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










Publication year range
1.
Depress Anxiety ; 39(5): 407-418, 2022 05.
Article in English | MEDLINE | ID: mdl-35357051

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is characterized by a high rate of treatment resistance. Omega (ω)-3 polyunsaturated fatty acids (PUFAs) were shown to correlate with depressive phenotype both in rodents and in humans. However, few studies to date have investigated the role of PUFAs in antidepressant response. The primary aim of this study was to assess the link between baseline PUFA composition and changes in depressive symptoms as well as antidepressant response in a multicenter study of depressed patients. METHODS: Sixty depressed adults who met criteria for MDD according to DSM-IV-TR were recruited. Neuropsychiatric evaluations occurred at baseline and after 4 and 8 weeks of treatment with standard antidepressants, including escitalopram (N = 45), sertraline (N = 13) and venlafaxine (N = 2). At study endpoint, patients were stratified into responders (R) or non-responders (NR) based on their MADRS (Montgomery-Åsberg Depression Rating Scale) score. Baseline PUFA levels were assessed and their association with clinical response was determined. RESULTS: Lower ω-3 PUFA levels were associated to worse baseline symptomatology. Baseline levels of PUFAs were significantly different between R and NR, with R exhibiting lower docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA) and ω-3 index; and higher ω-6/ω-3 ratio than NR before the start of antidepressant treatment. DHA levels as well as the ω-3 index and ω-6/ω-3 ratio significantly predicted response to antidepressants at study endpoint. CONCLUSIONS: These results show that baseline levels of PUFAs predict later response to standard antidepressants in depressed subjects. They suggest that PUFA intake and/or metabolism represent a novel modifiable tool for the management of unresponsive depressed patients.


Subject(s)
Depressive Disorder, Major , Fatty Acids, Omega-3 , Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Docosahexaenoic Acids/therapeutic use , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Unsaturated/therapeutic use , Humans
2.
J Neuroinflammation ; 18(1): 290, 2021 Dec 11.
Article in English | MEDLINE | ID: mdl-34895261

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) represents a major public health concern, particularly due to its steadily rising prevalence and the poor responsiveness to standard antidepressants notably in patients afflicted with chronic inflammatory conditions, such as obesity. This highlights the need to improve current therapeutic strategies, including by targeting inflammation based on its role in the pathophysiology and treatment responsiveness of MDD. Nevertheless, dissecting the relative contribution of inflammation in the development and treatment of MDD remains a major issue, further complicated by the lack of preclinical depression models suitable to experimentally dissociate inflammation-related vs. inflammation-unrelated depression. METHODS: While current models usually focus on one particular MDD risk factor, we compared in male C57BL/6J mice the behavioral, inflammatory and neurobiological impact of chronic exposure to high-fat diet (HFD), a procedure known to induce inflammation-related depressive-like behaviors, and unpredictable chronic mild stress (UCMS), a stress-induced depression model notably renowned for its responsivity to antidepressants. RESULTS: While both paradigms induced neurovegetative, depressive-like and anxiety-like behaviors, inflammation and downstream neurobiological pathways contributing to inflammation-driven depression were specifically activated in HFD mice, as revealed by increased circulating levels of inflammatory factors, as well as brain expression of microglial activation markers and enzymes from the kynurenine and tetrahydrobiopterin (BH4) pathways. In addition, serotoninergic and dopaminergic systems were differentially impacted, depending on the experimental condition. CONCLUSIONS: These data validate an experimental design suitable to deeply study the mechanisms underlying inflammation-driven depression comparatively to non-inflammatory depression. This design could help to better understand the pathophysiology of treatment resistant depression.


Subject(s)
Brain/metabolism , Depressive Disorder, Major/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Inflammation Mediators/metabolism , Animals , Depressive Disorder, Major/etiology , Depressive Disorder, Major/psychology , Male , Mice , Mice, Inbred C57BL
3.
Neurobiol Stress ; 15: 100353, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34189193

