocyte-Stimulating Hormones/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , Rats, Wistar , Suprachiasmatic Nucleus/anatomy & histology
3.
Obes Rev ; 16(10): 871-82, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26214605

ABSTRACT

The circadian system, headed by the suprachiasmatic nucleus, synchronizes behaviour and metabolism according to the external light-dark cycle through neuroendocrine and autonomic signals. Metabolic diseases, such as steatosis, obesity and glucose intolerance, have been associated with conditions of circadian misalignment wherein the feeding schedule has been moved to the resting phase. Here we describe the physiological processes involved in liver lipid accumulation and show how they follow a circadian pattern importantly regulated by both the autonomic nervous system and the feeding-fasting cycle. We propose that an unbalanced activity of the sympathetic-parasympathetic branches between organs induced by circadian misalignment provides the conditions for the development and progression of non-alcoholic fatty liver disease.


Subject(s)
Autonomic Nervous System/physiopathology , Fatty Acids, Nonesterified/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Period Circadian Proteins/metabolism , Adipose Tissue , Circadian Rhythm , Gene Expression Regulation , Humans , Lipolysis , Molecular Sequence Data , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/physiopathology , Obesity/complications , Obesity/physiopathology
4.
Neuroscience ; 266: 197-207, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24583038

ABSTRACT

The suprachiasmatic nucleus (SCN) is typically considered our autonomous clock synchronizing behavior with physiological parameters such as blood pressure (BP), just transmitting time independent of physiology. Yet several studies show that the SCN is involved in the etiology of hypertension. Here, we demonstrate that the SCN is incorporated in a neuronal feedback circuit arising from the nucleus tractus solitarius (NTS), modulating cardiovascular reactivity. Tracer injections into the SCN of male Wistar rats revealed retrogradely filled neurons in the caudal NTS, where BP information is integrated. These NTS projections to the SCN were shown to be glutamatergic and to terminate in the ventrolateral part of the SCN where light information also enters. BP elevations not only induced increased neuronal activity as measured by c-Fos in the NTS but also in the SCN. Lesioning the caudal NTS prevented this activation. The increase of SCN neuronal activity by hypertensive stimuli suggested involvement of the SCN in counteracting BP elevations. Examining this possibility we observed that elevation of BP, induced by α1-agonist infusion, was more than twice the magnitude in SCN-lesioned animals as compared to in controls, indicating indeed an active involvement of the SCN in short-term BP regulation. We propose that the SCN receives BP information directly from the NTS enabling it to react to hemodynamic perturbations, suggesting the SCN to be part of a homeostatic circuit adapting BP response. We discuss how these findings could explain why lifestyle conditions violating signals of the biological clock may, in the long-term, result in cardiovascular disease.


Subject(s)
Blood Pressure/physiology , Neural Pathways/physiology , Solitary Nucleus/physiology , Suprachiasmatic Nucleus/physiology , Animals , Feedback , Immunohistochemistry , Male , Neural Pathways/anatomy & histology , Rats , Rats, Wistar , Solitary Nucleus/anatomy & histology , Suprachiasmatic Nucleus/anatomy & histology
5.
Neuroscience ; 265: 184-95, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24508747

ABSTRACT

The arcuate nucleus is the main receptive area of the brain for peripheral and central metabolic cues and its integrity is essential for the maintenance of energy homeostasis. In the arcuate nucleus, different neuronal populations process metabolic signals and transmit this information to other nuclei of the hypothalamus by means of neurotransmitters and a combination of neuropeptides whose expression is modulated by the nutritional status. Here we investigated the changes in expression and synthesis of the polypeptide VGF in the arcuate nucleus of rats, in relation to the two main categories of neurons that show colocalization with VGF: the orexigenic NPY-expressing cells and the anorexigenic POMC-expressing cells. The results show that fasting is the most important stimulus for VGF expression, and that the up-regulation of VGF mRNA is restricted to the NPY area of the arcuate nucleus. POMC neurons express VGF under all feeding conditions, but especially in ad libitum-fed and fasted-refed animals. We also show that VGF arcuate neurons project to the pre-autonomic neurons of the paraventricular nucleus of the hypothalamus, providing anatomical evidence suggesting VGF as a central modulator of the autonomic nervous system.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Energy Metabolism , Neurons/metabolism , Neuropeptides/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Animals , Fasting/metabolism , Homeostasis , Male , Rats , Rats, Wistar
6.
Endocrinology ; 155(2): 525-35, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24265453

