Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167283, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38851305

ABSTRACT

Obesity represents a significant health challenge, intricately linked to conditions such as type II diabetes, metabolic syndrome, and hepatic steatosis. Several existing obesity treatments exhibit limited efficacy, undesirable side effects or a limited capability to maintain therapeutics effects in the long-term. Recently, modulation Coenzyme Q (CoQ) metabolism has emerged as a promising target for treatment of metabolic syndrome. This potential intervention could involve the modulation of endogenous CoQ biosynthesis by the use of analogs of the precursor of its biosynthesis, such as ß-resorcylic acid (ß-RA). Here, we show that oral supplementation with ß-RA, incorporated into the diet of diet-induced obese (DIO) mice, leads to substantial weight loss. The anti-obesity effects of ß-RA are partially elucidated through the normalization of mitochondrial CoQ metabolism in white adipose tissue (WAT). Additionally, we identify an HFN4α/LXR-dependent transcriptomic activation of the hepatic lipid metabolism that contributes to the anti-obesity effects of ß-RA. Consequently, ß-RA mitigates WAT hypertrophy, prevents hepatic steatosis, counteracts metabolic abnormalities in WAT and liver, and enhances glucose homeostasis by reducing the insulin/glucagon ratio and plasma levels of gastric inhibitory peptide (GIP). Moreover, pharmacokinetic evaluation of ß-RA supports its translational potential. Thus, ß-RA emerges as an efficient, safe, and translatable therapeutic option for the treatment and/or prevention of obesity, metabolic dysfunction-associated steatotic liver disease (MASLD).

2.
Cell Rep ; 43(5): 114148, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38697100

ABSTRACT

Coenzyme Q (CoQ) deficiency syndrome is conventionally treated with limited efficacy using exogenous CoQ10. Poor outcomes result from low absorption and bioavailability of CoQ10 and the clinical heterogenicity of the disease. Here, we demonstrate that supplementation with 4-hydroxybenzoic acid (4HB), the precursor of the benzoquinone ring in the CoQ biosynthetic pathway, completely rescues multisystemic disease and perinatal lethality in a mouse model of CoQ deficiency. 4HB stimulates endogenous CoQ biosynthesis in tissues of Coq2 mutant mice, normalizing mitochondrial function and rescuing cardiac insufficiency, edema, and neurodevelopmental delay. In contrast, exogenous CoQ10 supplementation falls short in fully restoring the phenotype. The treatment is translatable to human use, as proven by in vitro studies in skin fibroblasts from patients with pathogenic variants in COQ2. The therapeutic approach extends to other disorders characterized by deficiencies in the production of 4HB and early steps of CoQ biosynthesis and instances of secondary CoQ deficiency.


Subject(s)
Disease Models, Animal , Mitochondrial Diseases , Parabens , Ubiquinone , Animals , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/pathology , Mitochondrial Diseases/metabolism , Parabens/pharmacology , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Ubiquinone/metabolism , Ubiquinone/deficiency , Mice , Mitochondria/metabolism , Mitochondria/drug effects , Humans , Fibroblasts/metabolism , Fibroblasts/drug effects , Mice, Inbred C57BL , Muscle Weakness/drug therapy , Muscle Weakness/metabolism , Muscle Weakness/pathology , Ataxia/drug therapy , Ataxia/pathology , Ataxia/metabolism
3.
Front Pharmacol ; 13: 925740, 2022.
Article in English | MEDLINE | ID: mdl-35924056