ABSTRACT

Stress is a fundamental biological response that can be associated with alterations in cognitive processes. Unhealthy dietary habits are proposed to modulate this effect, notably through their pro-inflammatory potential. This cross-sectional study aimed to evaluate the influence of an obesogenic dietary pattern with inflammatory potential on stress-induced cognitive alterations in healthy volunteers. Fifty healthy adult participants were stratified into two diet groups: obesogenic vs. non-obesogenic, based on their self-reported consumption of fat, sugar, and salt, assessed by the French National Program for Nutrition and Health questionnaire and a food frequency questionnaire. Serum high-sensitive C-reactive protein (hsCRP) was measured as a marker of systemic inflammation using ELISA. Verbal memory and sustained attention were evaluated through the Verbal Recognition Memory (VRM) test and the Rapid Visual Information Processing (RVP) test respectively, from the Cambridge Neuropsychological Test Automated Battery. Assessments were performed before and after exposure to the psychological stressor Trier Social Stress Test (TSST). Stress response was evaluated by subjective stress perception, salivary cortisol, blood pressure, and heart rate. Twenty-two participants (44%) presented an obesogenic diet. Systemic inflammation was significantly higher in the obesogenic diet group (p=0.005). The TSST induced a significant stress response, regardless of dietary habits (Time effect p < 0.001). In the whole sample, exposure to TSST was associated with cognitive changes in the form of impaired performance on the VRM test and overall improved RVP scores. However, the obesogenic diet group exhibited an increased total number of false alarms (Time x Diet: p=0.014) on the RVP test after TSST exposure as well as a greater impairment in immediate verbal recognition on the VRM test (Time x Diet: p=0.002). This effect was not associated with the inflammatory component of the obesogenic diet. These results suggest that an obesogenic diet may sensitize healthy individuals to the detrimental effects of acute stress on cognitive performance.

4.
Brain Behav Immun ; 94: 104-110, 2021 05.
Article in English | MEDLINE | ID: mdl-33684502

ABSTRACT

OBJECTIVE: Neuropsychiatric symptoms are frequent in obese individuals. Mounting evidence suggests that adiposity-related inflammation contributes to this effect. This study assessed the relationship between adiposity, neuropsychiatric symptom dimensions and systemic inflammation in subjects stratified by body-mass-index (BMI). METHODS: The study included 165 subjects, of whom 70 were very severely obese (BMI ≥ 40 kg/m2), 50 severely obese (BMI: 35-39.99 kg/m2), 21 overweight or moderately obese (BMI: 25-34.9 kg/m2), and 24 lean (BMI < 25 kg/m2). Depressive symptoms were assessed using the Montgomery-Asberg Depression Rating Scale (MADRS) and the Mini-International Neuropsychiatric Interview (MINI). Fatigue and general neurobehavioral symptoms were assessed using the Multidimensional Fatigue Inventory (MFI) and Neurotoxicity Rating Scale (NRS) respectively. Serum levels of the inflammatory markers, high-sensitive (hs) CRP and hsIL-6, were determined by ELISA. RESULTS: Severely obese subjects exhibited higher MADRS, MFI and NRS scores and were more frequently afflicted with current diagnosis of major depression than lean participants. Scores on psychometric scales were also increased in very severely obese subjects, although to a lesser extent. Alterations in neuropsychiatric dimensions were highly inter-related. HsCRP was significantly increased in subjects with severe or very severe obesity, while hsIL-6 was augmented in all obese groups. Overall, increased neuropsychiatric comorbidity was associated with greater systemic inflammation, notably hsCRP. CONCLUSION: Obesity is characterized by an increased prevalence of inter-related neuropsychiatric symptoms together with low-grade systemic inflammation augmenting with adiposity. The association between adiposity, systemic inflammation and neuropsychiatric alterations supports the contribution of adiposity-related inflammatory processes to neuropsychiatric comorbidities in obesity. These data suggest that consideration of adiposity characteristics may help identifying subjects at increased risk for neuropsychiatric comorbidity.


Subject(s)
C-Reactive Protein , Obesity , Adiposity , Body Mass Index , C-Reactive Protein/metabolism , Humans , Inflammation/complications , Obesity/complications
5.
Brain Behav Immun ; 84: 23-35, 2020 02.
Article in English | MEDLINE | ID: mdl-31731013

