Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 190
1.
Neuropharmacology ; : 110007, 2024 May 23.
Article En | MEDLINE | ID: mdl-38795953

Feeding, like many other biological functions, displays a daily rhythm. This daily rhythmicity is controlled by the circadian timing system of which the central master clock is located in the hypothalamic suprachiasmatic nucleus (SCN). Other brain areas and tissues throughout the body also display rhythmic functions and contain the molecular clock mechanism known as peripheral oscillators. To generate the daily feeding rhythm, the SCN signals to different hypothalamic areas with the lateral hypothalamus, paraventricular nucleus and arcuate nucleus being the most prominent. With respect to the rewarding aspects of feeding behavior, the dopaminergic system is also under circadian influence. However the SCN projects only indirectly to the different reward regions, such as the ventral tegmental area where dopamine neurons are located. In addition, high palatable, high caloric diets have the potential to disturb the normal daily rhythms of physiology and have been shown to alter for example meal patterns. Around a meal several hormones and peptides are released that are also under circadian influence. For example, the release of postprandial insulin and glucagon-like peptide following a meal depend on the time of the day. Finally, we review the effect of deletion of different clock genes on feeding behavior. The most prominent effect on feeding behavior has been observed in Clock mutants, whereas deletion of Bmal1 and Per1/2 only disrupts the day-night rhythm, but not overall intake. Data presented here focus on the rodent literature as only limited data are available on the mechanisms underlying daily rhythms in human eating behavior.

2.
J Pineal Res ; 76(4): e12956, 2024 May.
Article En | MEDLINE | ID: mdl-38695262

The circadian timing system controls glucose metabolism in a time-of-day dependent manner. In mammals, the circadian timing system consists of the main central clock in the bilateral suprachiasmatic nucleus (SCN) of the anterior hypothalamus and subordinate clocks in peripheral tissues. The oscillations produced by these different clocks with a period of approximately 24-h are generated by the transcriptional-translational feedback loops of a set of core clock genes. Glucose homeostasis is one of the daily rhythms controlled by this circadian timing system. The central pacemaker in the SCN controls glucose homeostasis through its neural projections to hypothalamic hubs that are in control of feeding behavior and energy metabolism. Using hormones such as adrenal glucocorticoids and melatonin and the autonomic nervous system, the SCN modulates critical processes such as glucose production and insulin sensitivity. Peripheral clocks in tissues, such as the liver, muscle, and adipose tissue serve to enhance and sustain these SCN signals. In the optimal situation all these clocks are synchronized and aligned with behavior and the environmental light/dark cycle. A negative impact on glucose metabolism becomes apparent when the internal timing system becomes disturbed, also known as circadian desynchrony or circadian misalignment. Circadian desynchrony may occur at several levels, as the mistiming of light exposure or sleep will especially affect the central clock, whereas mistiming of food intake or physical activity will especially involve the peripheral clocks. In this review, we will summarize the literature investigating the impact of circadian desynchrony on glucose metabolism and how it may result in the development of insulin resistance. In addition, we will discuss potential strategies aimed at reinstating circadian synchrony to improve insulin sensitivity and contribute to the prevention of type 2 diabetes.


Circadian Rhythm , Glucose , Humans , Animals , Circadian Rhythm/physiology , Glucose/metabolism , Circadian Clocks/physiology , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/physiology
3.
Acta Neuropathol ; 147(1): 64, 2024 Mar 31.
Article En | MEDLINE | ID: mdl-38556574

Prader-Willi Syndrome (PWS) is a rare neurodevelopmental disorder of genetic etiology, characterized by paternal deletion of genes located at chromosome 15 in 70% of cases. Two distinct genetic subtypes of PWS deletions are characterized, where type I (PWS T1) carries four extra haploinsufficient genes compared to type II (PWS T2). PWS T1 individuals display more pronounced physiological and cognitive abnormalities than PWS T2, yet the exact neuropathological mechanisms behind these differences remain unclear. Our study employed postmortem hypothalamic tissues from PWS T1 and T2 individuals, conducting transcriptomic analyses and cell-specific protein profiling in white matter, neurons, and glial cells to unravel the cellular and molecular basis of phenotypic severity in PWS sub-genotypes. In PWS T1, key pathways for cell structure, integrity, and neuronal communication are notably diminished, while glymphatic system activity is heightened compared to PWS T2. The microglial defect in PWS T1 appears to stem from gene haploinsufficiency, as global and myeloid-specific Cyfip1 haploinsufficiency in murine models demonstrated. Our findings emphasize microglial phagolysosome dysfunction and altered neural communication as crucial contributors to the severity of PWS T1's phenotype.