ABSTRACT

Timing of metabolic processes is crucial for balanced physiology; many studies have shown the deleterious effects of untimely food intake. The basis for this might be an interaction between the arcuate nucleus (ARC) as the main integration site for metabolic information and the suprachiasmatic nucleus (SCN) as the master clock. Here we show in male rats that the SCN influences ARC daily neuronal activity by imposing a daily rhythm on the α-MSH neurons with a peak in neuronal activity at the end of the dark phase. Bilateral SCN lesions showed a complete disappearance of ARC neuronal rhythms and unilateral SCN lesions showed a decreased activation in the ARC at the lesioned side. Moreover light exposure during the dark phase inhibited ARC and α-MSH neuronal activity. The daily inhibition of ARC neuronal activity occurred in light-dark conditions as well as in dark-dark conditions, demonstrating the inhibitory effect to be mediated by increased SCN (subjective) day neuronal activity. Injections into the SCN with the neuronal tracer cholera toxin B showed that α-MSH neurons receive direct projections from the SCN. The present study demonstrates that the SCN activates and possibly also inhibits depending on the moment of the circadian cycle ARC α-MSH neurons via direct neuronal input. The persistence of these activity patterns in fasted animals demonstrates that this SCN-ARC interaction is not necessarily satiety associated but may support physiological functions associated with changes in the sleep-wake cycle.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Circadian Rhythm/physiology , Neurons/physiology , Suprachiasmatic Nucleus/physiology , alpha-MSH/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Male , Motor Activity/physiology , Neural Pathways/metabolism , Neural Pathways/physiology , Neurons/metabolism , Rats , Rats, Wistar , Suprachiasmatic Nucleus/metabolism
7.
Eur J Vasc Endovasc Surg ; 46(5): 542-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24091093

ABSTRACT

OBJECTIVES: Abdominal aortic aneurysm (AAA) is a major cause of death in developed countries. The AAA diameter is still the only validated prognostic measure for rupture, and therapeutic interventions are initiated accordingly. This still leads to unnecessary interventions in some cases or unidentified impending ruptures. Vascular calcification has been validated abundantly as a risk factor in the cardiovascular field and may strengthen the rupture risk assessment of the AAA. With this study we aim to assess the correlation between AAA calcification and rupture risk in a retrospective unmatched case-control population. METHODS: A database of 334 AAA patients was evaluated. Three groups were formed: elective (eAAA; n = 233), ruptured (rAAA; n = 73) and symptomatic non-ruptured (sAAA; n = 28) AAA patients. The Abdominal Aortic Calcification-8 score (AAC-8) was used to measure the severity of vascular calcification. RESULTS: The AAA diameter (61 ± 12 mm vs. 74 ± 21 mm; p < .001) and AAC-8 score (3.4 ± 2 points vs. 4.9 ± 2.3 points; p < .001) of the eAAA and the combined rAAA and sAAA groups, respectively, were significantly different after univariate analysis. Multivariate analysis showed that larger AAA diameter (odds ratio [OR]: 1.048/mm increase; 95% confidence interval [CI]: 1.042-1.082; p < .001) and a higher AAC-8 score (OR: 1.34/point increase; 95% CI: 1.19-1.53; p < .001) were significantly associated with development into a sAAA or rAAA. Peripheral artery disease was significantly correlated to eventual elective treatment (OR: 0.39; 95% CI: .15-1; p = .049). CONCLUSION: This study suggests a trend of an increased degree of calcification in symptomatic or even ruptured AAA patients compared with elective AAA patients.


