Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Neurotherapeutics ; 21(2): e00334, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38368170

ABSTRACT

Psychosis in Parkinson's disease is a common phenomenon associated with poor outcomes. To clarify the pathophysiology of this condition and the mechanisms of antipsychotic treatments, we have here characterized the neurophysiological brain states induced by clozapine, pimavanserin, and the novel prospective antipsychotic mesdopetam in a rodent model of Parkinson's disease psychosis, based on chronic dopaminergic denervation by 6-OHDA lesions, levodopa priming, and the acute administration of an NMDA antagonist. Parallel recordings of local field potentials from eleven cortical and sub-cortical regions revealed shared neurophysiological treatment effects for the three compounds, despite their different pharmacological profiles, involving reversal of features associated with the psychotomimetic state, such as a reduction of aberrant high-frequency oscillations in prefrontal structures together with a decrease of abnormal synchronization between different brain regions. Other drug-induced neurophysiological features were more specific to each treatment, affecting network oscillation frequencies and entropy, pointing to discrete differences in mechanisms of action. These findings indicate that neurophysiological characterization of brain states is particularly informative when evaluating therapeutic mechanisms in conditions involving symptoms that are difficult to assess in rodents such as psychosis, and that mesdopetam should be further explored as a potential novel antipsychotic treatment option for Parkinson psychosis.


Subject(s)
Antipsychotic Agents , Clozapine , Parkinson Disease , Phenyl Ethers , Piperidines , Propylamines , Psychotic Disorders , Urea/analogs & derivatives , Animals , Clozapine/pharmacology , Parkinson Disease/complications , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Rodentia , Prospective Studies , Psychotic Disorders/etiology , Psychotic Disorders/complications
2.
Front Pharmacol ; 14: 1087171, 2023.
Article in English | MEDLINE | ID: mdl-36778010

ABSTRACT

The dopamine D4 receptor (D4R) is expressed in the retina, prefrontal cortex, and autonomic nervous system and has been implicated in attention deficit hyperactivity disorder (ADHD), substance use disorders, and erectile dysfunction. D4R has also been investigated as a target for antipsychotics due to its high affinity for clozapine. As opposed to the closely related dopamine D2 receptor (D2R), dopamine-induced arrestin recruitment and desensitization at the D4R have not been studied in detail. Indeed, some earlier investigations could not detect arrestin recruitment and desensitization of this receptor upon its activation by agonist. Here, we used a novel nanoluciferase complementation assay to study dopamine-induced recruitment of ß-arrestin2 (ßarr2; also known as arrestin3) and G protein-coupled receptor kinase-2 (GRK2) to the D4R in HEK293T cells. We also studied desensitization of D4R-evoked G protein-coupled inward rectifier potassium (GIRK; also known as Kir3) current responses in Xenopus oocytes. Furthermore, the effect of coexpression of GRK2 on ßarr2 recruitment and GIRK response desensitization was examined. The results suggest that coexpression of GRK2 enhanced the potency of dopamine to induce ßarr2 recruitment to the D4R and accelerated the rate of desensitization of D4R-evoked GIRK responses. The present study reveals new details about the regulation of arrestin recruitment to the D4R and thus increases our understanding of the signaling and desensitization of this receptor.