Prader-Willi Syndrome , Humans , Mice , Animals , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/psychology , Microglia , Carrier Proteins/genetics , Phenotype , Phagosomes , Adaptor Proteins, Signal Transducing/genetics
4.
Metabolism ; 150: 155696, 2024 Jan.
Article En | MEDLINE | ID: mdl-37804881

BACKGROUND: Growing evidence demonstrates the role of the striatal dopamine system in the regulation of glucose metabolism. Treatment with dopamine antagonists is associated with insulin resistance and hyperglycemia, while dopamine agonists are used in treatment of type 2 diabetes. The mechanism underlying striatal dopamine effects in glucose metabolism, however is not fully understood. Here, we provide mechanistic insights into the role of nucleus accumbens shell (sNAc) dopaminergic signaling in systemic glucose metabolism. METHODS: Endogenous glucose production (EGP), blood glucose and mRNA expression in the lateral hypothalamic area (LHA) in male Wistar rats were measured following infusion of vanoxerine (VNX, dopamine reuptake inhibitor) in the sNAc. Thereafter, we analyzed projections from sNAc Drd1-expressing neurons to LHA using D1-Cre male Long-Evans rats, Cre-dependent viral tracers and fluorescence immunohistochemistry. Brain slice electrophysiology in adult mice was used to study spontaneous excitatory postsynaptic currents of sNAc Drd1-expressing neurons following VNX application. Finally, we assessed whether GABAergic LHA activity and hepatic vagal innervation were required for the effect of sNAc-VNX on glucose metabolism by combining infusion of sNAc-VNX with LHA-bicuculline, performing vagal recordings and combining infusion of sNAc-VNX with hepatic vagal denervation. RESULTS: VNX infusion in the sNAc strongly decreased endogenous glucose production, prevented glucose increases over time, reduced Slc17A6 and Hcrt mRNA in LHA, and increased vagal activity. Furthermore, sNAc Drd1-expressing neurons increased spontaneous firing following VNX application, and viral tracing of sNAc Drd1-expressing neurons revealed direct projections to LHA with on average 67 % of orexin cells directly targeted by sNAc Drd1-expressing neurons. Importantly, the sNAc-VNX-induced effect on glucose metabolism was dependent on GABAergic signaling in the LHA and on intact hepatic vagal innervation. CONCLUSIONS: We show that sNAc dopaminergic signaling modulates hepatic glucose metabolism through GABAergic inputs to glutamatergic LHA cells and hepatic vagal innervation. This demonstrates that striatal control of glucose metabolism involves a dopaminergic sNAc-LHA-liver axis and provides a potential explanation for the effects of dopamine agonists and antagonists on glucose metabolism.


Diabetes Mellitus, Type 2 , Hypothalamic Area, Lateral , Rats , Male , Mice , Animals , Hypothalamic Area, Lateral/metabolism , Nucleus Accumbens/metabolism , Dopamine/metabolism , Rodentia/metabolism , Dopamine Agonists/metabolism , Dopamine Agonists/pharmacology , Diabetes Mellitus, Type 2/metabolism , Rats, Wistar , Rats, Long-Evans , Glucose/metabolism , Liver/metabolism , RNA, Messenger/metabolism
5.
Cell Rep Med ; 4(12): 101299, 2023 12 19.
Article En | MEDLINE | ID: mdl-38016481

Lipid homeostasis in humans follows a diurnal pattern in muscle and pancreatic islets, altered upon metabolic dysregulation. We employ tandem and liquid-chromatography mass spectrometry to investigate daily regulation of lipid metabolism in subcutaneous white adipose tissue (SAT) and serum of type 2 diabetic (T2D) and non-diabetic (ND) human volunteers (n = 12). Around 8% of ≈440 lipid metabolites exhibit diurnal rhythmicity in serum and SAT from ND and T2D subjects. The spectrum of rhythmic lipids differs between ND and T2D individuals, with the most substantial changes observed early morning, as confirmed by lipidomics in an independent cohort of ND and T2D subjects (n = 32) conducted at a single morning time point. Strikingly, metabolites identified as daily rhythmic in both serum and SAT from T2D subjects exhibit phase differences. Our study reveals massive temporal and tissue-specific alterations of human lipid homeostasis in T2D, providing essential clues for the development of lipid biomarkers in a temporal manner.