Subject(s)
Aortic Aneurysm, Abdominal/complications , Aortic Rupture/etiology , Vascular Calcification/complications , Aged , Aged, 80 and over , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/surgery , Aortic Rupture/diagnostic imaging , Aortic Rupture/surgery , Aortography/methods , Elective Surgical Procedures , Emergencies , Female , Humans , Logistic Models , Male , Middle Aged , Multivariate Analysis , Odds Ratio , Predictive Value of Tests , Prognosis , Retrospective Studies , Risk Factors , Severity of Illness Index , Tomography, X-Ray Computed , Vascular Calcification/diagnostic imaging , Vascular Calcification/surgery , Vascular Surgical Procedures
8.
Neuroscience ; 246: 291-300, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23680526

ABSTRACT

The intergeniculate leaflet (IGL) is classically known as the area of the Thalamic Lateral Geniculate Complex providing the suprachiasmatic nucleus (SCN) non-photic information. In the present study we investigated whether this information might be related to the metabolic state of the animal. The following groups of male Wistar rats were used for analysis of neuropeptide Y (NPY) and c-Fos in the IGL and SCN. (1) Fed ad libitum. (2) Fasted for 48 h. (3) Fasted for 48 h followed by refeeding for 3 h. (4) Monosodium glutamate-lesioned and 48 h fasted. (5) Electrolytic lesion in the IGL and 48 h fasted. The results were quantified by optical densitometry. Neuronal tracers were injected in two brain areas that receive metabolic information from the periphery, the arcuate nucleus (ARC) and Nucleus of the Tractus Solitarius to investigate whether there is an anatomical relationship with the IGL. Lesion studies showed the IGL, and not the ARC, as origin of most NPY projections to the SCN. Fasting induced important changes in the NPY expression in the IGL, coinciding with similar changes of NPY/glutamate decarboxylase projections of the IGL to the SCN. These changes revealed that the IGL is involved in the transmission of metabolic information to the SCN. In fasted animals IGL lesion resulted in a significant increase of c-Fos in the SCN as compared to intact fasted animals demonstrating the inhibitory influence of the IGL to the SCN in fasting conditions. When the animal after fasting was refed, an increase of c-Fos in the SCN indicated a removal of this inhibitory input. Together these observations show that in addition to increased inhibitory IGL input during fasting, the negative metabolic condition also results in increased excitatory input to the SCN via other pathways. Consequently the present observations show that at least part of the non-photic input to the SCN, arising from the IGL contains information about metabolic conditions.


Subject(s)
Geniculate Bodies/metabolism , Neuropeptide Y/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Fasting/metabolism , Male , Neural Pathways/metabolism , Random Allocation , Rats , Rats, Wistar
9.
Neurogastroenterol Motil ; 24(2): 191-200, e93, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22118533

ABSTRACT

BACKGROUND: The cholinergic anti-inflammatory pathway is proposed to be part of the so-called vago-vagal 'inflammatory reflex'. The aim of this study is to provide neuro-anatomical evidence to support the existence of a functional neuronal circuit and its activation in response to intestinal inflammation. METHODS: The expression of c-fos was evaluated at different levels of the neurocircuitry in the course of postoperative ileus (POI) in a mouse model. Specific activation of the motor neurons innervating the inflamed intestine and the spleen was monitored by retrograde tracing using cholera toxin-b. The role of the vagal afferent pathway nerve was evaluated by selective vagal denervation of the intestine. KEY RESULTS: Abdominal surgery resulted in subtle inflammation of the manipulated intestine at 24 h (late phase), but not after 2 and 6 h (early) after surgery. This local inflammation was associated with activation of neurons in the nucleus of the solitary tract and in the dorsal nucleus of the vagus. The vagal output mainly targeted the inflamed zone: 42% of motor neurons innervating the intestine expressed c-fos IR in contrast to 7% of those innervating the spleen. Vagal denervation of the intestine abolished c-fos expression in the brain nuclei involved in the neuronal network activated by intestinal inflammation. CONCLUSIONS & INFERENCES: Our data demonstrate that intestinal inflammation triggers a vagally mediated circuit leading mainly to activation of vagal motor neurons connected to the inflamed intestine. These findings for the first time provide neuro-anatomical evidence for the existence of the endogenous 'inflammatory reflex' and its activation during inflammation.