3.
Article in English | MEDLINE | ID: mdl-36038021

ABSTRACT

Major depressive disorder is a highly prevalent psychiatric condition. Metalloproteinase 9 (MMP-9), a gelatinase involved in synaptic plasticity, learning and memory processes, is elevated in both chronic stress animal models and human peripheral blood samples of depressed patients. In this study we have evaluated the MMP-9 activity and protein expression in brain areas relevant to depression using the chronic corticosterone mouse model of depression. These mice show a depressive- and anxious-like behaviour. The MMP-9 activity and protein levels are significantly elevated in both the hippocampus and the cortex, and nectin-3 levels are lower in these brain areas in this model. In particular, these mice display an increased gelatinase activity in the CA1 and CA3 subfields of the hippocampus and in the internal layer of the prefrontal cortex. Moreover, the immobility time in the tail suspension test presents a positive correlation with the cortical MMP-9 activity, and a negative correlation with nectin-3 levels. In conclusion, the chronic corticosterone model of depression leads to an increase in the protein expression and activity of MMP-9 and a reduction of its substrate nectin-3 in relevant areas implicated in this disease. The MMP-9 activity correlates with behavioural despair in this model of depression. All these findings support the role of MMP-9 in the pathophysiology of depression, and as a putative target to develop novel antidepressant drugs.


Subject(s)
Corticosterone , Depressive Disorder, Major , Animals , Humans , Mice , Antidepressive Agents/therapeutic use , Behavior, Animal , Depression/metabolism , Depressive Disorder, Major/drug therapy , Disease Models, Animal , Hippocampus/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/therapeutic use , Nectins/metabolism
4.
Brain Struct Funct ; 226(8): 2603-2616, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34363521

ABSTRACT

Ketamine has rapid and robust antidepressant effects. However, unwanted psychotomimetic effects limit its widespread use. Hence, several studies examined whether GluN2B-subunit selective NMDA antagonists would exhibit a better therapeutic profile. Although preclinical work has revealed some of the mechanisms of action of ketamine at cellular and molecular levels, the impact on brain circuitry is poorly understood. Several neuroimaging studies have examined the functional changes in the brain induced by acute administration of ketamine and Ro 25-6981 (a GluN2B-subunit selective antagonist), but the changes in the microstructure of gray and white matter have received less attention. Here, the effects of ketamine and Ro 25-6981 on gray and white matter integrity in male Sprague-Dawley rats were determined using diffusion-weighted magnetic resonance imaging (DWI). In addition, DWI-based structural brain networks were estimated and connectivity metrics were computed at the regional level. Immunohistochemical analyses were also performed to determine whether changes in myelin basic protein (MBP) and neurofilament heavy-chain protein (NF200) may underlie connectivity changes. In general, ketamine and Ro 25-6981 showed some opposite structural alterations, but both compounds coincided only in increasing the fractional anisotropy in infralimbic prefrontal cortex and dorsal raphe nucleus. These changes were associated with increments of NF200 in deep layers of the infralimbic cortex (together with increased MBP) and the dorsal raphe nucleus. Our results suggest that the synthesis of NF200 and MBP may contribute to the formation of new dendritic spines and myelination, respectively. We also suggest that the increase of fractional anisotropy of the infralimbic and dorsal raphe nucleus areas could represent a biomarker of a rapid antidepressant response.


Subject(s)
Antidepressive Agents , Ketamine , Phenols , Piperidines , Animals , Antidepressive Agents/pharmacology , Dorsal Raphe Nucleus , Ketamine/pharmacology , Magnetic Resonance Imaging , Male , Phenols/metabolism , Piperidines/metabolism , Prefrontal Cortex , Rats , Rats, Sprague-Dawley
5.
Int J Mol Sci ; 22(16)2021 Aug 12.
Article in English | MEDLINE | ID: mdl-34445375

ABSTRACT

Fast and sustained antidepressant effects of ketamine identified the mammalian target of rapamycin (mTOR) signaling pathway as the main modulator of its antidepressive effects. Thus, mTOR signaling has become integral for the preclinical evaluation of novel compounds to treat depression. However, causality between mTOR and depression has yet to be determined. To address this, we knocked down mTOR expression in mice using an acute intracerebral infusion of small interfering RNAs (siRNA) in the infralimbic (IL) or prelimbic (PrL) cortices of the medial prefrontal cortex (mPFC), and evaluated depressive- and anxious-like behaviors. mTOR knockdown in IL, but not PrL, cortex produced a robust depressive-like phenotype in mice, as assessed in the forced swimming test (FST) and the tail suspension test (TST). This phenotype was associated with significant reductions of mTOR mRNA and protein levels 48 h post-infusion. In parallel, decreased brain-derived neurotrophic factor (BDNF) expression was found bilaterally in both IL and PrL cortices along with a dysregulation of serotonin (5-HT) and glutamate (Glu) release in the dorsal raphe nucleus (DRN). Overall, our results demonstrate causality between mTOR expression in the IL cortex and depressive-like behaviors, but not in anxiety.