Diabetes Mellitus, Type 2 , Lipid Metabolism , Humans , Lipid Metabolism/physiology , Subcutaneous Fat/metabolism , Adipose Tissue, White/metabolism , Lipids , Diabetes Mellitus, Type 2/metabolism
6.
Acta Neuropathol Commun ; 11(1): 107, 2023 07 03.
Article En | MEDLINE | ID: mdl-37400893

Evidence from animal experiments has shown that the hypothalamic paraventricular nucleus (PVN) plays a key role in regulating body weight and blood glucose levels. However, it is unclear whether neuron populations in the human PVN are involved in the development of type 2 diabetes mellitus (T2DM). To address this, we investigated the neuronal and glial populations in the PVN of 26 T2DM patients and 20 matched controls. Our findings revealed a significant reduction in oxytocin (Oxt) neuron density in the PVN of T2DM patients compared to controls, while other neuronal populations remained unchanged. This suggests that Oxt neurons may play a specific role in the pathophysiology of T2DM. Interestingly, the reduction in Oxt neurons was accompanied by a decreased melanocortinergic input in to the PVN as reflected by a reduction in alpha-MSH immunoreactivity. We also analysed two glial cell populations, as they are important for maintaining a healthy neural microenvironment. We found that microglial density, phagocytic capacity, and their proximity to neurons were not altered in T2DM patients, indicating that the loss of Oxt neurons is independent of changes in microglial immunity. However, we did observe a reduction in the number of astrocytes, which are crucial for providing trophic support to local neurons. Moreover, a specific subpopulation of astrocytes characterized by aquaporin 4 expression was overrepresented in T2DM patients. Since this subset of astrocytes is linked to the glymphatic system, their overrepresentation might point to alterations in the hypothalamic waste clearance system in T2DM. Our study shows selective loss of Oxt neurons in the PVN of T2DM individuals in association with astrocytic reduction and gliovascular remodelling. Therefore, hypothalamic Oxt neurons may represent a potential target for T2DM treatment modalities.


Diabetes Mellitus, Type 2 , Oxytocin , Humans , Body Weight , Diabetes Mellitus, Type 2/metabolism , Neurons/metabolism , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism
7.
Adv Biol (Weinh) ; 7(11): e2200324, 2023 11.
Article En | MEDLINE | ID: mdl-37017509

One possible pathological mechanism underlying hypertension and its related health consequences is dysfunction of the circadian system-a network of coupled circadian clocks that generates and orchestrates rhythms of ≈24 h in behavior and physiology. To better understand the role of circadian function during the development of hypertension, circadian regulation of motor activity is investigated in spontaneously hypertensive rats (SHRs) before the onset of hypertension and in their age-matched controls-Wistar Kyoto rats (WKYs). Two complementary properties in locomotor activity fluctuations are examined to assessthe multiscale regulatory function of the circadian control network: 1) rhythmicity at ≈24 h and 2) fractal patterns-similar temporal correlation at different time scales (≈0.5-8 h). Compared to WKYs, SHRs have more stable and less fragmented circadian activity rhythms but the changes in the rhythms (e.g., period and amplitude) from constant dark to light conditions are reduced or opposite. SHRs also have altered fractal activity patterns, displaying activity fluctuations with excessive regularity at small timescales that are linked to rigid physiological states. These different rhythmicity/fractal patterns and their different responses to light in SHRs indicate that an altered circadian function may be involved in the development of hypertension.


