Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis ; 96(4): 1593-1607, 2023.
Article in English | MEDLINE | ID: mdl-38007646

ABSTRACT

BACKGROUND: The need for early diagnosis biomarkers in Alzheimer's disease (AD) is growing. Only few studies have reported gustatory dysfunctions in AD using subjective taste tests. OBJECTIVE: The main purpose of the study was to explore gustatory functions using subjective taste tests and recordings of gustatory evoked potentials (GEPs) for sucrose solution in patients with minor or major cognitive impairment (CI) linked to AD, and to compare them with healthy controls. The secondary objective was to evaluate the relationships between GEPs and the results of cognitive assessments and fasting blood samples. METHODS: A total of 45 subjects (15 healthy subjects, 15 minor CI patients, 15 major CI patients) were included to compare their gustatory functions and brain activity by recording GEPs in response to a sucrose stimulation. CI groups were combined in second analyses in order to keep a high power in the study. Correlations were made with cognitive scores and hormone levels (ghrelin, leptin, insulin, serotonin). RESULTS: Increased P1 latencies and reduced N1 amplitudes were observed in minor or major patients compared to controls. GEPs were undetectable in 6 major and 4 minor CI patients. Thresholds for sucrose detection were significantly higher in the major CI group than in controls or the minor CI group. No correlation was found with hormone levels. CONCLUSIONS: The cortical processing of sensory taste information seems to be altered in patients with minor or major CI linked to AD. This disturbance was identifiable with subjective taste tests only later, at the major CI stage.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Taste Perception/physiology , Alzheimer Disease/complications , Evoked Potentials , Cognitive Dysfunction/complications , Sucrose , Hormones
2.
Article in English | MEDLINE | ID: mdl-36656675

ABSTRACT

Aims: Part of hypothalamic (mediobasal hypothalamus [MBH]) neurons detect changes in blood glucose levels that in turn coordinate the vagal control of insulin secretion. This control cascade requires the production of mitochondrial reactive oxygen species (mROS), which is altered in models of obesity and insulin resistance. Obese, insulin-resistant Zücker rats are characterized by hypothalamic hypersensitivity to glucose. This initiates an abnormal vagus-induced insulin secretion, associated with an overproduction of mROS in response to a low glucose dose. Here, we hypothesized that ghrelin, known to buffer reactive oxygen species (ROS) via mitochondrial function, may be a major component of the hypothalamic glucose hypersensitivity in the hypoghrelinemic obese Zücker rat. Results: Hypothalamic glucose hypersensitivity-induced insulin secretion of Zücker obese rats was reversed by ghrelin pretreatment. The overproduction of MBH mROS in response to a low glucose load no longer occurred in obese rats that had previously received the cerebral ghrelin infusion. This decrease in mROS production was accompanied by a normalization of oxidative phosphorylation (OXPHOS). Conversely, blocking the action of ghrelin with a growth hormone secretagogue receptor antagonist in a model of hyperghrelinemia (fasted rats) completely restored hypothalamic glucose sensing-induced insulin secretion that was almost absent in this physiological situation. Accordingly, ROS signaling and mitochondrial activity were increased by the ghrelin receptor antagonist. Innovation: These results demonstrate for the first time that ghrelin addressed only to the brain could have a protective effect on the defective control of insulin secretion in the insulin-resistant, hypoghrelinemic obese subject. Conclusions: Ghrelin, through its action on OXPHOS, modulates mROS signaling in response to cerebral hyperglycemia and the consequent vagal control of insulin secretion. In insulin-resistant obese states, brain hypoghrelinemia could be responsible for the nervous defect in insulin secretion.