ABSTRACT

Background: Hypoxic-ischemic (HI) insults have important deleterious consequences in newborns, including short-term morbidity with neuromotor and cognitive disturbances. Cannabidiol (CBD) has demonstrated robust neuroprotective effects and shows anxiolytic/antidepressant effects as well. These effects are thought to be related to serotonin 5-HT1A receptor (5HT1AR) activation. We hereby aimed to study the role of 5HT1AR in the neuroprotective and behavioral effects of CBD in HI newborn piglets. Methods: 1-day-old piglets submitted to 30 min of hypoxia (FiO2 10%) and bilateral carotid occlusion were then treated daily with vehicle, CBD 1 mg/kg, or CBD with the 5HT1AR antagonist WAY 100635 1 mg/kg 72 h post-HI piglets were studied using amplitude-integrated EEG to detect seizures and a neurobehavioral test to detect neuromotor impairments. In addition, behavioral performance including social interaction, playful activity, hyperlocomotion, and motionless periods was assessed. Then, brain damage was assessed using histology (Nissl and TUNEL staining) and biochemistry (proton magnetic resonance spectroscopy studies. Results: HI led to brain damage as assessed by histologic and biochemistry studies, associated with neuromotor impairment and increased seizures. These effects were not observed in HI piglets treated with CBD. These beneficial effects of CBD were not reversed by the 5HT1AR antagonist, which is in contrast with previous studies demonstrating that 5HT1AR antagonists eliminated CBD neuroprotection as assessed 6 h after HI in piglets. HI led to mood disturbances, with decreased social interaction and playfulness and increased hyperlocomotion. Mood disturbances were not observed in piglets treated with CBD, but in this case, coadministration of the 5HT1AR antagonist eliminates the beneficial effects of CBD. Conclusion: CBD prevented HI-induced mood disturbances in newborn piglets by acting on 5HT1AR. However, 5HT1AR activation seems to be necessary for CBD neuroprotection only in the first hours after HI.

4.
Redox Biol ; 55: 102403, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35863266

ABSTRACT

Defects in Coenzyme Q (CoQ) metabolism have been associated with primary mitochondrial disorders, neurodegenerative diseases and metabolic conditions. The consequences of CoQ deficiency have not been fully addressed, and effective treatment remains challenging. Here, we use mice with primary CoQ deficiency (Coq9R239X), and we demonstrate that CoQ deficiency profoundly alters the Q-junction, leading to extensive changes in the mitochondrial proteome and metabolism in the kidneys and, to a lesser extent, in the brain. CoQ deficiency also induces reactive gliosis, which mediates a neuroinflammatory response, both of which lead to an encephalopathic phenotype. Importantly, treatment with either vanillic acid (VA) or ß-resorcylic acid (ß-RA), two analogs of the natural precursor for CoQ biosynthesis, partially restores CoQ metabolism, particularly in the kidneys, and induces profound normalization of the mitochondrial proteome and metabolism, ultimately leading to reductions in gliosis, neuroinflammation and spongiosis and, consequently, reversing the phenotype. Together, these results provide key mechanistic insights into defects in CoQ metabolism and identify potential disease biomarkers. Furthermore, our findings clearly indicate that the use of analogs of the CoQ biosynthetic precursor is a promising alternative therapy for primary CoQ deficiency and has potential for use in the treatment of more common neurodegenerative and metabolic diseases that are associated with secondary CoQ deficiency.

6.
Front Pharmacol ; 10: 1131, 2019.
Article in English | MEDLINE | ID: mdl-31611802

ABSTRACT

Neonatal hypoxia-ischemia (HI) is a risk factor for myelination disturbances, a key factor for cerebral palsy. Cannabidiol (CBD) protects neurons and glial cells after HI insult in newborn animals. We hereby aimed to study CBD's effects on long-lasting HI-induced myelination deficits in newborn rats. Thus, P7 Wistar rats received s.c. vehicle (HV) or cannabidiol (HC) after HI brain damage (left carotid artery electrocoagulation plus 10% O2 for 112 min). Controls were non-HI pups. At P37, neurobehavioral tests were performed and immunohistochemistry [quantifying mature oligodendrocyte (mOL) populations and myelin basic protein (MBP) density] and electron microscopy (determining axon number, size, and myelin thickness) studies were conducted in cortex (CX) and white matter (WM). Expression of brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF) were analyzed by western blot at P14. HI reduced mOL or MBP in CX but not in WM. In both CX and WM, axon density and myelin thickness were reduced. MBP impairment correlated with functional deficits. CBD administration resulted in normal function associated with normal mOL and MBP, as well as normal axon density and myelin thickness in all areas. CBD's effects were not associated with increased BDNF or GDNF expression. In conclusion, HI injury in newborn rats resulted in long-lasting myelination disturbance, associated with functional impairment. CBD treatment preserved function and myelination, likely as a part of a general neuroprotective effect.