Hypertension , Prehypertension , Rats , Animals , Rats, Inbred SHR , Rats, Inbred WKY , Fractals , Motor Activity/physiology
8.
Adv Biol (Weinh) ; 7(11): e2200116, 2023 11.
Article En | MEDLINE | ID: mdl-35818679

Eating during the rest phase is associated with metabolic syndrome, proposed to result from a conflict between food consumption and the energy-saving state imposed by the circadian system. However, in nocturnal rodents, eating during the rest phase (day-feeding, DF) also implies food intake during light exposure. To investigate whether light exposure contributes to DF-induced metabolic impairments, animals receive food during the subjective day without light. A skeleton photoperiod (SP) is used to entrain rats to a 12:12 cycle with two short light pulses framing the subjective day. DF-induced adiposity is prevented by SP, suggesting that the conflict between light and feeding stimulates fat accumulation. However, all animals under SP conditions develop glucose intolerance regardless of their feeding schedule. Moreover, animals under SP with ad libitum or night-feeding have increased adiposity. SP animals show a delayed onset of the daily rise in body temperature and energy expenditure and shorter duration of nighttime activity, which may contribute to the metabolic disturbances. These data emphasize that metabolic homeostasis can only be achieved when all daily cycling variables are synchronized. Even small shifts in the alignment of different metabolic rhythms, such as those induced by SP, may predispose individuals to metabolic disease.


Glucose Intolerance , Photoperiod , Rats , Animals , Adiposity , Feeding Behavior , Circadian Rhythm , Glucose Intolerance/etiology , Obesity/etiology , Skeleton
9.
Front Physiol ; 13: 973461, 2022.
Article En | MEDLINE | ID: mdl-36105299

Nocturnal light pollution has been rapidly increasing during the last decades and even though dim artificial light at night (ALAN) has been associated with metabolic diseases, its mechanism is still far from clear. Therefore, the aim of our study was to thoroughly analyze the effects of ALAN on energy metabolism, metabolites, metabolic hormones, and gene expression. Male Wistar rats were kept in either the standard light:dark (12:12) cycle or exposed to ALAN (∼2 lx) during the whole 12-h dark phase for 2 weeks. Energy metabolism was measured in metabolic cages. In addition, we measured plasma and hepatic metabolites, clock and metabolic gene expression in the liver and epididymal adipose tissue, and plasma hormone levels. In ALAN rats, we observed an unexpected transitory daytime peak of locomotor activity and a suppression of the peak in locomotor activity at the beginning of the dark period. These changes were mirrored in the respiratory exchange ratio. Plasma metabolites became arrhythmic, and plasma and hepatic cholesterol levels were increased. Lost rhythmicity of metabolites was associated with disrupted behavioral rhythms and expression of metabolic genes. In the liver, the rhythms of metabolic sensors were either phase-advanced (Ppara, Pgc1a, Nampt) or arrhythmic (Sirt1, Lxra) after ALAN. The rhythmic pattern of Ppara and Sirt1 was abolished in the adipose tissue. In the liver, the amplitude of the daily rhythm in glycogen content was attenuated, the Glut2 rhythm was phase-advanced and Foxo1 lost its daily rhythmicity. Moreover, hepatic Foxo1 and Gck were up-regulated after ALAN. Interestingly, several parameters of lipid metabolism gained rhythmicity (adiponectin, Hmgcs2, Lpl, Srebf1c) in the liver, whereas Noct became arrhythmic in the adipose tissue. Peripheral clock genes maintained their robust oscillations with small shifts in their acrophases. Our data show that even a low level of ALAN can induce changes in the daily pattern of behavior and energy metabolism, and disturb daily rhythms of genes encoding key metabolic sensors and components of metabolic pathways in the liver and adipose tissue. Disturbed metabolic rhythms by ALAN could represent a serious risk factor for the development and progression of metabolic diseases.