3.
FEBS J ; 2022 Jul 26.
Article in English | MEDLINE | ID: mdl-35880408

ABSTRACT

Maintaining energy balance is essential for survival and health. This physiological function is controlled by the brain, which adapts food intake to energy needs. Indeed, the brain constantly receives a multitude of biological signals that are derived from digested foods or that originate from the gastrointestinal tract, energy stores (liver and adipose tissues) and other metabolically active organs (muscles). These signals, which include circulating nutrients, hormones and neuronal inputs from the periphery, collectively provide information on the overall energy status of the body. In the brain, several neuronal populations can specifically detect these signals. Nutrient-sensing neurons are found in discrete brain areas and are highly enriched in the hypothalamus. In turn, specialized brain circuits coordinate homeostatic responses acting mainly on appetite, peripheral metabolism, activity and arousal. Accumulating evidence shows that hypothalamic microglial cells located at the vicinity of these circuits can influence the brain control of energy balance. However, microglial cells could have opposite effects on energy balance, that is homeostatic or detrimental, and the conditions for this shift are not totally understood yet. One hypothesis relies on the extent of microglial activation, and nutritional lipids can considerably change it.

4.
Cell Rep ; 30(9): 3067-3078.e5, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32130907

ABSTRACT

Mechanistic studies in rodents evidenced synaptic remodeling in neuronal circuits that control food intake. However, the physiological relevance of this process is not well defined. Here, we show that the firing activity of anorexigenic POMC neurons located in the hypothalamus is increased after a standard meal. Postprandial hyperactivity of POMC neurons relies on synaptic plasticity that engages pre-synaptic mechanisms, which does not involve structural remodeling of synapses but retraction of glial coverage. These functional and morphological neuroglial changes are triggered by postprandial hyperglycemia. Chemogenetically induced glial retraction on POMC neurons is sufficient to increase POMC activity and modify meal patterns. These findings indicate that synaptic plasticity within the melanocortin system happens at the timescale of meals and likely contributes to short-term control of food intake. Interestingly, these effects are lost with a high-fat meal, suggesting that neuroglial plasticity of POMC neurons is involved in the satietogenic properties of foods.


Subject(s)
Hyperglycemia/physiopathology , Hypothalamus/metabolism , Meals , Neuroglia/pathology , Neuronal Plasticity , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Blood Glucose/metabolism , Electrophysiological Phenomena , Feeding Behavior , Hyperglycemia/blood , Mice, Inbred C57BL , Mice, Transgenic , Postprandial Period , Synapses/metabolism
5.
Diabetes Metab Syndr ; 13(4): 2489-2494, 2019.
Article in English | MEDLINE | ID: mdl-31405666

ABSTRACT

AIMS: GLP-1 analogues decrease food intake and have great promise for the fight against obesity. Little is known about their effects on food hedonic sensations and taste perception in poor controlled patients with type 2 diabetes (T2D). MATERIALS AND METHODS: Eighteen T2D patients with BMI ≥25 kg/m2 and poor controlled glycemia were studied before and after 3 months of treatment with Liraglutide. Detection thresholds for salty, sweet and bitter tastes, optimal preferences, olfactory liking, wanting and recalled liking for several food items were assessed. Subjects also answered questionnaires to measure their attitudes to food. RESULTS: T2D patients had a significant decrease in bodyweight and HbA1c after treatment with Liraglutide. Liraglutide improved gustative detection threshold of sweet flavors, and decreased wanting for sweet foods and recalled liking for fatty foods. It also led to a decrease in feelings of hunger. CONCLUSIONS: Liraglutide increases sensitivity to sweet tastes and decreases pleasure responses for fatty foods in poor controlled T2D patients, and is of particular interest in the understanding of the mechanisms of weight loss. CLINICAL TRIAL: NCT02674893.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Eating/psychology , Feeding Behavior/psychology , Food Preferences/psychology , Glucagon-Like Peptide 1/agonists , Liraglutide/therapeutic use , Mental Recall , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/analysis , Choice Behavior , Diabetes Mellitus, Type 2/psychology , Female , Follow-Up Studies , Humans , Hunger/physiology , Hypoglycemic Agents/therapeutic use , Male , Middle Aged , Obesity/prevention & control , Prognosis , Taste/physiology , Young Adult
6.
J Clin Invest ; 129(6): 2417-2430, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30938715