Subject(s)
Intestines/physiology , Neurons/physiology , Reflex/physiology , Vagus Nerve/physiology , Afferent Pathways/physiology , Animals , Cholinergic Neurons/physiology , Ileus/metabolism , Mice , Proto-Oncogene Proteins c-fos/metabolism , Spleen/physiology
10.
Mol Cell Endocrinol ; 349(1): 20-9, 2012 Feb 05.
Article in English | MEDLINE | ID: mdl-21782883

ABSTRACT

The pronounced daily variation in the release of adrenal hormones has been at the heart of the deciphering and understanding of the circadian timing system. Indeed, the first demonstration of an endocrine day/night rhythm was provided by Pincus (1943), by showing a daily pattern of 17-keto-steroid excretion in the urine of 7 healthy males. Twenty years later the adrenal gland was one of the very first organs to show, in vitro, that circadian rhythmicity was maintained. In the seventies, experimental manipulation of the daily corticosterone rhythm served as evidence for the identification of respectively the light- and food-entrainable oscillator. Another 20 years later the hypothalamo-pituitary-adrenal (HPA)-axis was key in furthering our understanding of the way in which rhythmic signals generated by the central pacemaker in the hypothalamic suprachiasmatic nuclei (SCN) are forwarded to the rest of the brain and to the organism as a whole. To date, the adrenal gland is still of prime importance for understanding how the oscillations of clock genes in peripheral tissues result in functional rhythms of these tissues, whereas it has become even more evident that adrenal glucocorticoids are key in the resetting of the circadian system after a phase-shift. The HPA-axis thus still is an excellent model for studying the transmission of circadian information in the body.


Subject(s)
Circadian Rhythm , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Adrenal Glands/metabolism , Animals , Anticipation, Psychological/physiology , Circadian Clocks , Hormones/blood , Hormones/metabolism , Hormones/physiology , Humans , Hypothalamus/anatomy & histology , Hypothalamus/metabolism
11.
J Biol Rhythms ; 26(4): 324-34, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21775291

ABSTRACT

Circadian desynchrony occurs when individuals are exposed to abrupt phase shifts of the light-dark cycle, as in jet lag. For reducing symptoms and for speeding up resynchronization, several strategies have been suggested, including scheduled exercise, exposure to bright light, drugs, and especially exogenous melatonin administration. Restricted feeding schedules have shown to be powerful entraining signals for metabolic and hormonal daily cycles, as well as for clock genes in tissues and organs of the periphery. This study explored in a rat model of jet lag the contribution of exogenous melatonin or scheduled feeding on the re-entrainment speed of spontaneous general activity and core temperature after a 6-h phase advance of the light-dark cycle. In a first phase, the treatment was scheduled for 5 days prior to the phase shift, while in a second stage, the treatment was simultaneous with the phase advance of the light-dark cycle. Melatonin administration and especially scheduled feeding simultaneous with the phase shift improved significantly the re-entrainment speed. The evaluation of the free-running activity and temperature following the 5-day treatment proved that both exogenous melatonin and specially scheduled feeding accelerated re-entrainment of the SCN-driven general activity and core temperature, respectively, with 7, 5 days (p < 0.01) and 3, 3 days (p < 0.001). The present results show the relevance of feeding schedules as entraining signals for the circadian system and highlight the importance of using them as a strategy for preventing internal desynchrony.


Subject(s)
Circadian Rhythm , Darkness , Food , Light , Melatonin/administration & dosage , Animals , Behavior, Animal , Male , Rats , Rats, Wistar
12.
Br J Pharmacol ; 163(5): 1007-16, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21371006

ABSTRACT

BACKGROUND AND PURPOSE: Electrical stimulation of the vagus nerve reduces intestinal inflammation following mechanical handling, thereby shortening post-operative ileus in mice. Previous studies in a sepsis model showed that this cholinergic anti-inflammatory pathway can be activated pharmacologically by central administration of semapimod, an inhibitor of p38 mitogen-activated protein kinase. We therefore evaluated the effect of intracerebroventricular (i.c.v.) semapimod on intestinal inflammation and post-operative ileus in mice. EXPERIMENTAL APPROACH: Mice underwent a laparotomy or intestinal manipulation 1 h after i.c.v. pre-treatment with semapimod (1 µg·kg(-1) ) or saline. Drugs were administered through a cannula placed in the left lateral ventricle 1 week prior to experimentation. Twenty-four hours after surgery, gastric emptying was measured using scintigraphy, and the degree of intestinal inflammation was assessed. Finally, activation of brain regions was assessed using quantitative immunohistochemistry for c-fos. KEY RESULTS: Intestinal manipulation induced inflammation of the manipulated intestine and significantly delayed gastric emptying, 24 h after surgery in saline-treated animals. Semapimod significantly reduced this inflammation and improved gastric emptying. Vagotomy enhanced the inflammatory response induced by intestinal manipulation and abolished the anti-inflammatory effect of semapimod. Semapimod but not saline induced a significant increase in c-fos expression in the paraventricular nucleus, the nucleus of the solitary tract and the dorsal motor nucleus of the vagus nerve. CONCLUSIONS AND IMPLICATIONS: Our findings show that i.c.v. semapimod reduces manipulation-induced intestinal inflammation and prevented post-operative ileus. This anti-inflammatory effect depends on central activation of the vagus nerve.