ABSTRACT

Metabolic syndrome represents a major risk factor for severe comorbidities such as cardiovascular diseases or diabetes. It is also associated with an increased prevalence of emotional and cognitive alterations that in turn aggravate the disease and related outcomes. Identifying therapeutic strategies able to improve those alterations is therefore a major socioeconomical and public health challenge. We previously reported that both hippocampal inflammatory processes and neuronal plasticity contribute to the development of emotional and cognitive alterations in db/db mice, an experimental model of metabolic syndrome that displays most of the classical features of the syndrome. In that context, nutritional interventions with known impact on those neurobiological processes appear as a promising alternative to limit the development of neurobiological comorbidities of metabolic syndrome. We therefore tested here whether n-3 polyunsaturated fatty acids (n-3 PUFAs) associated with a cocktail of antioxidants can protect against the development of behavioral alterations that accompany the metabolic syndrome. Thus, this study aimed: 1) to evaluate if a diet supplemented with the plant-derived n-3 PUFA α-linolenic acid (ALA) and antioxidants (provided by n-3 PUFAs-rich rapeseed oil fortified with a mix of naturally constituting antioxidant micronutrients, including coenzyme Q10, tocopherol, and the phenolic compound canolol) improved behavioral alterations in db/db mice, and 2) to decipher the biological mechanisms underlying this behavioral effect. Although the supplemented diet did not improve anxiety-like behavior and inflammatory abnormalities, it reversed hippocampus-dependent spatial memory deficits displayed by db/db mice in a water maze task. It concomitantly changed subunit composition of glutamatergic AMPA and NMDA receptors in the hippocampus that has been shown to modulate synaptic function related to spatial memory. These data suggest that changes in local neuronal plasticity may underlie cognitive improvements in db/db mice fed the supplemented diet. The current findings might therefore provide valuable data for introducing new nutritional strategies for the treatment of behavioral complications associated with MetS.


Subject(s)
Cognition Disorders/diet therapy , Cognition/drug effects , Food, Fortified , Metabolic Syndrome/diet therapy , Micronutrients/pharmacology , Rapeseed Oil/chemistry , Rapeseed Oil/pharmacology , Animals , Cognition Disorders/complications , Cognition Disorders/physiopathology , Disease Models, Animal , Male , Metabolic Syndrome/complications , Metabolic Syndrome/physiopathology , Mice
6.
Brain Behav Immun ; 77: 25-36, 2019 03.
Article in English | MEDLINE | ID: mdl-30508579

ABSTRACT

Although the high prevalence of anxiety in obesity increasingly emerges as significant risk factor for related severe health complications, the underlying pathophysiological mechanisms remain poorly understood. Considering that chronic inflammation is a key component of obesity and is well known to impact brain function and emotional behavior, we hypothesized that it may similarly contribute to the development of obesity-related anxiety. This hypothesis was experimentally tested by measuring whether chronic food restriction, a procedure known to reduce inflammation, or chronic anti-inflammatory treatment with ibuprofen improved anxiety-like behavior and concomitantly decreased peripheral and/or hippocampal inflammation characterizing a model of severe obesity, the db/db mice. In both experiments, reduced anxiety-like behaviors in the open-field and/or elevated plus-maze were selectively associated with decreased hippocampal tumor necrosis factor-α (TNF-α) mRNA expression. Highlighting the causality of both events, chronic central infusion of the TNF-α blocker etanercept was then shown to be sufficient to improve anxiety-like behavior in db/db mice. Lastly, by measuring the impact of ex-vivo etanercept on hippocampal synaptic processes underlying anxiety-like behaviors, we showed that the anxiolytic effect of central TNF-α blockade likely involved modulation of synaptic transmission within the ventral hippocampus. Altogether, these results uphold the role of brain TNF-α in mediating obesity-related anxiety and provide important clues about how it may modulate brain function and behavior. They may therefore help to introduce novel therapeutic strategies to reduce anxiety associated with inflammatory conditions.


Subject(s)
Anxiety/metabolism , Obesity/psychology , Tumor Necrosis Factor-alpha/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Anxiety Disorders/metabolism , Behavior, Animal/physiology , Brain/metabolism , Disease Models, Animal , Etanercept/pharmacology , Hippocampus/metabolism , Inflammation/metabolism , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Tumor Necrosis Factor-alpha/physiology
7.
Psychoneuroendocrinology ; 91: 55-61, 2018 05.
Article in English | MEDLINE | ID: mdl-29525586