ABSTRACT

Glial cells have emerged as key players in the central control of energy balance and etiology of obesity. Astrocytes play a central role in neural communication via the release of gliotransmitters. Acyl-CoA binding protein (ACBP)-derived endozepines are secreted peptides that modulate the GABAA receptor. In the hypothalamus, ACBP is enriched in arcuate nucleus (ARC) astrocytes, ependymocytes and tanycytes. Central administration of the endozepine octadecaneuropeptide (ODN) reduces feeding and improves glucose tolerance, yet the contribution of endogenous ACBP in energy homeostasis is unknown. We demonstrated that ACBP deletion in GFAP+ astrocytes, but not in Nkx2.1-lineage neural cells, promoted diet-induced hyperphagia and obesity in both male and female mice, an effect prevented by viral rescue of ACBP in ARC astrocytes. ACBP-astrocytes were observed in apposition with proopiomelanocortin (POMC) neurons and ODN selectively activated POMC neurons through the ODN-GPCR but not GABAA, and supressed feeding while increasing carbohydrate utilization via the melanocortin system. Similarly, ACBP overexpression in ARC astrocytes reduced feeding and weight gain. Finally, the ODN-GPCR agonist decreased feeding and promoted weight loss in ob/ob mice. These findings uncover ACBP as an ARC gliopeptide playing a key role in energy balance control and exerting strong anorectic effects via the central melanocortin system.


Subject(s)
Astrocytes/metabolism , Diazepam Binding Inhibitor/metabolism , Eating , Energy Metabolism , Hyperphagia/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Astrocytes/pathology , Cell Line , Diazepam Binding Inhibitor/genetics , Female , Hyperphagia/genetics , Hyperphagia/pathology , Male , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Obesity/genetics , Obesity/pathology , Pro-Opiomelanocortin/genetics
7.
Mol Metab ; 20: 166-177, 2019 02.
Article in English | MEDLINE | ID: mdl-30553770

ABSTRACT

OBJECTIVE: Hypothalamic glucose sensing (HGS) initiates insulin secretion (IS) via a vagal control, participating in energy homeostasis. This requires mitochondrial reactive oxygen species (mROS) signaling, dependent on mitochondrial fission, as shown by invalidation of the hypothalamic DRP1 protein. Here, our objectives were to determine whether a model with a HGS defect induced by a short, high fat-high sucrose (HFHS) diet in rats affected the fission machinery and mROS signaling within the mediobasal hypothalamus (MBH). METHODS: Rats fed a HFHS diet for 3 weeks were compared with animals fed a normal chow. Both in vitro (calcium imaging) and in vivo (vagal nerve activity recordings) experiments to measure the electrical activity of isolated MBH gluco-sensitive neurons in response to increased glucose level were performed. In parallel, insulin secretion to a direct glucose stimulus in isolated islets vs. insulin secretion resulting from brain glucose stimulation was evaluated. Intra-carotid glucose load-induced hypothalamic DRP1 translocation to mitochondria and mROS (H2O2) production were assessed in both groups. Finally, compound C was intracerebroventricularly injected to block the proposed AMPK-inhibited DRP1 translocation in the MBH to reverse the phenotype of HFHS fed animals. RESULTS: Rats fed a HFHS diet displayed a decreased HGS-induced IS. Responses of MBH neurons to glucose exhibited an alteration of their electrical activity, whereas glucose-induced insulin secretion in isolated islets was not affected. These MBH defects correlated with a decreased ROS signaling and glucose-induced translocation of the fission protein DRP1, as the vagal activity was altered. AMPK-induced inhibition of DRP1 translocation increased in this model, but its reversal through the injection of the compound C, an AMPK inhibitor, failed to restore HGS-induced IS. CONCLUSIONS: A hypothalamic alteration of DRP1-induced fission and mROS signaling in response to glucose was observed in HGS-induced IS of rats exposed to a 3 week HFHS diet. Early hypothalamic modifications of the neuronal activity could participate in a primary defect of the control of IS and ultimately, the development of diabetes.


