Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Neuroendocrinology ; : 1-16, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38852578

ABSTRACT

INTRODUCTION: Protein-enriched diets improve glycemic control in diabetes or emotional behavior in depressive patients. In mice, these benefits depend on intestinal gluconeogenesis activation by di-/tripeptides. Intestinal di-/tripeptides absorption is carried out by the peptide transporter 1, PEPT1. The lack of PEPT1 might thus alter glucose and emotional balance. METHODS: To determine the effects of PEPT1 deficiency under standard dietary conditions or during a dietary challenge known to promote both metabolic and cognitive dysfunction, insulin sensitivity, anxiety, and depressive-like traits, hippocampal serotonin (5-HT) and insulin signaling pathway were measured in wild-type (WT) and Pept1-/- mice fed either a chow or a high-fat high-sucrose (HF-HS) diet. RESULTS: Pept1-/- mice exhibited slight defects in insulin sensitivity and emotional behavior, which were aggravated by an HF-HS diet. Pept1-/- mice fed a chow diet had lower hippocampal 5-HT levels and exhibited cerebral insulin resistance under HF-HS diet. These defects were independent of intestinal gluconeogenesis but might be linked to increased plasma amino acids levels. CONCLUSION: Pept1-/- mice develop prediabetic and depressive-like traits and could thus be used to develop strategies to prevent or cure both diseases.

2.
Eur J Neurosci ; 59(7): 1558-1566, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38308520

ABSTRACT

The excitation-inhibition imbalance manifesting as epileptic activities in Alzheimer's disease is gaining more and more attention, and several potentially involved cellular and molecular pathways are currently under investigation. Based on in vitro studies, dopamine D1-type receptors in the anterior cingulate cortex and the hippocampus have been proposed to participate in this peculiar co-morbidity in mouse models of amyloidosis. Here, we tested the implication of dopaminergic transmission in vivo in the Tg2576 mouse model of Alzheimer's disease by monitoring epileptic activities via intracranial EEG before and after treatment with dopamine antagonists. Our results show that neither the D1-like dopamine receptor antagonist SCH23390 nor the D2-like dopamine receptor antagonist haloperidol reduces the frequency of epileptic activities. While requiring further investigation, our results indicate that on a systemic level, dopamine receptors are not significantly contributing to epilepsy observed in vivo in this mouse model of Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloidosis , Epilepsy , Mice , Animals , Dopamine Antagonists/pharmacology , Alzheimer Disease/drug therapy , Receptors, Dopamine D2/metabolism , Benzazepines/pharmacology , Benzazepines/therapeutic use , Receptors, Dopamine D1/metabolism , Epilepsy/drug therapy , Disease Models, Animal , Amyloidosis/drug therapy
3.
Molecules ; 29(2)2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38276574

ABSTRACT

This review highlights the advantages of high-precision liquid chromatography with an electrochemical detector (HPLC-ECD) in detecting and quantifying biological samples obtained through intracerebral microdialysis, specifically the serotonergic and dopaminergic systems: Serotonin (5-HT), 5-hydroxyindolacetic acid (5-HIAA), 3,4-dihydroxyphenylacetic acid (DOPAC), dopamine (DA), 3-metoxytryptamin (3-MT) and homovanillic acid (HVA). Recognized for its speed and selectivity, HPLC enables direct analysis of intracerebral microdialysis samples without complex derivatization. Various chromatographic methods, including reverse phase (RP), are explored for neurotransmitters (NTs) and metabolites separation. Electrochemical detector (ECD), particularly with glassy carbon (GC) electrodes, is emphasized for its simplicity and sensitivity, aimed at enhancing reproducibility through optimization strategies such as modified electrode materials. This paper underscores the determination of limits of detection (LOD) and quantification (LOQ) and the linear range (L.R.) showcasing the potential for real-time monitoring of compounds concentrations. A non-exhaustive compilation of literature values for LOD, LOQ, and L.R. from recent publications is included.