ABSTRACT

BACKGROUND: Recent reports suggest that the risk of depressive symptoms in obesity is potentiated in subjects presenting a metabolically unhealthy phenotype. Inflammation is often considered a defining criteria of metabolic health. However, this factor may drive the association of metabolic health with depressive symptoms given its well-known role in the pathophysiology of depression. This study aimed at determining the relative contribution of inflammation and metabolic abnormalities to depressive symptoms in obesity. METHODS: One-hundred severely obese adults (BMI ≥ 35-40 kg/m2) and 25 non-obese control individuals (BMI < 30 kg/m2) were recruited. Depressive symptoms were assessed using the Montgomery-Asberg Depression Rating Scale (MADRS) and Mini-International Neuropsychiatric Interview (MINI). Serum high-sensitive C-reactive protein (hs-CRP) was measured as a marker of systemic inflammation. Metabolically unhealthy obesity was defined as obesity associated with two or more metabolic alterations, including low high-density lipoprotein cholesterol, hypertriglyceridemia, high fasting glucose and hypertension. RESULTS: Total MADRS scores were significantly higher in obese subjects with significant inflammation (hs-CRP ≥ 5 mg/L) compared to those with low inflammation (hs-CRP < 5 mg/L) and non-obese controls. Interestingly, hs-CRP levels significantly predicted MADRS scores in the whole population under study and in the group of obese subjects. Overall, no association was found between MADRS scores and individual metabolic alterations or the composite measure of metabolically unhealthy obesity. Similarly, the association of hs-CRP with MADRS scores in obese patients was not modulated by metabolic health factors. CONCLUSIONS: These results indicate that systemic inflammation represents a stronger contributor of obesity-related depressive symptoms than metabolic health per se. This supports the notion that inclusion of inflammation in the definition of metabolically unhealthy obesity drives the association found between poor metabolic health and depressive symptoms.


Subject(s)
Depression/physiopathology , Obesity/metabolism , Obesity/physiopathology , Adult , Blood Glucose/metabolism , Body Mass Index , C-Reactive Protein/metabolism , Depression/metabolism , Fasting/blood , Female , Humans , Inflammation/complications , Insulin Resistance/physiology , Male , Middle Aged , Risk Factors
8.
Brain Behav Immun ; 64: 33-49, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28027925

ABSTRACT

Mounting evidence shows that the gut microbiota, an important player within the gut-brain communication axis, can affect metabolism, inflammation, brain function and behavior. Interestingly, gut microbiota composition is known to be altered in patients with metabolic syndrome (MetS), who also often display neuropsychiatric symptoms. The use of prebiotics, which beneficially alters the microbiota, may therefore be a promising way to potentially improve physical and mental health in MetS patients. This hypothesis was tested in a mouse model of MetS, namely the obese and type-2 diabetic db/db mice, which display emotional and cognitive alterations associated with changes in gut microbiota composition and hippocampal inflammation compared to their lean db/+ littermates. We assessed the impact of chronic administration (8weeks) of prebiotics (oligofructose) on both metabolic (body weight, food intake, glucose homeostasis) and behavioral (increased anxiety-like behavior and impaired spatial memory) alterations characterizing db/db mice, as well as related neurobiological correlates, with particular attention to neuroinflammatory processes. Prebiotic administration improved excessive food intake and glycemic dysregulations (glucose tolerance and insulin resistance) in db/db mice. This was accompanied by an increase of plasma anti-inflammatory cytokine IL-10 levels and hypothalamic mRNA expression of the anorexigenic cytokine IL-1ß, whereas unbalanced mRNA expression of hypothalamic orexigenic (NPY) and anorexigenic (CART, POMC) peptides was unchanged. We also detected signs of improved blood-brain-barrier integrity in the hypothalamus of oligofructose-treated db/db mice (normalized expression of tight junction proteins ZO-1 and occludin). On the contrary, prebiotic administration did not improve behavioral alterations and associated reduction of hippocampal neurogenesis displayed by db/db mice, despite normalization of increased hippocampal IL-6 mRNA expression. Of note, we found a relationship between the effect of treatment on dentate gyrus neurons and spatial memory. These findings may prove valuable for introducing novel approaches to treat some of the comorbidities associated with MetS.