7.
Respir Res ; 20(1): 130, 2019 Jun 24.
Article in English | MEDLINE | ID: mdl-31234835

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is an age-related, progressive and lethal disease, whose pathogenesis is associated with fibroblasts/myofibroblasts foci that produce excessive extracellular matrix accumulation in lung parenchyma. Hypoxia has been described as a determinant factor in its development and progression. However, the role of distinct members of this pathway is not completely described. METHODS: By western blot, quantitative PCR, Immunohistochemistry and Immunocitochemistry were evaluated, the expression HIF alpha subunit isoforms 1, 2 & 3 as well, as their role in myofibroblast differentiation in lung tissue and fibroblast cell lines derived from IPF patients. RESULTS: Hypoxia signaling pathway was found very active in lungs and fibroblasts from IPF patients, as demonstrated by the abundance of alpha subunits 1 and 2, which further correlated with the increased expression of myofibroblast marker αSMA. In contrast, HIF-3α showed reduced expression associated with its promoter hypermethylation. CONCLUSIONS: This study lends further support to the involvement of hypoxia in the pathogenesis of IPF, and poses HIF-3α expression as a potential negative regulator of these phenomena.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Idiopathic Pulmonary Fibrosis/metabolism , Myofibroblasts/metabolism , Repressor Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Cell Line , Gene Expression , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Myofibroblasts/pathology , Repressor Proteins/genetics
8.
ACS Chem Neurosci ; 10(7): 3318-3326, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31244055

ABSTRACT

Depression is a chronic and debilitating illness that interferes severely with many human behaviors, and is the leading cause of disability in the world. There is data suggesting that deficits in serotonin neurotransmission can contribute to the development of depression. Indeed, >90% of prescribed antidepressant drugs act by increasing serotonergic transmission at the synapse. However, this increase is offset by a negative feedback operating at the level of the cell body of the serotonin neurons in the raphe nuclei. In the present work, we demonstrate: first, the intracortical infusion of ketamine induced an antidepressant-like effect in the forced swim test, comparable to that produced by systemic ketamine; second, systemic and intracortical ketamine increased serotonin and noradrenaline efflux in the prefrontal cortex, but not in the dorsal raphe nucleus; third, systemic and intracortical administration of ketamine increased the efflux of glutamate in the prefrontal cortex and dorsal raphe nucleus; fourth, systemic ketamine did not alter the functionality of 5-HT1A receptors in the dorsal raphe nucleus. Taken together, these findings suggest that the antidepressant-like effects of ketamine are caused by the stimulation of the prefrontal projection to the dorsal raphe nucleus and locus coeruleus caused by an elevated glutamate in the medial prefrontal cortex, which would stimulate release of serotonin and noradrenaline in the same area. The impact of both monoamines in the antidepressant response to ketamine seems to have different time frames.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Dorsal Raphe Nucleus/drug effects , Ketamine/pharmacology , Norepinephrine/metabolism , Serotonergic Neurons/drug effects , Serotonin/metabolism , Animals , Antidepressive Agents/therapeutic use , Depression/metabolism , Dorsal Raphe Nucleus/metabolism , Glutamic Acid/metabolism , Ketamine/therapeutic use , Male , Motor Activity/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1A/metabolism , Serotonergic Neurons/metabolism
9.
Pediatr Res ; 85(4): 539-545, 2019 03.
Article in English | MEDLINE | ID: mdl-30675020