Subject(s)
Dopamine , Serotonin , Chromatography, High Pressure Liquid/methods , Reproducibility of Results , Dopamine/metabolism , Chromatography, Liquid , Serotonin/metabolism , Neurotransmitter Agents , 3,4-Dihydroxyphenylacetic Acid/metabolism , Hydroxyindoleacetic Acid/analysis , Hydroxyindoleacetic Acid/metabolism , Biogenic Monoamines
4.
Mol Psychiatry ; 29(3): 718-729, 2024 03.
Article in English | MEDLINE | ID: mdl-38123728

ABSTRACT

Chronic stress causes cognitive deficits, such as impairments in episodic-like hippocampus-dependent memory. Stress regulates an opioid-related neuropeptide named Nociceptin/Orphanin FQ (N/OFQ), the ligand of the G protein-coupled receptor NOP. Since this peptide has deleterious effects on memory, we hypothesized that the N/OFQ system could be a mediator of the negative effects of stress on memory. Chronic stress was mimicked by chronic exposure to corticosterone (CORT). The NOP receptor was either acutely blocked using selective antagonists, or knocked-down specifically in the hippocampus using genetic tools. Long-term memory was assessed in the object recognition (OR) and object location (OL) paradigms. Acute injection of NOP antagonists before learning had a negative impact on memory in naive mice whereas it restored memory performances in the chronic stress model. This rescue was associated with a normalization of neuronal cell activity in the CA3 part of the hippocampus. Chronic CORT induced an upregulation of the N/OFQ precursor in the hippocampus. Knock-down of the NOP receptor in the CA3/Dentate Gyrus region prevented memory deficits in the CORT model. These data demonstrate that blocking the N/OFQ system can be beneficial for long-term memory in a neuroendocrine model of chronic stress. We therefore suggest that NOP antagonists could be useful for the treatment of memory deficits in stress-related disorders.


Subject(s)
Corticosterone , Disease Models, Animal , Hippocampus , Memory, Long-Term , Nociceptin Receptor , Nociceptin , Opioid Peptides , Receptors, Opioid , Stress, Psychological , Animals , Receptors, Opioid/metabolism , Mice , Stress, Psychological/metabolism , Male , Hippocampus/metabolism , Hippocampus/drug effects , Opioid Peptides/metabolism , Memory, Long-Term/drug effects , Memory, Long-Term/physiology , Narcotic Antagonists/pharmacology , Mice, Inbred C57BL , Cognition/drug effects , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/drug therapy
5.
J Neuroendocrinol ; 35(12): e13344, 2023 12.
Article in English | MEDLINE | ID: mdl-37857383

ABSTRACT

OBJECTIVES: Intensive insulin therapy provides optimal glycemic control in patients with diabetes. However, intensive insulin therapy causes so-called iatrogenic hypoglycemia as a major adverse effect. The ventromedial hypothalamus (VMH) has been described as the primary brain area initiating the counter-regulatory response (CRR). Nevertheless, the VMH receives projections from other brain areas which could participate in the regulation of the CRR. In particular, studies suggest a potential role of the serotonin (5-HT) network. Thus, the objective of this study was to determine the contribution of 5-HT neurons in CRR control. METHODS: Complementary approaches have been used to test this hypothesis in quantifying the level of 5-HT in several brain areas by HPLC in response to insulin-induced hypoglycemia, measuring the electrical activity of dorsal raphe (DR) 5-HT neurons in response to insulin or decreased glucose level by patch-clamp electrophysiology; and measuring the CRR hormone glucagon as an index of the CRR to the modulation of the activity of 5-HT neurons using pharmacological or pharmacogenetic approaches. RESULTS: HPLC measurements show that the 5HIAA/5HT ratio is increased in several brain regions including the VMH in response to insulin-induced hypoglycemia. Patch-clamp electrophysiological recordings show that insulin, but not decreased glucose level, increases the firing frequency of DR 5-HT neurons in the DR. In vivo, both the pharmacological inhibition of 5-HT neurons by intraperitoneal injection of the 5-HT1A receptor agonist 8-OH-DPAT or the chemogenetic inhibition of these neurons reduce glucagon secretion, suggesting an impaired CRR. CONCLUSION: Taken together, these data highlight a new neuronal network involved in the regulation of the CRR. In particular, this study shows that DR 5-HT neurons detect iatrogenic hypoglycemia in response to the increased insulin level and may play an important role in the regulation of CRR.