Subject(s)
Blood Glucose/metabolism , Dynamins/metabolism , Hypothalamus/metabolism , Mitochondria/metabolism , Sensory Receptor Cells/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Carotid Arteries/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Male , Protein Kinases/metabolism , Protein Transport , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Signal Transduction
8.
Front Nutr ; 5: 125, 2018.
Article in English | MEDLINE | ID: mdl-30619871

ABSTRACT

High variability exists in individual susceptibility to develop overweight in an obesogenic environment and the biological underpinnings of this heterogeneity are poorly understood. In this brief report, we show in mice that the vulnerability to diet-induced obesity is associated with low level of polysialic acid-neural cell adhesion molecule (PSA-NCAM), a factor of neural plasticity, in the hypothalamus. As we previously shown that reduction of hypothalamic PSA-NCAM is sufficient to alter energy homeostasis and promote fat storage under hypercaloric pressure, inter-individual variability in hypothalamic PSA-NCAM might account for the vulnerability to diet-induced obesity. These data support the concept that reduced plasticity in brain circuits that control appetite, metabolism and body weight confers risk for eating disorders and obesity.

9.
Diabetes ; 66(2): 314-324, 2017 02.
Article in English | MEDLINE | ID: mdl-27899482

ABSTRACT

The mediobasal hypothalamus (MBH) contains neurons capable of directly detecting metabolic signals such as glucose to control energy homeostasis. Among them, glucose-excited (GE) neurons increase their electrical activity when glucose rises. In view of previous work, we hypothesized that transient receptor potential canonical type 3 (TRPC3) channels are involved in hypothalamic glucose detection and the control of energy homeostasis. To investigate the role of TRPC3, we used constitutive and conditional TRPC3-deficient mouse models. Hypothalamic glucose detection was studied in vivo by measuring food intake and insulin secretion in response to increased brain glucose level. The role of TRPC3 in GE neuron response to glucose was studied by using in vitro calcium imaging on freshly dissociated MBH neurons. We found that whole-body and MBH TRPC3-deficient mice have increased body weight and food intake. The anorectic effect of intracerebroventricular glucose and the insulin secretory response to intracarotid glucose injection are blunted in TRPC3-deficient mice. TRPC3 loss of function or pharmacological inhibition blunts calcium responses to glucose in MBH neurons in vitro. Together, the results demonstrate that TRPC3 channels are required for the response to glucose of MBH GE neurons and the central effect of glucose on insulin secretion and food intake.


Subject(s)
Body Weight/genetics , Eating/genetics , Energy Metabolism/genetics , Glucose/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Neurons/metabolism , TRPC Cation Channels/genetics , Animals , Blotting, Western , Fasting , Glucose Tolerance Test , Homeostasis , Hypothalamus/cytology , Insulin Secretion , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , TRPC Cation Channels/metabolism
10.
Cell Metab ; 23(2): 324-34, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26621107

ABSTRACT

The composition of gut microbiota has been associated with host metabolic phenotypes, but it is not known if gut bacteria may influence host appetite. Here we show that regular nutrient provision stabilizes exponential growth of E. coli, with the stationary phase occurring 20 min after nutrient supply accompanied by bacterial proteome changes, suggesting involvement of bacterial proteins in host satiety. Indeed, intestinal infusions of E. coli stationary phase proteins increased plasma PYY and their intraperitoneal injections suppressed acutely food intake and activated c-Fos in hypothalamic POMC neurons, while their repeated administrations reduced meal size. ClpB, a bacterial protein mimetic of α-MSH, was upregulated in the E. coli stationary phase, was detected in plasma proportional to ClpB DNA in feces, and stimulated firing rate of hypothalamic POMC neurons. Thus, these data show that bacterial proteins produced after nutrient-induced E. coli growth may signal meal termination. Furthermore, continuous exposure to E. coli proteins may influence long-term meal pattern.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/growth & development , Gastrointestinal Tract/microbiology , Satiety Response , Adenosine Triphosphate/biosynthesis , Amygdala/metabolism , Animals , Electrophysiological Phenomena , Endopeptidase Clp , Escherichia coli/metabolism , Feeding Behavior , Female , Glucagon-Like Peptide 1/metabolism , Heat-Shock Proteins/metabolism , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Peptide YY/metabolism , Pro-Opiomelanocortin/metabolism , Proteomics , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Rats, Wistar
11.
Behav Brain Res ; 268: 40-7, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24675157