Subject(s)
Behavior, Animal , Gastrointestinal Microbiome , Metabolic Syndrome/metabolism , Metabolic Syndrome/microbiology , Prebiotics/administration & dosage , Animals , Bifidobacterium , Blood-Brain Barrier/metabolism , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/microbiology , Hippocampus/metabolism , Hypothalamus/metabolism , Inflammation/metabolism , Inflammation/microbiology , Male , Mice, Inbred C57BL , Mice, Obese , Spatial Memory
9.
J Neurosci ; 35(9): 4092-103, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25740536

ABSTRACT

In addition to metabolic and cardiovascular disorders, obesity is associated with adverse cognitive and emotional outcomes. Its growing prevalence during adolescence is particularly alarming since recent evidence indicates that obesity can affect hippocampal function during this developmental period. Adolescence is a decisive period for maturation of the amygdala and the hypothalamic-pituitary-adrenal (HPA) stress axis, both required for lifelong cognitive and emotional processing. However, little data are available on the impact of obesity during adolescence on amygdala function. Herein, we therefore evaluate in rats whether juvenile high-fat diet (HFD)-induced obesity alters amygdala-dependent emotional memory and whether it depends on HPA axis deregulation. Exposure to HFD from weaning to adulthood, i.e., covering adolescence, enhances long-term emotional memories as assessed by odor-malaise and tone-shock associations. Juvenile HFD also enhances emotion-induced neuronal activation of the basolateral complex of the amygdala (BLA), which correlates with protracted plasma corticosterone release. HFD exposure restricted to adulthood does not modify all these parameters, indicating adolescence is a vulnerable period to the effects of HFD-induced obesity. Finally, exaggerated emotional memory and BLA synaptic plasticity after juvenile HFD are alleviated by a glucocorticoid receptor antagonist. Altogether, our results demonstrate that juvenile HFD alters HPA axis reactivity leading to an enhancement of amygdala-dependent synaptic and memory processes. Adolescence represents a period of increased susceptibility to the effects of diet-induced obesity on amygdala function.


Subject(s)
Amygdala/physiopathology , Emotions , Glucocorticoids/metabolism , Memory , Neuronal Plasticity , Obesity/psychology , Animals , Anxiety/psychology , Avoidance Learning , Fear/psychology , Male , Obesity/physiopathology , Rats , Rats, Wistar , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
10.
Brain Behav Immun ; 40: 9-17, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24662056

ABSTRACT

In addition to metabolic and cardiovascular disorders, obesity pandemic is associated with chronic low-grade inflammation as well as adverse cognitive outcomes. However, the existence of critical periods of development that differ in terms of sensitivity to the effects of diet-induced obesity remains unexplored. Using short exposure to a high-fat diet (HFD) exerting no effects when given to adult mice, we recently found impairment of hippocampal-dependent memory and plasticity after similar HFD exposure encompassing adolescence (from weaning to adulthood) showing the vulnerability of the juvenile period (Boitard et al., 2012). Given that inflammatory processes modulate hippocampal functions, we evaluated in rats whether the detrimental effect of juvenile HFD (jHFD) on hippocampal-dependent memory is associated with over-expression of hippocampal pro-inflammatory cytokines. jHFD exposure impaired long-term spatial reference memory in the Morris water maze without affecting acquisition or short-term memory. This suggests an effect on consolidation processes. Moreover, jHFD consumption delayed spatial reversal learning. jHFD intake did neither affect basal expression of pro-inflammatory cytokines at the periphery nor in the brain, but potentiated the enhancement of Interleukin-1-beta and Tumor Necrosis Factor-alpha expression specifically in the hippocampus after a peripheral immune challenge with lipopolysaccharide. Interestingly, whereas the same duration of HFD intake at adulthood induced similar weight gain and metabolic alterations as jHFD intake, it did neither affect spatial performance (long-term memory or reversal learning) nor lipopolysaccharide-induced cytokine expression in the hippocampus. Finally, spatial reversal learning enhanced Interleukin-1-beta in the hippocampus, but not in the frontal cortex and the hypothalamus, of jHFD-fed rats. These results indicate that juvenile HFD intake promotes exaggerated pro-inflammatory cytokines expression in the hippocampus which is likely to contribute to spatial memory impairment.


Subject(s)
Cytokines/blood , Diet, High-Fat/adverse effects , Encephalitis/immunology , Hippocampus/immunology , Memory/physiology , Animals , Brain/immunology , Eating , Male , Maze Learning/physiology , Memory, Long-Term/physiology , Memory, Short-Term/physiology , Obesity/immunology , Obesity/physiopathology , Rats , Rats, Wistar , Spatial Memory/physiology
11.
PLoS One ; 9(2): e87421, 2014.
Article in English | MEDLINE | ID: mdl-24505289