Subject(s)
Glucagon , Hypoglycemia , Humans , Serotonergic Neurons , Serotonin/pharmacology , Hypoglycemia/chemically induced , Insulin/pharmacology , Glucose , Iatrogenic Disease
6.
Neuropharmacology ; 241: 109730, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37758019

ABSTRACT

Type 2 diabetes and obesity characterized by hallmarks of insulin resistance along with an imbalance in brain oxidative metabolism would impair intrinsic capacities (ICs), a new concept for assessing mental and physical functioning. Here, we explored the impact of physical activity on antioxidant responses and oxidative metabolism in discrete brain areas of HFD or standard diet (STD) fed mice but also its consequences on specific domains of ICs. 6-week-old Swiss male mice were exposed to a STD or a HFD for 16 weeks and half of the mice in each group had access to an activity wheel and the other half did not. As expected HFD mice displayed peripheral insulin resistance but also a persistent inhibition of aconitase activity in cortices revealing an increase in mitochondrial reactive oxygen species (ROS) production. Animals with access to the running wheel displayed an improvement of insulin sensitivity regardless of the diet factor whereas ROS production remained impaired. Moreover, although the access of the running wheel did not influence mitochondrial biomass, in the oxidative metabolism area, it produced a slight decrease in brain SOD1 and catalase expression notably in HFD fed mice. At the behavioural level, physical exercise produced anxiolytic/antidepressant-like responses and improved motor coordination in both STD and HFD fed mice. However, this non-pharmacological intervention failed to enhance cognitive performance. These findings paint a contrasting landscape about physical exercise as a non-pharmacological intervention for positively orienting the aging trajectory.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Animals , Male , Mice , Brain/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance/physiology , Mice, Inbred C57BL , Oxidative Stress , Reactive Oxygen Species , Physical Conditioning, Animal/physiology
7.
Transl Psychiatry ; 13(1): 302, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37775532

ABSTRACT

Selective serotonin reuptake inhibitors (SSRI) are common first-line treatments for major depression. However, a significant number of depressed patients do not respond adequately to these pharmacological treatments. In the present preclinical study, we demonstrate that organic cation transporter 2 (OCT2), an atypical monoamine transporter, contributes to the effects of SSRI by regulating the routing of the essential amino acid tryptophan to the brain. Contrarily to wild-type mice, OCT2-invalidated mice failed to respond to prolonged fluoxetine treatment in a chronic depression model induced by corticosterone exposure recapitulating core symptoms of depression, i.e., anhedonia, social withdrawal, anxiety, and memory impairment. After corticosterone and fluoxetine treatment, the levels of tryptophan and its metabolites serotonin and kynurenine were decreased in the brain of OCT2 mutant mice compared to wild-type mice and reciprocally tryptophan and kynurenine levels were increased in mutants' plasma. OCT2 was detected by immunofluorescence in several structures at the blood-cerebrospinal fluid (CSF) or brain-CSF interface. Tryptophan supplementation during fluoxetine treatment increased brain concentrations of tryptophan and, more discreetly, of 5-HT in wild-type and OCT2 mutant mice. Importantly, tryptophan supplementation improved the sensitivity to fluoxetine treatment of OCT2 mutant mice, impacting chiefly anhedonia and short-term memory. Western blot analysis showed that glycogen synthase kinase-3ß (GSK3ß) and mammalian/mechanistic target of rapamycin (mTOR) intracellular signaling was impaired in OCT2 mutant mice brain after corticosterone and fluoxetine treatment and, conversely, tryptophan supplementation recruited selectively the mTOR protein complex 2. This study provides the first evidence of the physiological relevance of OCT2-mediated tryptophan transport, and its biological consequences on serotonin homeostasis in the brain and SSRI efficacy.


Subject(s)
Depressive Disorder, Major , Organic Cation Transporter 2 , Selective Serotonin Reuptake Inhibitors , Animals , Mice , Anhedonia , Antidepressive Agents/therapeutic use , Brain/metabolism , Corticosterone/pharmacology , Depressive Disorder, Major/drug therapy , Fluoxetine/pharmacology , Kynurenine/metabolism , Organic Cation Transporter 2/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin/metabolism , Tryptophan/metabolism
8.
Transl Psychiatry ; 13(1): 227, 2023 06 26.
Article in English | MEDLINE | ID: mdl-37365183