ABSTRACT

Learned association between odor, taste and further post-ingestive consequence is known as flavor nutrient conditioned preference. Amygdala is supposed to be one of the areas involved in these associations. In the present study, one flavor was associated with a 16% glucose (CS(+)) whereas another flavor was paired with less reinforcing 4% glucose (CS(-)). We showed that CS(+) presentation after conditioning increased Fos expression in the basolateral nucleus of amygdala (BLA). Furthermore, we performed electrophysiological recordings in the BLA in free moving rats. After preference acquisition, rats were exposed to either the CS(+) or the CS(-). The proportion of neurons showing a decreased activity during the CS(-) presentation was significantly higher in conditioned rats compared to controls. Among this neuronal population recorded in conditioned rats, we noticed a significant proportion of neurons that also showed a decreased activity during the CS(+) presentation. Our data indicate an involvement of BLA during retrieval of learned flavors. It also suggests that both flavors might have acquired a biological value through conditioning.


Subject(s)
Basolateral Nuclear Complex/physiology , Conditioning, Psychological/physiology , Food Preferences/physiology , Neurons/physiology , Taste Perception/physiology , Action Potentials , Animals , Eating , Electrodes, Implanted , Food Deprivation , Glucose/administration & dosage , Immunohistochemistry , Male , Oncogene Proteins v-fos/metabolism , Rats, Wistar , Signal Processing, Computer-Assisted
12.
Behav Brain Res ; 184(1): 1-10, 2007 Nov 22.
Article in English | MEDLINE | ID: mdl-17686536

ABSTRACT

The long-chain polyunsaturated n-3 fatty acids (n-3 PUFA), particularly docosahexaenoic acid (DHA), are abundantly present in the central nervous system and play an important role in cognitive functions such as learning and memory. We, therefore, investigated the effects of n-3 PUFA-depletion in rats (F2 generation) on the learning of an olfactory discrimination task, progressively acquired within a four-arm maze, and on the mRNA expression of some candidate genes, i.e., c-fos, Gir and glucose transporter (Glut1), which could reflect the level of cerebral activity. We observed that DHA contents were dramatically decreased in the olfactory bulb, the piriform cortex and the neocortex of n-3-depleted rats. Furthermore, the n-3 deficiency resulted in a mild olfactory learning impairment as these rats required more days to master the olfactory task compared to control rats. Real-time RT-PCR experiments revealed that the training induced the expression of c-fos mRNA in all the three regions of the brain whereas Gir and Glut1 mRNA were induced only in olfactory bulb and neocortex. However, such an increase was less marked in the n-3-deficient rats. Taken together, these results allow us to assume that the behavioural impairment in n-3-deficient rats is linked to the depletion of n-3 fatty acids in brain regions processing olfactory cues. Data are discussed in view of the possible role of some of these genes in learning-induced neuronal olfactory plasticity.


Subject(s)
Brain/metabolism , Discrimination, Psychological/physiology , Fatty Acids, Omega-3/metabolism , Glucose Transporter Type 1/genetics , Proto-Oncogene Proteins c-fos/genetics , Receptors, G-Protein-Coupled/genetics , Smell/physiology , Analysis of Variance , Animals , Behavior, Animal/physiology , Body Weight/physiology , Diet, Fat-Restricted/methods , Discrimination Learning/physiology , Gene Expression Regulation/physiology , Glucose Transporter Type 1/metabolism , Male , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors
13.
Learn Mem ; 12(3): 307-17, 2005.
Article in English | MEDLINE | ID: mdl-15897253

ABSTRACT

Fos protein immunodetection was used to investigate the neuronal activation elicited in some olfactory-related areas after either learning of an olfactory discrimination task or its reactivation 10 d later. Trained rats (T) progressively acquired the association between one odor of a pair and water-reward in a four-arm maze. Two groups of pseudotrained rats were used: PO rats were not water restricted and were submitted to the olfactory stimuli in the maze without any reinforcement, whereas PW rats were water-deprived and systematically received water in the maze without any odorous stimulation. When the discrimination task was well mastered, a significantly lower Fos immunoreactivity was observed in T rats compared to PW and PO rats in most of the analyzed brain areas, which could reflect the post-acquisition consolidation process. Following memory reactivation, differences in Fos immunoreactivity between trained and some pseudotrained rats were found in the anterior part of piriform cortex, CA3, and orbitofrontal cortex. We also observed that Fos labeling was significantly higher in trained rats after memory reactivation than after acquisition of the olfactory task in most of the brain areas examined. Our results support the assumption of a differential involvement of neuronal networks after either learning or reactivation of an olfactory discrimination task.