Subject(s)
Depression/psychology , Prefrontal Cortex/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Depression/genetics , Depression/metabolism , Disease Models, Animal , Dorsal Raphe Nucleus/metabolism , Gene Knockdown Techniques , Glutamic Acid/metabolism , Hindlimb Suspension , Male , Mice , Serotonin/metabolism , Swimming
6.
ACS Chem Neurosci ; 12(11): 2036-2044, 2021 06 02.
Article in English | MEDLINE | ID: mdl-33974408

ABSTRACT

Clinical and preclinical studies report the implication of 5-hydroxytryptamine 4 receptors (5-HT4Rs) in depression and anxiety. Here, we tested whether the absence of 5-HT4Rs influences the response to the antidepressant fluoxetine in mice subjected to chronic corticosterone administration, an animal model of depression and anxiety. Therefore, the effects of chronic administration of fluoxetine in corticosterone-treated wild-type (WT) and 5-HT4R knockout (KO) mice were evaluated in the open-field and novelty suppressed feeding tests. As 5-HT1A receptor (5-HT1AR) and brain-derived neurotrophic factor (BDNF) are critically involved in depression and anxiety, we further evaluated 5-HT1A receptor functionality by [35S]GTPγS autoradiography and BDNF mRNA expression by in situ hybridization techniques. We found that 5-HT4R KO and WT mice displayed anxiety- and depressive-like behavior following chronic administration of corticosterone, as evidenced in the open-field and novelty suppressed feeding tests. In the open-field, a decreased central activity was observed in naïve and corticosterone-treated mice of both genotypes following chronic fluoxetine administration. In the novelty suppressed feeding test, a predictive paradigm of antidepressant activity, chronic treatment with fluoxetine reverted the latency to eat in both genotypes. The antidepressant also potentiated the corticosterone-induced desensitization of the 5-HT1AR in the dorsal raphe nucleus. Further, chronic fluoxetine increased BDNF mRNA expression in the dentate gyrus of the hippocampus in corticosterone-treated mice of both genotypes. Therefore, our findings indicate that the behavioral effects of fluoxetine in the corticosterone model of depression and anxiety appear not to be dependent on 5-HT4Rs.


Subject(s)
Corticosterone , Fluoxetine , Animals , Anxiety , Depression/drug therapy , Fluoxetine/pharmacology , Hippocampus , Mice , Serotonin
7.
Biochem Pharmacol ; 185: 114433, 2021 03.
Article in English | MEDLINE | ID: mdl-33513342