ABSTRACT

Mood disorders are associated with hypothalamic-pituitary-adrenal axis overactivity resulting from a decreased inhibitory feedback exerted by the hippocampus on this brain structure. Growing evidence suggests that antidepressants would regulate hippocampal excitatory/inhibitory balance to restore an effective inhibition on this stress axis. While these pharmacological compounds produce beneficial clinical effects, they also have limitations including their long delay of action. Interestingly, non-pharmacological strategies such as environmental enrichment improve therapeutic outcome in depressed patients as in animal models of depression. However, whether exposure to enriched environment also reduces the delay of action of antidepressants remains unknown. We investigated this issue using the corticosterone-induced mouse model of depression, submitted to antidepressant treatment by venlafaxine, alone or in combination with enriched housing. We found that the anxio-depressive phenotype of male mice was improved after only two weeks of venlafaxine treatment when combined with enriched housing, which is six weeks earlier than mice treated with venlafaxine but housed in standard conditions. Furthermore, venlafaxine combined with exposure to enriched environment is associated with a reduction in the number of parvalbumin-positive neurons surrounded by perineuronal nets (PNN) in the mouse hippocampus. We then showed that the presence of PNN in depressed mice prevented their behavioral recovery, while pharmacological degradation of hippocampal PNN accelerated the antidepressant action of venlafaxine. Altogether, our data support the idea that non-pharmacological strategies can shorten the onset of action of antidepressants and further identifies PV interneurons as relevant actors of this effect.


Subject(s)
Parvalbumins , Selective Serotonin Reuptake Inhibitors , Mice , Male , Animals , Venlafaxine Hydrochloride/pharmacology , Parvalbumins/metabolism , Serotonin/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Antidepressive Agents/metabolism , Interneurons/metabolism
9.
Hum Mol Genet ; 32(2): 244-261, 2023 01 06.
Article in English | MEDLINE | ID: mdl-35951020

ABSTRACT

The neuronal-specific SNORD115 has gathered interest because its deficiency may contribute to the pathophysiology of Prader-Willi syndrome (PWS), possibly by altering post-transcriptional regulation of the gene encoding the serotonin (HTR2C) receptor. Yet, Snord115-KO mice do not resume the main symptoms of PWS, and only subtle-altered A-to-I RNA editing of Htr2c mRNAs was uncovered. Because HTR2C signaling fine-tunes the activity of monoaminergic neurons, we addressed the hypothesis that lack of Snord115 alters monoaminergic systems. We first showed that Snord115 was expressed in both monoaminergic and non-monoaminergic cells of the ventral tegmental area (VTA) and the dorsal raphe nucleus (DRN) harboring cell bodies of dopaminergic and serotonergic neurons, respectively. Measuring the tissue level of monoamines and metabolites, we found very few differences except that the content of homovanillic acid-a metabolite of dopamine-was decreased in the orbitofrontal and prefrontal cortex of Snord115-KO mice. The latter effects were, however, associated with a few changes in monoamine tissue content connectivity across the 12 sampled brain regions. Using in vivo single-cell extracellular recordings, we reported that the firing rate of VTA dopaminergic neurons and DRN serotonergic neurons was significantly increased in Snord115-KO mice. These neural circuit dysfunctions were not, however, associated with apparent defects in binge eating, conditioned place preference to cocaine, cocaine-induced hyperlocomotion or compulsive behavior. Altogether, our multiscale study shows that the absence of Snord115 impacts central monoaminergic circuits to an extent that does not elicit gross behavioral abnormalities.


Subject(s)
Brain , Prader-Willi Syndrome , Mice , Animals , Brain/metabolism , Neurons/metabolism , Dopamine/metabolism , Prefrontal Cortex/metabolism , Serotonin/metabolism , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/metabolism
10.
Fundam Clin Pharmacol ; 37(1): 94-106, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35996325