ABSTRACT

Normal aging is associated with vasopressin neuron adaptation, but little is known about its effects on the release of apelin, an aquaretic peptide colocalized with vasopressin. We found that plasma vasopressin concentrations were higher and plasma apelin concentrations lower in aged rats than in younger adults. The response of AVP/apelin neurons to osmotic challenge was impaired in aged rats. The overactivity of vasopressin neurons was sustained partly by the increased expression of Transient receptor potential vanilloid2 (Trpv2), because central Trpv blocker injection reversed the age-induced increase in plasma vasopressin concentration without modifying plasma apelin concentration. The morphofunctional plasticity of the supraoptic nucleus neuron-astrocyte network normally observed during chronic dehydration in adults appeared to be impaired in aged rats as well. IL-6 overproduction by astrocytes and low-grade microglial neuroinflammation may contribute to the modification of neuronal functioning during aging. Indeed, central treatment with antibodies against IL-6 decreased plasma vasopressin levels and increased plasma apelin concentration toward the values observed in younger adults. Conversely, minocycline treatment (inhibiting microglial metabolism) did not affect plasma vasopressin concentration, but increased plasma apelin concentration toward control values for younger adults. This study is the first to demonstrate dual vasopressin/apelin adaptation mediated by inflammatory molecules and neuronal Trpv2, during aging.


Subject(s)
Aging/blood , Astrocytes/metabolism , Intercellular Signaling Peptides and Proteins/blood , Neurons/metabolism , Vasopressins/blood , Aging/pathology , Animals , Anti-Bacterial Agents/pharmacology , Apelin , Astrocytes/pathology , Gene Expression Regulation/drug effects , Interleukin-6/metabolism , Male , Minocycline/pharmacology , Neurons/pathology , Osmotic Pressure , Rats , Rats, Wistar , TRPV Cation Channels/biosynthesis
12.
Hippocampus ; 22(11): 2095-100, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22593080

ABSTRACT

Increased consumption of high-fat diet (HFD) leads to obesity and adverse neurocognitive outcomes. Childhood and adolescence are important periods of brain maturation shaping cognitive function. These periods could consequently be particularly sensitive to the detrimental effects of HFD intake. In mice, juvenile and adulthood consumption of HFD induce similar morphometric and metabolic changes. However, only juvenile exposure to HFD abolishes relational memory flexibility, assessed after initial radial-maze concurrent spatial discrimination learning, and decreases neurogenesis. Our results identify a critical period of development covering adolescence with higher sensitivity to HFD-induced hippocampal dysfunction at both behavioral and cellular levels.


Subject(s)
Diet, High-Fat/adverse effects , Hippocampus/drug effects , Memory/drug effects , Neurogenesis/drug effects , Overweight/etiology , Age Factors , Animals , Blood Glucose/analysis , Body Weight/drug effects , Corticosterone/blood , Dentate Gyrus/chemistry , Dentate Gyrus/pathology , Discrimination Learning/drug effects , Doublecortin Domain Proteins , Energy Intake/drug effects , Hippocampus/pathology , Immunoenzyme Techniques , Leptin/blood , Lipids/blood , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/analysis , Nerve Tissue Proteins/analysis , Neuropeptides/analysis , Overweight/blood , Spatial Behavior/drug effects
13.
Neurobiol Dis ; 39(3): 393-401, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20470889

ABSTRACT

Interleukin-1beta acts on the CNS to induce fever, neuroendocrine activation, and behavioral changes, but cannot passively cross the blood-brain barrier. According to a widely accepted hypothesis interleukin-1beta induces the synthesis of cyclooxygenase-2 at the blood-brain interface, which produces prostaglandins that diffuse into brain parenchyma to activate neurons. We studied the role of brain cyclooxygenase-2 in interleukin-1beta-induced fever, neuroendocrine and behavioral responses and cellular activation by intracerebroventricular infusion of the cyclooxygenase-2 inhibitor NS-398. Central cyclooxygenase-2 inhibition attenuated extracellular signal-regulated kinase-1/2 phosphorylation and c-Fos induction in the median preoptic area and arcuate hypothalamus, but not in other hypothalamic or brainstem structures, after intraperitoneal interleukin-1beta administration. However, the same treatment did not affect interleukin-1beta-induced fever, rises in corticosterone or anorexia. These findings moderate the prevailing view and indicate that brain cyclooxygenase-2-dependent prostaglandin production is important to activation of the median preoptic and arcuate hypothalamus, but not necessarily involved in fever, rises in plasma corticosterone and anorexia after peripheral interleukin-1beta administration.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Cyclooxygenase 2/metabolism , Illness Behavior/drug effects , Interleukin-1beta/pharmacology , Neurons/metabolism , Preoptic Area/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Analysis of Variance , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Blotting, Western , Corticosterone/blood , Cyclooxygenase Inhibitors/pharmacology , Eating/drug effects , Interleukin-1beta/metabolism , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/drug effects , Nitrobenzenes/pharmacology , Phosphorylation/drug effects , Preoptic Area/drug effects , Random Allocation , Rats , Rats, Wistar , Sulfonamides/pharmacology
14.
Am J Physiol Endocrinol Metab ; 296(6): E1289-99, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19258490