ABSTRACT

Major Depression is a severe psychiatric condition with a still poorly understood etiology. In the last years, evidence supporting the neuroinflammatory hypothesis of depression has increased. In the current clinical scenario, in which the available treatments for depression is far from optimal, there is an urgent need to develop fast-acting drugs with fewer side effects. In this regard, recent pieces of evidence suggest that cannabidiol (CBD), the major non-psychotropic component of Cannabis sativa with anti-inflammatory properties, appears as a drug with antidepressant properties. In this work, CBD 30 mg/kg was administered systemically to mice 30 min before lipopolysaccharide (LPS; 0.83 mg/kg) administration as a neuroinflammatory model, and behavioral tests for depressive-, anhedonic- and anxious-like behavior were performed. NF-ĸB, IκBα and PPARγ levels were analyzed by western blot in nuclear and cytosolic fractions of cortical samples. IL-6 and TNFα levels were determined in plasma and prefrontal cortex using ELISA and qPCR techniques, respectively. The precursor tryptophan (TRP), and its metabolites kynurenine (KYN) and serotonin (5-HT) were measured in hippocampus and cortex by HPLC. The ratios KYN/TRP and KYN/5-HT were used to estimate indoleamine 2,3-dioxygenase (IDO) activity and the balance of both metabolic pathways, respectively. CBD reduced the immobility time in the tail suspension test and increased sucrose preference in the LPS model, without affecting locomotion and central activity in the open-field test. CBD diminished cortical NF-ĸB activation, IL-6 levels in plasma and brain, and the increased KYN/TRP and KYN/5-HT ratios in hippocampus and cortex in the LPS model. Our results demonstrate that CBD produced antidepressant-like effects in the LPS neuroinflammatory model, associated to a reduction in the kynurenine pathway activation, IL-6 levels and NF-ĸB activation. As CBD stands out as a promising antidepressant drug, more research is needed to completely understand its mechanisms of action in depression linked to inflammation.


Subject(s)
Antidepressive Agents/therapeutic use , Cannabidiol/therapeutic use , Depression/drug therapy , Depression/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Animals , Antidepressive Agents/pharmacology , Cannabidiol/pharmacology , Depression/chemically induced , Hindlimb Suspension/adverse effects , Hindlimb Suspension/psychology , Inflammation Mediators/antagonists & inhibitors , Male , Mice
8.
Mol Neurobiol ; 57(3): 1704-1715, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31823197

ABSTRACT

We previously reported that the inactivation (cKO) or the stabilization (cST) of ß-catenin in cells expressing the astrocyte-specific glutamate aspartate transporter (GLAST) is associated with the vulnerability or resilience to exhibit anxious/depressive-like behaviors, respectively, and to changes in hippocampal proliferation. Here, we used these cKO and cST ß-catenin mice to study the serotonergic system functionality associated with their behavioral/molecular phenotype. The activity of 5-HT1A receptors was assessed by (+)-8-OH-DPAT-induced hypothermia and [35S]GTPγS binding autoradiography. The animals' response to acute stress and the levels of extracellular serotonin (5-HT) in the medial prefrontal cortex (mPFC) were also assessed. cKO mice presented higher 5-HT1A autoreceptor functionality, lower 5-HT1A heteroreceptor functionality, and a decrease in extracellular 5-HT levels in the mPFC. These neurochemical changes were accompanied with a blunted physiological response to stress-induced hyperthermia. In contrast, cST mice showed a reduced 5-HT1A autoreceptor functionality and higher extracellular 5-HT levels in the mPFC after fluoxetine administration. Moreover, cST mice subjected to chronic corticosterone administration did not show a blunted response to fluoxetine. Our findings suggest the existence of a link between ß-catenin levels and 5-HT1A receptor functionality, which may be relevant to understand the neurobiological bases underlying the vulnerability or resilience to stress-related disorders.


Subject(s)
Anxiety/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin/metabolism , beta Catenin/metabolism , Animals , Behavior, Animal/drug effects , Corticosterone/metabolism , Depression/metabolism , Hippocampus/metabolism , Male , Mice , Prefrontal Cortex/metabolism
9.
Transl Psychiatry ; 9(1): 306, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31740729