10.
Neuroendocrinology ; 112(11): 1116-1128, 2022.
Article En | MEDLINE | ID: mdl-35316813

AIMS: Our study addresses underlying mechanisms of disruption of the circadian timing system by low-intensity artificial light at night (ALAN), which is a growing global problem, associated with serious health consequences. METHODS: Rats were exposed to low-intensity (∼2 lx) ALAN for 2 weeks. Using in situ hybridization, we assessed 24-h profiles of clock and clock-controlled genes in the suprachiasmatic nuclei (SCN) and other hypothalamic regions, which receive input from the master clock. Moreover, we measured the daily rhythms of hormones within the main neuroendocrine axes as well as the detailed daily pattern of feeding and drinking behavior in metabolic cages. RESULTS: ALAN strongly suppressed the molecular clockwork in the SCN, as indicated by the suppressed rhythmicity in the clock (Per1, Per2, and Nr1d1) and clock output (arginine vasopressin) genes. ALAN disturbed rhythmic Per1 expression in the paraventricular and dorsomedial hypothalamic nuclei, which convey the circadian signals from the master clock to endocrine and behavioral rhythms. Disruption of hormonal output pathways was manifested by the suppressed and phase-advanced corticosterone rhythm and lost daily variations in plasma melatonin, testosterone, and vasopressin. Importantly, ALAN altered the daily profile in food and water intake and eliminated the clock-controlled surge of drinking 2 h prior to the onset of the rest period, indicating disturbed circadian control of anticipatory thirst and fluid balance during sleep. CONCLUSION: Our findings highlight compromised time-keeping function of the central clock and multiple circadian outputs, through which ALAN disturbs the temporal organization of physiology and behavior.


Circadian Rhythm , Melatonin , Animals , Rats , Circadian Rhythm/genetics , Corticosterone/metabolism , Thirst , Light , Transcription Factors , Vasopressins , Arginine Vasopressin , Testosterone
11.
Int J Mol Sci ; 23(6)2022 Mar 08.
Article En | MEDLINE | ID: mdl-35328354

Obesity and type 2 diabetes mellitus (T2DM) are highly prevalent disorders, associated with insulin resistance and chronic inflammation. The brain is key for energy homeostasis and contains many insulin receptors. Microglia, the resident brain immune cells, are known to express insulin receptors (InsR) and to be activated by a hypercaloric environment. The aim of this study was to evaluate whether microglial insulin signaling is involved in the control of systemic energy homeostasis and whether this function is sex-dependent. We generated a microglia-specific knockout of the InsR gene in male and female mice and exposed them to control or obesogenic dietary conditions. Following 10 weeks of diet exposure, we evaluated insulin tolerance, energy metabolism, microglial morphology and phagocytic function, and neuronal populations. Lack of microglial InsR resulted in increased plasma insulin levels and insulin resistance in obese female mice. In the brain, loss of microglial InsR led to a decrease in microglial primary projections in both male and female mice, irrespective of the diet. In addition, in obese male mice lacking microglial InsR the number of proopiomelanocortin neurons was decreased, compared to control diet, while no differences were observed in female mice. Our results demonstrate a sex-dependent effect of microglial InsR-signaling in physiology and obesity, and stress the importance of a heterogeneous approach in the study of diseases such as obesity and T2DM.


Diabetes Mellitus, Type 2 , Insulin Resistance , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Female , Insulin/metabolism , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Microglia/metabolism , Obesity/genetics , Obesity/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
12.
FASEB J ; 36(2): e22133, 2022 02.
Article En | MEDLINE | ID: mdl-35032416

Shift-workers show an increased incidence of type 2 diabetes mellitus (T2DM). A possible mechanism is the disruption of the circadian timing of glucose homeostasis. Skeletal muscle mitochondrial function is modulated by the molecular clock. We used time-restricted feeding (TRF) during the inactive phase to investigate how mistimed feeding affects muscle mitochondrial metabolism. Rats on an ad libitum (AL) diet were compared to those that could eat only during the light (inactive) or dark (active) phase. Mitochondrial respiration, metabolic gene expressions, and metabolite concentrations were determined in the soleus muscle. Rats on AL feeding or dark-fed TRF showed a clear daily rhythm in muscle mitochondrial respiration. This rhythm in mitochondrial oxidative phosphorylation capacity was abolished in light-fed TRF animals and overall 24h respiration was lower. The expression of several genes involved in mitochondrial biogenesis and the fission/fusion machinery was altered in light-fed animals. Metabolomics analysis indicated that light-fed animals had lost rhythmic levels of α-ketoglutarate and citric acid. Contrastingly, lipidomics showed that light-fed animals abundantly gained rhythmicity in levels of triglycerides. Furthermore, while the RER shifted entirely with the food intake in the light-fed animals, many measured metabolic parameters (e.g., activity and mitochondrial respiration) did not strictly align with the shifted timing of food intake, resulting in a mismatch between expected metabolic supply/demand (as dictated by the circadian timing system and light/dark-cycle) and the actual metabolic supply/demand (as dictated by the timing of food intake). These data suggest that shift-work impairs mitochondrial metabolism and causes metabolic inflexibility, which can predispose to T2DM.