Subject(s)
Acetylcholine/agonists , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Enteritis/prevention & control , Hydrazones/therapeutic use , Ileus/prevention & control , Postoperative Complications/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Enteritis/etiology , Enteritis/immunology , Enteritis/metabolism , Female , Gastric Emptying/drug effects , Hydrazones/administration & dosage , Ileus/etiology , Ileus/immunology , Ileus/metabolism , Injections, Intraventricular , Mice , Mice, Inbred BALB C , Motor Neurons/drug effects , Motor Neurons/metabolism , Postoperative Complications/etiology , Postoperative Complications/immunology , Postoperative Complications/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Vagotomy , Vagus Nerve/drug effects , Vagus Nerve/metabolism
13.
J Neuroendocrinol ; 22(5): 362-72, 2010 May.
Article in English | MEDLINE | ID: mdl-20088910

ABSTRACT

The physiological effects of vasopressin as a peripheral hormone were first reported more than 100 years ago. However, it was not until the first immunocytochemical studies were carried out in the early 1970s, using vasopressin antibodies, and the discovery of an extensive distribution of vasopressin-containing fibres outside the hypothalamus, that a neurotransmitter role for vasopressin could be hypothesised. These studies revealed four additional vasopressin systems next to the classical magnocellular vasopressin system in the paraventricular and supraoptic nuclei: a sexually dimorphic system originating from the bed nucleus of the stria terminalis and the medial amygdala, an autonomic and endocrine system originating from the medial part of the paraventricular nucleus, and the circadian system originating from the hypothalamic suprachiasmatic nuclei (SCN). At about the same time as the discovery of the neurotransmitter function of vasopressin, it also became clear that the SCN contain the main component of the mammalian biological clock system (i.e. the endogenous pacemaker). This review will concentrate on the significance of the vasopressin neurones in the SCN for the functional output of the biological clock that is contained within it. The vasopressin-containing subpopulation is a characteristic feature of the SCN in many species, including humans. The activity of the vasopressin neurones in the SCN shows a pronounced daily variation in its activity that has also been demonstrated in human post-mortem brains. Animal experiments show an important role for SCN-derived vasopressin in the control of neuroendocrine day/night rhythms such as that of the hypothalamic-pituitary-adrenal and hypothalamic-pituitary-gonadal axes. The remarkable correlation between a diminished presence of vasopressin in the SCN and a deterioration of sleep-wake rhythms during ageing and depression make it likely that, also in humans, the vasopressin neurones contribute considerably to the rhythmic output of the SCN.


Subject(s)
Biological Clocks/physiology , Hypothalamus/physiology , Vasopressins/physiology , Animals , Humans , Male , Signal Transduction , Suprachiasmatic Nucleus/physiology , Suprachiasmatic Nucleus/physiopathology
14.
Neuroscience ; 165(4): 1115-26, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20004704

ABSTRACT

Daily feeding schedules entrain temporal patterns of behavior, metabolism, neuronal activity and clock gene expression in several brain areas and periphery while the suprachiasmatic nucleus (SCN), the biological clock, remains coupled to the light/dark cycle. Because bilateral lesions of the SCN do not abolish food entrained behavioral and hormonal rhythms it is suggested that food entrained and light entrained systems are independent of each other. Special circumstances indicate a possible interaction between the light and the food entrained systems and indicate modulation of SCN activity by restricted feeding. This study explores the influence of the SCN on food entrained rhythms. Food entrained temporal profiles of behavior, core temperature, corticosterone and glucose, as well as Fos and PER1 immunoreactivity in the hypothalamus and corticolimbic structures were explored in rats bearing bilateral SCN lesions (SCNX). In SCNX rats food anticipatory activity and the food entrained temperature and corticosterone increase were expressed with earlier onset and higher values than in intact controls. Glucose levels were lower in SCNX rats in all time points and SCNX rats anticipation to a meal induced higher c-Fos positive neurons in the hypothalamus, while a decreased c-Fos response was observed in corticolimbic structures. SCNX rats also exhibited an upregulation of the PER1 peak in hypothalamic structures, especially in the dorsomedial hypothalamic nucleus (DMH), while in some limbic structures PER1 rhythmicity was dampened. The present results indicate that the SCN participates actively during food entrainment modulating the response of hypothalamic and corticolimbic structures, resulting in an increased anticipatory response.