ABSTRACT

BACKGROUND: Newborn pigs offer theoretical advantages for studying newborn hypoxic-ischemic (HI) brain damage because of a development and structure similar to the human brain. However, the correlation between functional features and actual HI brain damage has not been reported. METHODS: Newborn pigs were examined daily for 3 days after a HI insult using amplitude-integrated EEG (aEEG), and a neurobehavioral score enriched with stress and social and object interaction-driven activity evaluation. Brain damage was then assessed using histologic, immunohistochemical, and proton magnetic resonance spectroscopy studies. Brain concentration of several neurotransmitters was determined by HPLC. RESULTS: HI insult led to aEEG amplitude decrease, muscle tone and activity impairment, eating disorders, poor environmental interaction, and increased motionless periods. Basal aEEG amplitude, muscle tone, and general behavior were the best predictive items for histological and biochemical (lactate/N-acetylaspartate ratio) brain damage. Hyperexcitable response to stress correlated inversely with brain damage. Motionless time, which correlated with brain damage severity, was inversely related to brain concentration of dopamine and norepinephrine. CONCLUSION: Standard neurologic examination of brain activity and motor and behavioral performance of newborn pigs is a valuable tool to assess HI brain damage, thus offering a powerful translational model for HI brain damage pathophysiology and management studies.


Subject(s)
Electroencephalography/methods , Hypoxia-Ischemia, Brain/pathology , Neurologic Examination , Animals , Hypoxia-Ischemia, Brain/physiopathology , Severity of Illness Index , Survival Analysis , Swine
10.
Neuropharmacology ; 116: 151-159, 2017 04.
Article in English | MEDLINE | ID: mdl-28012949

ABSTRACT

BACKGROUND: and purpose: Currently there is no effective treatment for neonatal arterial ischemic stroke (AIS). Cannabidiol (CBD) is neuroprotective in models of newborn hypoxic-ischemic brain damage and adult stroke. The purpose of this work was to study the protective effect of CBD in a neonatal rat model of AIS. METHODS: Middle Cerebral Artery Occlusion (MCAO) was achieved in neonatal Wistar rats by introducing a nylon filament to the left MCA for 3 h; 15 min after removing the occluder vehicle (MCAO-V) or CBD single dose 5 mg/kg (MCAO-C) were administered i. p. Similarly manipulated but non-occluded rats served as controls (SHM). A set of behavioral tests was then conducted one week (P15) or one month (P38) after MCAO. Brain damage was then assessed by magnetic resonance imaging (MRI), proton magnetic resonance spectroscopy (H+-MRS) and histologic (TUNEL for cell death, immunohistochemistry for neuron, astrocyte and microglia identification) studies. RESULTS: CBD administration improved neurobehavioral function regarding strength, hemiparesis, coordination and sensorimotor performance as assessed at P15 and P38. MRI indicated that CBD did not reduce the volume of infarct but reduced the volume of perilesional gliosis. H+-MRS indicated that CBD reduced metabolic derangement and excitotoxicty, and protected astrocyte function. Histologic studies indicated that CBD reduced neuronal loss and apoptosis, and modulated astrogliosis and microglial proliferation and activation. CONCLUSIONS: CBD administration after MCAO led to long-term functional recovery, reducing neuronal loss and astrogliosis, and modulating apoptosis, metabolic derangement, excitotoxicity and neuro-inflammation.


Subject(s)
Brain Ischemia/drug therapy , Brain/drug effects , Cannabidiol/pharmacology , Neuroprotective Agents/pharmacology , Stroke/drug therapy , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/pathology , Astrocytes/physiology , Brain/diagnostic imaging , Brain/pathology , Brain/physiopathology , Brain Ischemia/diagnostic imaging , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Disease Models, Animal , Disease Progression , Microglia/drug effects , Microglia/pathology , Microglia/physiology , Motor Activity/drug effects , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Random Allocation , Rats, Wistar , Recovery of Function/drug effects , Recovery of Function/physiology , Stroke/diagnostic imaging , Stroke/pathology , Stroke/physiopathology , Time Factors
11.
Neuropharmacology ; 108: 91-102, 2016 09.
Article in English | MEDLINE | ID: mdl-27108934