Cell Respiration/physiology , Circadian Clocks/physiology , Circadian Rhythm/physiology , Fasting/physiology , Mitochondria/physiology , Muscle, Skeletal/physiology , Animals , Diabetes Mellitus, Type 2/physiopathology , Diet/methods , Eating/physiology , Energy Metabolism/physiology , Feeding Behavior/physiology , Gene Expression/physiology , Male , Organelle Biogenesis , Oxidative Phosphorylation , Photoperiod , Rats , Rats, Wistar
13.
Neuropeptides ; 92: 102224, 2022 Apr.
Article En | MEDLINE | ID: mdl-34998113

In female mammals, reproductive senescence is a complex process involving progressive ovarian dysfunction, associated with altered central control of the hypothalamic-pituitary-gonadal axis and desynchronization of the circadian system. The objective of this study was to investigate age-dependent changes in the daily regulation of Arg-Phe amide-related peptide-3 (RFRP-3), a hypothalamic peptide involved in reproduction, in female C57BL/6 J mice of different age groups (4, 13, and 19 months old) sampled at their diestrus stage. We found an age-dependent decrease in the total number of RFRP-3 neurons and in the relative number of activated (i.e. c-Fos-positive) RFRP-3 neurons. RFRP-3 neuronal activation exhibited a daily variation in young and middle-aged mice, which was abolished in 19-month-old mice. We also found a daily variation in the number of RFRP-3 neurons receiving close vasopressin (AVP)- and vasoactive intestinal peptide (VIP)-ergic fiber appositions in mice aged 4 and 13 months, but not in 19-month-old mice. However, we found no daily or age-dependent changes in the AVP and VIP fiber density in the dorsomedial hypothalamus. Plasma LH levels were similar in mice aged 4 and 13 months, but were markedly increased in 19-month-old mice. The present findings indicate that the number of RFRP-3 positive neurons is downregulated during old age and that the daily changes in their innervation by the circadian peptides AVP and VIP are abolished. This age-associated reduced (rhythmic) activity of the inhibitory RFRP-3 system could be implicated in the elevated LH secretion observed during reproductive senescence.


Luteinizing Hormone , Neuropeptides , Animals , Female , Mammals , Mice , Mice, Inbred C57BL , Neurons , Neuropeptides/pharmacology , Vasoactive Intestinal Peptide
14.
J Neuroendocrinol ; 33(10): e13036, 2021 Aug 26.
Article En | MEDLINE | ID: mdl-34528311

Opioids are known to affect blood glucose levels but their exact role in the physiological control of glucose metabolism remains unclear. Although there are numerous studies investigating the peripheral effects of opioid stimulation, little is known about how central opioids control blood glucose and which brain areas are involved. One brain area possibly involved is the nucleus accumbens because, as well as being a key site for opioid effects on food intake, it has also been implicated in the control of blood glucose levels. Within the nucleus accumbens, µ-opioid receptors are most abundantly expressed. Therefore, in the present study, we investigated the role of µ-opioid receptors in the nucleus accumbens in the control of glucose metabolism. We show that infusion of the µ-opioid receptor agonist [d-Ala2 , N-MePhe4 , Gly-ol]-enkephalin (DAMGO) in the nucleus accumbens by itself does not affect blood glucose levels, but it enhances the glycaemic response after both an insulin tolerance test, as well as a glucose tolerance test. These findings indicate that the nucleus accumbens plays a role in the central effects of opioids on glucose metabolism, and highlight the possibility of nucleus accumbens µ-opioid receptors as a therapeutic target for enhancing the counter-regulatory response.