ABSTRACT

The increase of plasma arginin-vasopressin (AVP) release, which translates hypothalamic AVP neuron activation in response to immune challenge, appears to occur independently of plasma osmolality or blood pressure changes. Many studies have shown that major inflammatory mediators produced in response to peripheral inflammation, such as prostaglandin (PG)-E(2) and interleukin (IL)-1beta, excite AVP neurons. However, in vivo electrical activation of AVP neurons was still not assessed in relation to plasma AVP release, osmolality, or blood pressure or to the expression and role of inflammatory molecules like PG-E(2), IL-1beta, IL-6, and tumor necrosis factor-alpha (TNFalpha). This study aims at elucidating those factors that underlie the activation of AVP neurons in response to immune stimulation mimicked by an intraperitoneal injection of lipopolysaccharide (LPS) in male Wistar rats. LPS treatment concomittanlty decreased diuresis and increased plasma AVP as well as AVP neuron activity in vivo, and these effects occurred as early as 30 min. Activation was sustained for more than 6 h. Plasma osmolality did not change, whereas blood pressure only transiently increased during the first hour post-LPS. PG-E(2), IL-1beta, and TNFalpha mRNA expression were raised 3 h after LPS, whereas IL-6 mRNA level increased 30 min post-LPS. In vivo electrophysiological recordings showed that brain IL-6 injection increased AVP neuron activity similarly to peripheral LPS treatment. In contrast, brain injection of anti-IL-6 antibodies prevented the LPS induced-activation of AVP neurons. Taken together, these results suggest that the early activation of AVP neurons in response to LPS injection is induced by brain IL-6.


Subject(s)
Arginine Vasopressin/blood , Inflammation/metabolism , Interleukin-6/genetics , Neurons/immunology , Supraoptic Nucleus/immunology , Animals , Antibodies/pharmacology , Blood Pressure/physiology , Dinoprostone/genetics , Dinoprostone/metabolism , Diuresis/physiology , Electric Stimulation , Inflammation/chemically induced , Inflammation/immunology , Interleukin-1beta/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Male , Membrane Potentials/physiology , Neurons/metabolism , Osmolar Concentration , RNA, Messenger/metabolism , Rats , Rats, Wistar , Supraoptic Nucleus/cytology , Tumor Necrosis Factor-alpha/genetics
15.
Dig Dis Sci ; 51(5): 986-95, 2006 May.
Article in English | MEDLINE | ID: mdl-16642426

ABSTRACT

Liver fibrosis is characterized by an activation of hepatic stellate cells (HSC). During primary culture HSC evolve from a quiescent into an activated phenotype which is characterized by alpha-smooth muscle actin (alpha-SMA) up-regulation, increase in cell growth, and extracellular matrix secretion. HSC culture with trans-resveratrol can lead to deactivation of myofibroblast-like HSC. We used an HSC line, PAV-1, to check the role of retinol and palmitic acid in the deactivation process of HSC. Using mass and metabolic-based methods, Western blot and immunocytochemistry assays, we demonstrated that treatment with palmitic acid (75 muM) alone or in combination with retinol (2 muM) significantly decreased cell proliferation and alpha-SMA expression. We also established that the association of both compounds strongly decreased collagen type I expression. Our results suggest the potential use of palmitic acid alone or in combination with retinol to induce HSC deactivation.


Subject(s)
Actins/metabolism , Collagen Type I/metabolism , Liver/drug effects , Palmitic Acid/pharmacology , Vitamin A/pharmacology , Actins/drug effects , Animals , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Collagen Type I/drug effects , Liver/cytology , Liver/metabolism , Liver Cirrhosis , Rats
SELECTION OF CITATIONS
SEARCH DETAIL