ABSTRACT

Brain insulin resistance has been pointed to as a possible link between diabetes and neuropsychiatric disorders; therefore, therapeutic approaches using anti-diabetic drugs to improve insulin levels or signaling could prevent type 1 (T1D) and type 2 diabetes mellitus (T2D)-induced brain dysfunction. The present study aimed to determine whether metformin exerts beneficial effects on metabolic and neurobehavioral outcomes in the streptozotocin (STZ)-induced T1D model and western diet (WD)-induced obesity model in male Swiss mice. T1D was induced by intraperitoneal injection of STZ (50 mg/kg, for five consecutive days). The animals were then treated daily with saline or metformin (200 mg/kg/day, oral gavage), and a battery of tests recapitulating different neurobehavioral anomalies related to anxiogenic/depressive-like phenotype was conducted after 18 days. WD-induced obesity was modeled in mice by high-fat and high-fructose diet (HFFD) feeding for 15 days. In the sequence, control and diet-induced obesity mice were treated daily with saline or metformin (200 mg/kg/day), and a battery of behavioral tests was performed after 17 days. STZ injection and WD feeding induced metabolic and neurobehavioral impairments in mice. Remarkably, metformin improved the metabolic and neurobehavioral parameters in WD-induced obesity mice. Moreover, metformin ameliorated STZ-induced neurobehavioral deficits while it failed to improve the associated metabolic impairments. The beneficial effects of metformin in STZ-induced neurobehavioral impairments were not mediated by improving peripheral insulin signaling. Our results suggest that conventional diabetes treatment could be repurposed to simultaneously improve neurobehavioral symptoms and diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Metformin , Mice , Male , Animals , Metformin/pharmacology , Metformin/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/therapeutic use , Streptozocin , Diet, Western/adverse effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/chemically induced , Insulin , Glucose/metabolism , Obesity/drug therapy , Blood Glucose , Diet, High-Fat/adverse effects
11.
Int J Mol Sci ; 23(21)2022 Oct 30.
Article in English | MEDLINE | ID: mdl-36362016

ABSTRACT

Studies suggest that astrocytic connexins (Cx) have an important role in the regulation of high brain functions through their ability to establish fine-tuned communication with neurons within the tripartite synapse. In light of these properties, growing evidence suggests a role of Cx in psychiatric disorders such as major depression but also in the therapeutic activity of antidepressant drugs. However, the real impact of Cx on treatment response and the underlying neurobiological mechanisms remain yet to be clarified. On this ground, the present study was designed to evaluate the functional activity of Cx in a mouse model of depression based on chronic corticosterone exposure and to determine to which extent their pharmacological inactivation influences the antidepressant-like activity of venlafaxine (VENLA). On the one hand, our results indicate that depressed mice have impaired Cx-based gap-junction and hemichannel activities. On the other hand, while VENLA exerts robust antidepressant-like activity in depressed mice; this effect is abolished by the pharmacological inhibition of Cx with carbenoxolone (CBX). Interestingly, the combination of VENLA and CBX is also associated with a higher rate of relapse after treatment withdrawal. To our knowledge, this study is one of the first to develop a model of relapse, and our results reveal that Cx-mediated dynamic neuroglial interactions play a critical role in the efficacy of monoaminergic antidepressant drugs, thus providing new targets for the treatment of depression.


Subject(s)
Astrocytes , Connexins , Depressive Disorder , Animals , Mice , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Astrocytes/drug effects , Astrocytes/metabolism , Carbenoxolone/pharmacology , Connexins/drug effects , Connexins/metabolism , Phenotype , Recurrence , Depression/drug therapy , Depression/metabolism , Depressive Disorder/drug therapy , Depressive Disorder/metabolism
12.
Mol Psychiatry ; 2022 Oct 07.
Article in English | MEDLINE | ID: mdl-36207585

ABSTRACT

Type-2 Diabetes (T2D) is characterized by insulin resistance and accompanied by psychiatric comorbidities including major depressive disorders (MDD). Patients with T2D are twice more likely to suffer from MDD and clinical studies have shown that insulin resistance is positively correlated with the severity of depressive symptoms. However, the potential contribution of central insulin signaling in MDD in patients with T2D remains elusive. Here we hypothesized that insulin modulates the serotonergic (5-HT) system to control emotional behavior and that insulin resistance in 5-HT neurons contributes to the development of mood disorders in T2D. Our results show that insulin directly modulates the activity of dorsal raphe (DR) 5-HT neurons to dampen 5-HT neurotransmission through a 5-HT1A receptor-mediated inhibitory feedback. In addition, insulin-induced 5-HT neuromodulation is necessary to promote anxiolytic-like effect in response to intranasal insulin delivery. Interestingly, such an anxiolytic effect of intranasal insulin as well as the response of DR 5-HT neurons to insulin are both blunted in high-fat diet-fed T2D animals. Altogether, these findings point to a novel mechanism by which insulin directly modulates the activity of DR 5-HT neurons to dampen 5-HT neurotransmission and control emotional behaviors, and emphasize the idea that impaired insulin-sensitivity in these neurons is critical for the development of T2D-associated mood disorders.