15.
Genes (Basel) ; 12(8)2021 07 31.
Article En | MEDLINE | ID: mdl-34440369

The circadian system interacts with the mesocorticolimbic reward system to modulate reward and memory in a time-of-day dependent manner. The circadian discrimination of reward, however, remains difficult to address between natural reinforcers and drugs of abuse. Circadian rhythms control cocaine sensitization and conversely cocaine causes long-term alteration in circadian periodicity in part through the serotonergic neurotransmission. Since neural circuits activated by cocaine and natural reinforcers do not completely overlap, we compared the effect of cocaine with that of sucrose, a strong reinforcer in rodents, by using passive chronic administration. The expression of fifteen genes playing a major role in DNA methylation (Dnmts, Tets), circadian rhythms (Clock, Bmal1, Per1/2, Cry1/2, Rev-Erbß, Dbp1), appetite, and satiety (Orexin, Npy) was analyzed in dopamine projection areas like the prefrontal cortex, the caudate putamen, and the hypothalamus interconnected with the reward system. The corresponding proteins of two genes (Orexin, Per2) were examined by IHC. For many factors controlling biological and cognitive functions, striking opposite responses were found between the two reinforcers, notably for genes controlling DNA methylation/demethylation processes and in global DNA methylation involved in chromatin remodeling. The data are consistent with a repression of critical core-clock genes by cocaine, suggesting that, consequently, both agents differentially modulate day/night cycles. Whether observed cocaine and sucrose-induced changes in DNA methylation in a time dependent manner are long lasting or contribute to the establishment of addiction requires further neuroepigenetic investigation. Understanding the mechanisms dissociating drugs of abuse from natural reinforcers remains a prerequisite for the design of selective therapeutic tools for compulsive behaviors.


Brain/drug effects , CLOCK Proteins/genetics , Chromatin Assembly and Disassembly , Cocaine/pharmacology , DNA Methylation , Sucrose/pharmacology , Animals , Body Weight , Brain/metabolism , Circadian Rhythm/genetics , Male , Rats , Rats, Wistar , Sucrose/administration & dosage
16.
Genes (Basel) ; 12(8)2021 08 19.
Article En | MEDLINE | ID: mdl-34440437

Based on studies describing an increased prevalence of addictive behaviours in several rare sleep disorders and shift workers, a relationship between circadian rhythms and addiction has been hinted for more than a decade. Although circadian rhythm alterations and molecular mechanisms associated with neuropsychiatric conditions are an area of active investigation, success is limited so far, and further investigations are required. Thus, even though compelling evidence connects the circadian clock to addictive behaviour and vice-versa, yet the functional mechanism behind this interaction remains largely unknown. At the molecular level, multiple mechanisms have been proposed to link the circadian timing system to addiction. The molecular mechanism of the circadian clock consists of a transcriptional/translational feedback system, with several regulatory loops, that are also intricately regulated at the epigenetic level. Interestingly, the epigenetic landscape shows profound changes in the addictive brain, with significant alterations in histone modification, DNA methylation, and small regulatory RNAs. The combination of these two observations raises the possibility that epigenetic regulation is a common plot linking the circadian clocks with addiction, though very little evidence has been reported to date. This review provides an elaborate overview of the circadian system and its involvement in addiction, and we hypothesise a possible connection at the epigenetic level that could further link them. Therefore, we think this review may further improve our understanding of the etiology or/and pathology of psychiatric disorders related to drug addiction.


Circadian Clocks/genetics , Circadian Rhythm/genetics , Epigenesis, Genetic/genetics , Substance-Related Disorders/genetics , DNA Methylation/genetics , Histones/genetics , Humans , Protein Processing, Post-Translational/genetics , Substance-Related Disorders/metabolism , Substance-Related Disorders/pathology
18.
Appetite ; 167: 105597, 2021 12 01.
Article En | MEDLINE | ID: mdl-34273421

Central dopamine signaling regulates reward-related aspects of feeding behavior, and during diet-induced obesity dopamine receptor signaling is altered. Yet, the influence of dopamine signaling on the consumption of specific dietary components remains to be elucidated. We have previously shown that 6-hydroxydopamine-mediated lesions of dopamine neuron terminals in the lateral shell of the nucleus accumbens promotes fat intake in rats fed a multi-component free-choice high-fat high-sugar (fcHFHS) diet. It is however not yet determined which dopamine receptors are responsible for this shift towards fat preference. In this study, we assess the effects of D1-or D2 receptor acute inhibition in the lateral shell of the nucleus accumbens on fcHFHS diet consumption. We report that infusion of the D1 receptor antagonist SCH2 3390, but not the D2 receptor antagonist raclopride, promotes dietary fat consumption in male Sprague Dawley rats on a fcHFHS diet during 2 h after infusion. Furthermore, anatomical analysis of infusion sites revealed that the rostral region, but not the caudal region, of the lateral shell of the nucleus accumbens is sensitive to the D1 receptor inhibition effects on fat consumption. Our data highlight a role for D1 receptors in the rostral region of the lateral shell of the nucleus accumbens to control dietary fat consumption.