Subject(s)
Circadian Rhythm/physiology , Feeding Behavior/physiology , Suprachiasmatic Nucleus/physiology , Animals , Behavior, Animal/physiology , Body Temperature/physiology , Brain/physiology , Corticosterone/metabolism , Glucose/metabolism , Male , Neurons/physiology , Period Circadian Proteins/metabolism , Periodicity , Photoperiod , Proto-Oncogene Proteins c-fos/metabolism , Random Allocation , Rats , Rats, Wistar
15.
Neuroscience ; 155(1): 297-307, 2008 Jul 31.
Article in English | MEDLINE | ID: mdl-18585440

ABSTRACT

The clock gene protein Per 1 (PER1) is expressed in several brain structures and oscillates associated with the suprachiasmatic nucleus (SCN). Restricted feeding schedules (RFS) induce anticipatory activity and impose daily oscillations of c-Fos and clock proteins in brain structures. Daily access to a palatable treat (chocolate) also elicits anticipatory activity and induces c-Fos expression mainly in corticolimbic structures. Here the influence of daily access to food or chocolate was explored by the analysis of the oscillatory patterns of PER1 in hypothalamic and corticolimbic structures. Wistar rats were exposed to RFS or to daily access to chocolate for 3 weeks. Persistence of food or chocolate entrained rhythms was determined 8 days after cessation of the feeding protocols. RFS and chocolate induced a phase shift in PER1 rhythmicity in corticolimbic structures with peak values at zeitgeber time 12 and a higher amplitude in the chocolate group. Both RFS and chocolate groups showed an upregulation of PER1 in the SCN. Food and chocolate entrained rhythms persisted for 8 days in behavior and in PER1 expression in the dorsomedial hypothalamic nucleus, accumbens, prefrontal cortex and central amygdala. The present data demonstrate the existence of different oscillatory systems in the brain that can be activated by entrainment to metabolic stimuli or to reward and suggest the participation of PER1 in both entraining pathways. Persistence and amplification of PER1 oscillations in structures associated with reward suggest that this oscillatory process is fundamental to food addictive behavior.


Subject(s)
Brain/metabolism , Cell Cycle Proteins/metabolism , Circadian Rhythm/physiology , Feeding Behavior/physiology , Motivation , Reward , Analysis of Variance , Animals , Behavior, Animal , Brain/anatomy & histology , Cacao , Cell Count , Food , Male , Motor Activity/physiology , Period Circadian Proteins , Rats , Rats, Wistar
16.
Neuroscience ; 149(3): 508-17, 2007 Nov 09.
Article in English | MEDLINE | ID: mdl-17920204

ABSTRACT

We recently discovered that human activity possesses a complex temporal organization characterized by scale-invariant/self-similar fluctuations from seconds to approximately 4 h-(statistical properties of fluctuations remain the same at different time scales). Here, we show that scale-invariant activity patterns are essentially identical in humans and rats, and exist for up to approximately 24 h: six-times longer than previously reported. Theoretically, such scale-invariant patterns can be produced by a neural network of interacting control nodes-system components with feedback loops-operating at different time scales. However such control nodes have not yet been identified in any neurophysiological model of scale invariance/self-similarity in mammals. Here we demonstrate that the endogenous circadian pacemaker (suprachiasmatic nucleus; SCN), known to modulate locomotor activity with a periodicity of approximately 24 h, also acts as a major neural control node responsible for the generation of scale-invariant locomotor patterns over a broad range of time scales from minutes to at least 24 h (rather than solely at approximately 24 h). Remarkably, we found that SCN lesion in rats completely abolished the scale-invariant locomotor patterns between 4 and 24 h and significantly altered the patterns at time scales <4 h. Identification of the control nodes of a neural network responsible for scale invariance is the critical first step in understanding the neurophysiological origin of scale invariance/self-similarity.