13.
Psychopharmacology (Berl) ; 239(9): 2735-2752, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35947166

ABSTRACT

Major depression (MD) is one of the most common psychiatric disorders worldwide. Currently, the first-line treatment for MD targets the serotonin system but these drugs, notably the selective serotonin reuptake inhibitors, usually need 4 to 6 weeks before the benefit is felt and a significant proportion of patients shows an unsatisfactory response. Numerous treatments have been developed to circumvent these issues as venlafaxine, a mixed serotonin-norepinephrine reuptake inhibitor that binds and blocks both the SERT and NET transporters. Despite this pharmacological profile, it is difficult to have a valuable insight into its ability to produce more robust efficacy than single-acting agents. In this review, we provide an in-depth characterization of the pharmacological properties of venlafaxine from in vitro data to preclinical and clinical efficacy in depressed patients and animal models of depression to propose an indirect comparison with the most common antidepressants. Preclinical studies show that the antidepressant effect of venlafaxine is often associated with an enhancement of serotonergic neurotransmission at low doses. High doses of venlafaxine, which elicit a concomitant increase in 5-HT and NE tone, is associated with changes in different forms of plasticity in discrete brain areas. In particular, the hippocampus appears to play a crucial role in venlafaxine-mediated antidepressant effects notably by regulating processes such as adult hippocampal neurogenesis or the excitatory/inhibitory balance. Overall, depending on the dose used, venlafaxine shows a high efficacy on depressive-like symptoms in relevant animal models but to the same extent as common antidepressants. However, these data are counterbalanced by a lower tolerance. In conclusion, venlafaxine appears to be one of the most effective treatments for treatment of major depression. Still, direct comparative studies are warranted to provide definitive conclusions about its superiority.


Subject(s)
Depressive Disorder, Major , Serotonin , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Cyclohexanols/pharmacology , Cyclohexanols/therapeutic use , Depressive Disorder, Major/drug therapy , Humans , Selective Serotonin Reuptake Inhibitors/pharmacology , Venlafaxine Hydrochloride/pharmacology , Venlafaxine Hydrochloride/therapeutic use
14.
Trends Endocrinol Metab ; 33(1): 18-35, 2022 01.
Article in English | MEDLINE | ID: mdl-34750064

ABSTRACT

The incidence of depression and anxiety is amplified by obesity. Mounting evidence reveals that the psychiatric consequences of obesity stem from poor diet, inactivity, and visceral adipose accumulation. Resulting metabolic and vascular dysfunction, including inflammation, insulin and leptin resistance, and hypertension, have emerged as key risks to depression and anxiety development. Recent research advancements are exposing the important contribution of these different corollaries of obesity and their impact on neuroimmune status and the neural circuits controlling mood and emotional states. Along these lines, this review connects the clinical manifestations of depression and anxiety in obesity to our current understanding of the origins and biology of immunometabolic threats to central nervous system function and behavior.


Subject(s)
Anxiety , Depression , Depression/epidemiology , Depression/etiology , Depression/metabolism , Humans , Inflammation/metabolism , Obesity/complications , Obesity/epidemiology , Prevalence
15.
NPJ Regen Med ; 6(1): 63, 2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34650070

ABSTRACT

Tissue repair after injury in adult mammals, usually results in scarring and loss of function in contrast to lower vertebrates such as the newt and zebrafish that regenerate. Understanding the regulatory processes that guide the outcome of tissue repair is therefore a concerning challenge for regenerative medicine. In multiple regenerative animal species, the nerve dependence of regeneration is well established, but the nature of the innervation required for tissue regeneration remains largely undefined. Using our model of induced adipose tissue regeneration in adult mice, we demonstrate here that nociceptive nerves promote regeneration and their removal impairs tissue regeneration. We also show that blocking the receptor for the nociceptive neuropeptide calcitonin gene-related peptide (CGRP) inhibits regeneration, whereas CGRP administration induces regeneration. These findings reveal that peptidergic nociceptive neurons are required for adult mice tissue regeneration.