Nucleus Accumbens , Receptors, Dopamine D1 , Animals , Dietary Fats , Male , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2
19.
Handb Clin Neurol ; 180: 45-63, 2021.
Article En | MEDLINE | ID: mdl-34225948

A major function of the nervous system is to maintain a relatively constant internal environment. The distinction between our external environment (i.e., the environment that we live in and that is subject to major changes, such as temperature, humidity, and food availability) and our internal environment (i.e., the environment formed by the fluids surrounding our bodily tissues and that has a very stable composition) was pointed out in 1878 by Claude Bernard (1814-1878). Later on, it was indicated by Walter Cannon (1871-1945) that the internal environment is not really constant, but rather shows limited variability. Cannon named the mechanism maintaining this limited variability homeostasis. Claude Bernard envisioned that, for optimal health, all physiologic processes in the body needed to maintain homeostasis and should be in perfect harmony with each other. This is illustrated by the fact that, for instance, during the sleep-wake cycle important elements of our physiology such as body temperature, circulating glucose, and cortisol levels show important variations but are in perfect synchrony with each other. These variations are driven by the biologic clock in interaction with hypothalamic target areas, among which is the paraventricular nucleus of the hypothalamus (PVN), a core brain structure that controls the neuroendocrine and autonomic nervous systems and thus is key for integrating central and peripheral information and implementing homeostasis. This chapter focuses on the anatomic connections between the biologic clock and the PVN to modulate homeostasis according to the daily sleep-wake rhythm. Experimental studies have revealed a highly specialized organization of the connections between the clock neurons and neuroendocrine system as well as preautonomic neurons in the PVN. These complex connections ensure a logical coordination between behavioral, endocrine, and metabolic functions that helps the organism maintain homeostasis throughout the day.


Hypothalamus , Paraventricular Hypothalamic Nucleus , Autonomic Nervous System , Humans , Neurons , Neurosecretory Systems
20.
Neurosci Lett ; 762: 136144, 2021 09 25.
Article En | MEDLINE | ID: mdl-34332031

Baroreflex sensitivity (BRS) is an important function of the nervous system and essential for maintaining blood pressure levels in the physiological range. In hypertension, BRS is decreased both in man and animals. Although increased sympathetic activity is thought to be the main cause of decreased BRS, hence the development of hypertension, the BRS is regulated by both sympathetic (SNS) and parasympathetic (PNS) nervous system. Here, we analyzed neuropeptide changes in the lateral hypothalamus (LH), which favours the SNS activity, as well as in PNS nuclei in the brainstem of spontaneously hypertensive rats (SHR) and their normotensive controls (Wistar Kyoto rats- WKY). The analyses revealed that in the WKY rats the hypothalamic orexin system, known for its role in sympathetic activation, showed a substantial decrease when animals age. At the same time, however, such a decrease was not observed when hypertension developed in the SHR. In contrast, Neuropeptide FF (NPFF) and Prolactin Releasing Peptide (PrRP) expression in the PNS associated Nucleus Tractus Solitarius (NTS) and Dorsal Motor Nucleus of the Vagus (DMV) diminished substantially, not only after the establishment of hypertension but also before its onset. Therefore, the current results indicate early changes in areas of the central nervous system involved in SNS and PNS control of blood pressure and associated with the development of hypertension.


Brain Stem/metabolism , Hypertension/physiopathology , Hypothalamus/metabolism , Neuropeptides/metabolism , Orexins/metabolism , Animals , Autonomic Nervous System/physiopathology , Baroreflex/physiology , Brain Stem/physiopathology , Hypothalamus/physiopathology , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY
...