ABSTRACT

A better understanding of the molecular mechanisms that participate in the development and clinical manifestations of schizophrenia can lead to improve our ability to diagnose and treat this disease. Previous data strongly associated the levels of deregulated ADAMTS2 expression in peripheral blood mononuclear cells (PBMCs) from patients at first episode of psychosis (up) as well as in clinical responders to treatment with antipsychotic drugs (down). In this current work, we performed an independent validation of such data and studied the mechanisms implicated in the control of ADAMTS2 gene expression. Using a new cohort of drug-naïve schizophrenia patients with clinical follow-up, we confirmed that the expression of ADAMTS2 was highly upregulated in PBMCs at the onset (drug-naïve patients) and downregulated, in clinical responders, after treatment with antipsychotics. Mechanistically, ADAMTS2 expression was activated by dopaminergic signalling (D1-class receptors) and downstream by cAMP/CREB and mitogen-activated protein kinase (MAPK)/ERK signalling. Incubation with antipsychotic drugs and selective PKA and MEK inhibitors abrogated D1-mediated activation of ADAMTS2 in neuronal-like cells. Thus, D1 receptors signalling towards CREB activation might participate in the onset and clinical responses to therapy in schizophrenia patients, by controlling ADAMTS2 expression and activity. The unbiased investigation of molecular mechanisms triggered by antipsychotic drugs may provide a new landscape of novel targets potentially associated with clinical efficacy.


Subject(s)
ADAMTS Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dopamine/metabolism , Mitogen-Activated Protein Kinases/metabolism , Schizophrenia/physiopathology , 8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , ADAMTS Proteins/genetics , Animals , Antipsychotic Agents/pharmacology , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Humans , Leukocytes, Mononuclear/metabolism , Mice , Phosphorylation , Schizophrenia/genetics , Schizophrenia/metabolism , Signal Transduction
10.
Mol Neurobiol ; 56(1): 553-566, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29737454

ABSTRACT

ß-catenin (key mediator in the Wnt signaling pathway) contributes to the pathophysiology of mood disorders, associated to neurogenesis and neuroplasticity. Decreased ß-catenin protein levels have been observed in the hippocampus and prefrontal cortex of depressed subjects. Additionally, the antidepressants exert, at least in part, their neurogenic effects by increasing ß-catenin levels in the subgranular zone of the hippocampus. To further understand the role of ß-catenin in depression and anxiety, we generated two conditional transgenic mice in which ß-catenin was either inactivated or stabilized in cells expressing CreERT under the control of the astrocyte-specific glutamate transporter (GLAST) promoter inducible by tamoxifen, which presents high expression levels on the subgranular zone of the hippocampus. Here, we show that ß-catenin inactivation in GLAST-expressing cells enhanced anxious/depressive-like responses. These behavioral changes were associated with impaired hippocampal proliferation and markers of immature neurons as doublecortin. On the other hand, ß-catenin stabilization induced an anxiolytic-like effect in the novelty suppressed feeding test and tended to ameliorate depressive-related behaviors. In these mice, the control over the Wnt/ß-catenin pathway seems to be tighter as evidenced by the lack of changes in some proliferation markers. Moreover, animals with stabilized ß-catenin showed resilience to some anxious/depressive manifestations when subjected to the corticosterone model of depression. Our findings demonstrate that ß-catenin present in GLAST-expressing cells plays a critical role in the development of anxious/depressive-like behaviors and resilience, which parallels its regulatory function on hippocampal proliferation. Further studies need to be done to clarify the importance of these changes in other brain areas also implicated in the neurobiology of anxiety and depressive disorders.


Subject(s)
Anxiety/metabolism , Behavior, Animal , Depression/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Hippocampus/pathology , beta Catenin/metabolism , Animals , Anxiety/complications , Behavior, Animal/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Corticosterone/administration & dosage , Corticosterone/pharmacology , Dentate Gyrus/pathology , Depression/complications , Glial Fibrillary Acidic Protein/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Protein Stability/drug effects , SOXB1 Transcription Factors/metabolism
11.
Mol Neurobiol ; 56(6): 4135-4150, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30284123