16.
Prog Brain Res ; 259: 197-228, 2021.
Article in English | MEDLINE | ID: mdl-33541677

ABSTRACT

Astrocytes were traditionally regarded as cells important to neuronal activity, providing both metabolic and structural supports. Recent evidence suggests that they may also play a crucial role in the control of higher brain functions. In keeping with this hypothesis, it is now well accepted that astrocytes contribute to stress but also react to antidepressant drugs as they express serotonergic transporters and receptors. However, the downstream mechanisms leading to the fine-tuned regulation of mood are still unknown. This chapter pays attention to the role of astrocytes in the regulation of emotional behavior and related serotonergic neurotransmission. In particular, it gives a current state of the clinical and preclinical evidence showing that astrocytes respond to environmental conditions and antidepressant drugs through the release of gliotransmitters and neurotrophic factors which in turn, influence serotonergic tone in discrete brain areas. This state-of-the-art review aims at demonstrating the remarkable potential for novel therapeutic antidepressant strategies targeting these glial cells.


Subject(s)
Astrocytes , Serotonergic Neurons , Antidepressive Agents/pharmacology , Humans , Mood Disorders/drug therapy , Synaptic Transmission
17.
Neuroendocrinology ; 111(12): 1249-1265, 2021.
Article in English | MEDLINE | ID: mdl-33429400

ABSTRACT

INTRODUCTION: Several studies have suggested that diet, especially the one enriched in microbiota-fermented fibers or fat, regulates behavior. The underlying mechanisms are currently unknown. We previously reported that certain macronutrients (fermentable fiber and protein) regulate energy homeostasis via the activation of intestinal gluconeogenesis (IGN), which generates a neural signal to the brain. We hypothesized that these nutriments might control behavior using the same gut-brain circuit. METHODS: Wild-type and IGN-deficient mice were fed chow or diets enriched in protein or fiber. Changes in their behavior were assessed using suited tests. Hippocampal neurogenesis, extracellular levels of serotonin, and protein expression levels were assessed by immunofluorescence, in vivo dialysis, and Western blotting, respectively. IGN was rescued by infusing glucose into the portal vein of IGN-deficient mice. RESULTS: We show here that both fiber- and protein-enriched diets exert beneficial actions on anxiety-like and depressive-like behaviors. These benefits do not occur in mice lacking IGN. Consistently, IGN-deficient mice display hallmarks of depressive-like disorders, including decreased hippocampal neurogenesis, basal hyperactivity, and deregulation of the hypothalamic-pituitary-adrenal axis, which are associated with increased expression of the precursor of corticotropin-releasing hormone in the hypothalamus and decreased expression of the glucocorticoid receptor in the hippocampus. These neurobiological alterations are corrected by portal glucose infusion mimicking IGN. CONCLUSION: IGN translates nutritional information, allowing the brain to finely coordinate energy metabolism and behavior.


Subject(s)
Anxiety/metabolism , Behavior, Animal/physiology , Depression/metabolism , Dietary Fiber/metabolism , Dietary Proteins/metabolism , Gluconeogenesis/physiology , Intestine, Small/metabolism , Animals , Disease Models, Animal , Mice
18.
J Neuroendocrinol ; 33(4): e12928, 2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33506507

ABSTRACT

The idea that insulin could influence emotional behaviours has long been suggested. However, the underlying mechanisms have yet to be solved and there is no direct and clear-cut evidence demonstrating that such action involves brain serotonergic neurones. Indeed, initial arguments in favour of the association between insulin, serotonin and mood arise from clinical or animal studies showing that impaired insulin action in type 1 or type 2 diabetes causes anxiety- and depressive symptoms along with blunted plasma and brain serotonin levels. The present review synthesises the main mechanistic hypotheses that might explain the comorbidity between diabetes and depression. It also provides a state of knowledge of the direct and indirect experimental evidence that insulin modulates brain serotonergic neurones. Finally, it highlights the literature suggesting that antidiabetic drugs present antidepressant-like effects and, conversely, that serotonergic antidepressants impact glucose homeostasis. Overall, this review provides mechanistic insights into how insulin signalling alters serotonergic neurotransmission and related behaviours bringing new targets for therapeutic options.