Subject(s)
Discrimination Learning/physiology , Discrimination, Psychological/physiology , Learning/physiology , Olfactory Pathways/metabolism , Oncogene Proteins v-fos/biosynthesis , Smell/physiology , Animals , Functional Laterality/physiology , Habenula/physiology , Immunohistochemistry , Limbic System/physiology , Psychomotor Performance/physiology , Rats , Rats, Wistar
14.
Behav Brain Res ; 157(1): 127-37, 2005 Feb 10.
Article in English | MEDLINE | ID: mdl-15617779

ABSTRACT

By using Fos immunocytochemistry, we investigated the activation in olfactory-related areas at three stages (the first and fourth days of conditioning and complete acquisition) of an olfactory discrimination learning task. The trained rats (T) had to associate one odour of a pair with water-reward within a four-arm maze whereas pseudo-trained (P) rats were only submitted to the olfactory cues without any reinforcement. In the piriform cortex, both T and P rats exhibited a higher immunoreactivity on the first day, which seemed to indicate a novelty-related Fos expression in this area, but whatever the learning-stage, no significant difference in Fos expression between T and P rats was observed. In hippocampus, Fos expression was significantly different between T and P rats in CA1 and CA3 on the first and fourth days respectively. Thus we showed a differential activation of CA1 and CA3 subfields which might support a possible functional heterogeneity. In the orbitofrontal cortex, Fos immunoreactivity was significantly higher in T rats compared to P rats when mastery of the discrimination task was complete. In contrast, no learning-related Fos expression was found in infralimbic and prelimbic cortices. The present data suggest an early implication of the hippocampal formation and a later involvement of neocortical areas throughout different stages of a progressively acquired olfactory learning task.


Subject(s)
Brain Mapping , Cerebral Cortex/metabolism , Discrimination Learning/physiology , Nerve Net/metabolism , Oncogene Proteins v-fos/metabolism , Analysis of Variance , Animals , Hippocampus/metabolism , Immunochemistry , Limbic System/metabolism , Male , Memory/physiology , Parahippocampal Gyrus/metabolism , Rats , Rats, Wistar , Smell/physiology
15.
Brain Res Bull ; 64(1): 31-8, 2004 Jul 30.
Article in English | MEDLINE | ID: mdl-15275954

ABSTRACT

The piriform cortex (PCx) and related structures such as hippocampus and frontal cortex could play an important role in olfactory memory. We investigated their involvement in learning the biological value of an odor cue, i.e. predicting reward or non-reward in a two-odor discrimination task. Rats were sacrificed after stimulation by either rewarded or non-rewarded odor and Fos immunocytochemistry was performed. The different experimental groups of rats did not show strongly differentiated Fos expression pattern in either the PCx or the hippocampus. A few differences were noted in frontal areas. In the ventro-lateral orbital cortex, rats, ramdomly rewarded during the conditionning had a higher Fos level in comparison with other groups. In infralimbic cortex, rats, which learned the reward value of the olfactory cue and were water-reinforced the day of sacrifice, showed a higher Fos expression. Data are discussed in view of the olfactory learning paradigm and of the accuracy of the control groups used in the present experimental design. The behavioural conditions leading to Fos expression are further discussed since Fos is a marker of learning-induced plasticity as well as a general activity marker which can be activated by a wide range of stimuli not directly linked to memory.


Subject(s)
Conditioning, Operant/physiology , Cues , Discrimination Learning/physiology , Olfactory Pathways/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Animals , Behavior, Animal/physiology , Cell Count , Cerebral Cortex/metabolism , Gene Expression Regulation/physiology , Hippocampus/metabolism , Immunohistochemistry/methods , Male , Maze Learning/physiology , Neurons/metabolism , Olfactory Pathways/cytology , Rats , Rats, Wistar , Smell/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...