ABSTRACT

Pain affects both sensory and emotional aversive responses, often provoking depression and anxiety-related conditions when it becomes chronic. As the opioid receptors in the locus coeruleus (LC) have been implicated in pain, stress responses, and opioid drug effects, we explored the modifications to LC opioid neurotransmission in a chronic constriction injury (CCI) model of short- and long-term neuropathic pain (7 and 30 days after nerve injury). No significant changes were found after short-term CCI, yet after 30 days, CCI provoked an up-regulation of cAMP (cyclic 5'-adenosine monophosphate), pCREB (phosphorylated cAMP response element binding protein), protein kinase A, tyrosine hydroxylase, and electrical activity in the LC, as well as enhanced c-Fos expression. Acute mu opioid receptor desensitization was more intense in these animals, measured as the decline of the peak current caused by [Met5]-enkephalin and the reduction of forskolin-stimulated cAMP produced in response to DAMGO. Sustained morphine treatment did not markedly modify certain LC parameters in CCI-30d animals, such as [Met5]-enkephalin-induced potassium outward currents or burst activity and c-Fos rebound after naloxone precipitation, which may limit the development of some typical opioid drug-related adaptations. However, other phenomena were impaired by long-term CCI, including the reduction in forskolin-stimulated cAMP accumulation by DAMGO after naloxone precipitation in morphine dependent animals. Overall, this study suggests that long-term CCI leads to changes at the LC level that may contribute to the anxiodepressive phenotype that develops in these animals. Furthermore, opioid drugs produce complex adaptations in the LC in this model of chronic neuropathic pain.


Subject(s)
Analgesics, Opioid/therapeutic use , Chronic Pain/drug therapy , Locus Coeruleus/pathology , Neuralgia/drug therapy , Analgesics, Opioid/pharmacology , Animals , Chronic Pain/physiopathology , Constriction, Pathologic , Locus Coeruleus/drug effects , Locus Coeruleus/physiopathology , Male , Models, Biological , Morphine/pharmacology , Morphine/therapeutic use , Naloxone/pharmacology , Naloxone/therapeutic use , Neuralgia/physiopathology , Pain Threshold/drug effects , Rats, Sprague-Dawley
12.
ACS Chem Neurosci ; 8(11): 2393-2401, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28777913

ABSTRACT

Postsynaptic 5-HT1A receptors (5-HT1AR) play an important role in anxiety and stress, although their contribution is still controversial. Previous studies report that mice overexpressing postsynaptic 5-HT1ARs show no changes in basal anxiety, though the influence of stress conditions has not been addressed yet. In this study, we used this animal model to evaluate the role of 5-HT1ARs in anxiety response after pre-exposure to an acute stressor. Under basal conditions, 5-HT1AR overexpressing animals presented high corticosterone levels and a lower mineralocorticoid/glucocorticoid receptor ratio. After pre-exposure to a single stressor, they showed a high anxiety-like response, associated with a blunted increase in corticosterone levels and higher c-Fos activation in the prefrontal cortex. Moreover, these mice also presented a lack of downregulation of hippocampal long-term potentiation after stress exposure. Therefore, higher postsynaptic 5-HT1AR activation might predispose to a high anxious phenotype and an impaired stress coping behavior.


Subject(s)
Hippocampus/physiopathology , Hypothalamo-Hypophyseal System/physiopathology , Long-Term Potentiation/physiology , Nerve Tissue Proteins/biosynthesis , Receptor, Serotonin, 5-HT1A/physiology , Stress, Psychological/physiopathology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Adaptation, Psychological/drug effects , Adaptation, Psychological/physiology , Animals , Anxiety/etiology , Anxiety/physiopathology , Corticosterone/metabolism , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hippocampus/drug effects , Hypothalamo-Hypophyseal System/drug effects , Long-Term Potentiation/drug effects , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Receptor, Serotonin, 5-HT1A/biosynthesis , Receptor, Serotonin, 5-HT1A/genetics , Receptors, Glucocorticoid/biosynthesis , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/biosynthesis , Receptors, Mineralocorticoid/genetics , Recombinant Proteins/metabolism , Serotonin 5-HT1 Receptor Agonists/pharmacology , Stress, Psychological/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...