19.
Electrophoresis ; 42(9-10): 1108-1114, 2021 05.
Article in English | MEDLINE | ID: mdl-33469939

ABSTRACT

Capillary electrophoresis coupled to LED-induced fluorescence detection is a robust and sensitive technique used for amino acids (AA) analysis in biological media, after labeling with 3-(4-carboxybenzoyl)quinoline-2-carboxaldehyde (CBQCA). We wanted to quantitate in plasma tryptophan (Trp), tyrosine (Tyr), valine (Val), and isoleucine (Ile). Among the different labeled AA-CBQCA, Trp has the lowest fluorescence yield, which makes its detection and quantification very difficult in biological samples such as plasma. We tried to improve Trp analysis by CE/LED-induced fluorescence detection to its maximal sensitivity by using large volume sample stacking as a preconcentration step in our analytical protocol. At pH 9.5, this step caused a drop in resolution during the separation of the four AAs and it was therefore necessary to work at pH 10. We have found that Tyr, Val, Ile, and Trp are detected and well separated from the other AAs, but Trp cannot be quantified in plasma samples, mainly because of the low fluorescence yield of the Trp-CBQCA derivative. The recorded LOD is 0.18 µM for Trp-CBQCA in standard solution with a resolution between Trp and Tyr of 1.2, while the LOD is 6 µM in plasma with the same resolution. Trp, Tyr, Val, and Ile are, however, efficiently quantified when using a 3 M acetic acid electrolyte and CE associated with capacitively coupled contactless conductivity detection, which also has the advantage of not requiring derivatization or large volume sample stacking. This article demonstrates, for the CE user, that quantitative analysis of these four AA in mouse plasma can be performed by CE-fluorescence after CBQCA labeling, with the exception of Trp. It can be advantageously replaced by CE/capacitively coupled contactless conductivity detection, the only efficient one for Trp, Tyr, Val, and Ile quantification. In this case, the LOD for Trp is 2 µM. The four AAs are separated with resolution with neighbors above 1.5.


Subject(s)
Amino Acids/blood , Animals , Isoleucine , Mice , Quinolines , Tryptophan , Tyrosine , Valine
20.
Int J Mol Sci ; 22(2)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33477830

ABSTRACT

The forced swim stress test (FST) is widely used for screening pharmacological or non-pharmacological strategies with potential antidepressant activities. Recent data have suggested that repeated FST for five consecutive days (i.e., 5d-RFSS) could be used to generate a robust depressive-like phenotype in mice. However, the face, construct, and predictive validities of 5d-RFSS have been recently challenged. This study took advantage of recent findings showing that mice vulnerability to anxiety is enhanced when animals are stressed during the dark phase, to provide new insight into the relevance of this model. Our results showed a progressive increase in time of immobility in 5d-RFSS mice relative to control non-stressed animals (sham). Three weeks later, we noticed that 5d-RFSS mice injected with the vehicle compound (Veh) still exhibited a high level of immobility in the FST whereas this behavior was reversed by the antidepressant drug amitriptyline (AMI). However, 5d-RFSS/Veh and 5d-RFSS mice/AMI mice showed normal performances in the open field, the novelty suppressed feeding and the tail suspension tests. Despite this lack of generalized behavioral deficits, an impairment of different parameters characterizing the hypothalamic-pituitary-adrenal (HPA) axis reactivity was evidenced in 5d-RFSS mice/Veh but not in 5d-RFSS mice/AMI. Despite anomalies in the HPA axis, the activity of the central serotonergic system remained unaffected in 5d-RFSS mice relative to controls. From our results, it is suggested that learned immobility does not replicate the broad spectrum of depressive symptoms observed in other chronic models of depression such as the unpredictable chronic mild stress (UCMS) model, the chronic social defeat stress (CSDS) model or chronic corticosterone (CORT) exposure but its influence on the HPA axis is remarkable. Further experiments are warranted to makes this model suitable for modelling depression and therefore refine its translational applicability.


Subject(s)
Anxiety/drug therapy , Corticosterone/pharmacology , Depressive Disorder/drug therapy , Stress, Psychological/drug therapy , Animals , Antidepressive Agents/pharmacology , Anxiety/physiopathology , Anxiety Disorders/drug therapy , Anxiety Disorders/pathology , Behavior, Animal/drug effects , Depressive Disorder/pathology , Disease Models, Animal , Humans , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/pathology , Mice , Phenotype , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/pathology , Stress, Psychological/pathology , Swimming
SELECTION OF CITATIONS
SEARCH DETAIL