Subject(s)
Circadian Rhythm/physiology , Suprachiasmatic Nucleus/physiology , Activity Cycles/physiology , Adult , Animals , Darkness , Data Interpretation, Statistical , Female , Humans , Light , Male , Motor Activity/physiology , Rats , Rats, Wistar
17.
J Biol Rhythms ; 21(6): 458-69, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17107936

ABSTRACT

The circadian clock in the suprachiasmatic nucleus (SCN) is composed of thousands of oscillator neurons, each dependent on the cell-autonomous action of a defined set of circadian clock genes. Still, the major question remains how these individual oscillators are organized into a biological clock producing a coherent output able to time all the different daily changes in behavior and physiology. In the present review, the authors discuss the anatomical connections and neurotransmitters used by the SCN to control the daily rhythms in hormone release. The efferent SCN projections mainly target neurons in the medial hypothalamus surrounding the SCN. The activity of these preautonomic and neuroendocrine target neurons is controlled by differentially timed waves of, among others, vasopressin, GABA, and glutamate release from SCN terminals. Together, the data on the SCN control of neuroendocrine rhythms provide clear evidence not only that the SCN consists of phenotypically (i.e., according to neurotransmitter content) different subpopulations of neurons but also that subpopulations should be distinguished (within phenotypically similar groups of neurons) based on the acrophase of their (electrical) activity. Moreover, the specialization of the SCN may go as far as a single body structure, that is, the SCN seems to contain neurons that specifically target the liver, pineal, and adrenal.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Suprachiasmatic Nucleus/physiology , Animals , Autonomic Nervous System/physiology , Humans , Neurons/physiology , Suprachiasmatic Nucleus/metabolism , Vasopressins/physiology
18.
Ned Tijdschr Geneeskd ; 150(36): 1971-5, 2006 Sep 09.
Article in Dutch | MEDLINE | ID: mdl-17002185

ABSTRACT

The number of indications for the medical use of melatonin is slowly increasing. Melatonin is produced by the pineal gland and is a key signal in the circadian rhythm of the body. Melatonin plays an obvious role in the pathophysiology and treatment of sleep disorders and jetlag. Recent research has also demonstrated its favourable effect on blood-pressure regulation. By analogy, melatonin may play a role in a variety of other circadian processes. However, research into the precise effects is still insufficient.


Subject(s)
Circadian Rhythm/physiology , Melatonin/physiology , Melatonin/therapeutic use , Pineal Gland/physiology , Blood Pressure/physiology , Humans , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/etiology , Travel
20.
Chronobiol Int ; 23(3): 521-35, 2006.
Article in English | MEDLINE | ID: mdl-16753939

ABSTRACT

The circadian clock in the suprachiasmatic nuclei (SCN) is composed of thousands of oscillator neurons, each dependent on the cell-autonomous action of a defined set of circadian clock genes. A major question is still how these individual oscillators are organized into a biological clock that produces a coherent output capable of timing all the different daily changes in behavior and physiology. We investigated which anatomical connections and neurotransmitters are used by the biological clock to control the daily release pattern of a number of hormones. The picture that emerged shows projections contacting target neurons in the medial hypothalamus surrounding the SCN. The activity of these pre-autonomic and neuro-endocrine target neurons is controlled by differentially timed waves of vasopressin, GABA, and glutamate release from SCN terminals, among other factors. Together our data indicate that, with regard to the timing of their main release period within the LD cycle, at least four subpopulations of SCN neurons should be discernible. The different subgroups do not necessarily follow the phenotypic differences among SCN neurons. Thus, different subgroups can be found within neuron populations containing the same neurotransmitter. Remarkably, a similar distinction of four differentially timed subpopulations of SCN neurons was recently also discovered in experiments determining the temporal patterns of rhythmicity in individual SCN neurons by way of the electrophysiology or clock gene expression. Moreover, the specialization of the SCN may go as far as a single body structure, i.e., the SCN seems to contain neurons that specifically target the liver, pineal gland, and adrenal gland.


Subject(s)
Autonomic Nervous System/physiology , Biological Clocks/physiology , Animals , Circadian Rhythm/physiology , Glutamic Acid/physiology , Humans , Melatonin/physiology , Neurons/classification , Neurons/physiology , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/physiology , Vasopressins/physiology , gamma-Aminobutyric Acid/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...