ABSTRACT

Deep brain stimulation (DBS) is a treatment that has shown some efficacy in treatment-resistant depression. In particular, DBS of the subcallosal cingulate gyrus (Brodmann's area 25, Cg25) has been successfully applied to treat refractory depression. In the rat, we have demonstrated that DBS applied to infralimbic (IL) cortex elevates the levels of glutamate and monoamines in the prefrontal cortex, and requires the stimulation of cortical α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptors for its antidepressant-like effects. However, the molecular targets of IL DBS are not fully known. To gain insight into these pathways, we have investigated whether IL DBS is able to reverse the behavioral, biochemical and molecular changes exhibited by the olfactory bulbectomized (OBX) rat. Our results revealed that 1 h IL DBS diminished hyperlocomotion, hyperemotionality and anhedonia, and increased social interaction shown by the OBX rats. Further, IL DBS increased prefrontal efflux of glutamate and serotonin in both sham-operated and OBX rats. With regard to molecular targets, IL DBS increases the synthesis of brain-derived neurotrophic factor (BDNF) and the GluA1 AMPA receptor subunit, and stimulates the Akt/mammalian target of rapamycin (mTOR) as well as the AMPA receptor/c-AMP response element binding (CREB) pathways. Temsirolimus, a known in vivo mTOR blocker, suppressed the antidepressant-like effect of IL DBS in naïve rats in the forced swim test, thus demonstrating for the first time that mTOR signaling is required for the antidepressant-like effects of IL DBS, which is in line with the antidepressant response of other rapid-acting antidepressant drugs.


Subject(s)
Deep Brain Stimulation/methods , Depression/metabolism , Interpersonal Relations , Prefrontal Cortex/chemistry , Prefrontal Cortex/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Depression/surgery , Male , Olfactory Bulb/chemistry , Olfactory Bulb/metabolism , Olfactory Bulb/surgery , Prefrontal Cortex/surgery , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar
12.
Neuropharmacology ; 103: 16-26, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26711860

ABSTRACT

Cannabidiol (CBD), the main non-psychotomimetic component of marihuana, exhibits anxiolytic-like properties in many behavioural tests, although its potential for treating major depression has been poorly explored. Moreover, the mechanism of action of CBD remains unclear. Herein, we have evaluated the effects of CBD following acute and chronic administration in the olfactory bulbectomy mouse model of depression (OBX), and investigated the underlying mechanism. For this purpose, we conducted behavioural (open field and sucrose preference tests) and neurochemical (microdialysis and autoradiography of 5-HT1A receptor functionality) studies following treatment with CBD. We also assayed the pharmacological antagonism of the effects of CBD to dissect out the mechanism of action. Our results demonstrate that CBD exerts fast and maintained antidepressant-like effects as evidenced by the reversal of the OBX-induced hyperactivity and anhedonia. In vivo microdialysis revealed that the administration of CBD significantly enhanced serotonin and glutamate levels in vmPFCx in a different manner depending on the emotional state and the duration of the treatment. The potentiating effect upon neurotransmitters levels occurring immediately after the first injection of CBD might underlie the fast antidepressant-like actions in OBX mice. Both antidepressant-like effect and enhanced cortical 5-HT/glutamate neurotransmission induced by CBD were prevented by 5-HT1A receptor blockade. Moreover, adaptive changes in pre- and post-synaptic 5-HT1A receptor functionality were also found after chronic CBD. In conclusion, our findings indicate that CBD could represent a novel fast antidepressant drug, via enhancing both serotonergic and glutamate cortical signalling through a 5-HT1A receptor-dependent mechanism.


Subject(s)
Anti-Anxiety Agents/administration & dosage , Antidepressive Agents/administration & dosage , Cannabidiol/administration & dosage , Depressive Disorder/metabolism , Glutamic Acid/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin/metabolism , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Olfactory Bulb/surgery , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Synaptic Transmission/drug effects
13.
Cereb Cortex ; 26(6): 2778-2789, 2016 06.
Article in English | MEDLINE | ID: mdl-26088969

ABSTRACT

Although deep brain stimulation (DBS) has been used with success in treatment-resistant depression, little is known about its mechanism of action. We examined the antidepressant-like activity of short (1 h) DBS applied to the infralimbic prefrontal cortex in the forced swim test (FST) and the novelty-suppressed feeding test (NSFT). We also used in vivo microdialysis to evaluate the release of glutamate, γ-aminobutyric acid, serotonin, dopamine, and noradrenaline in the prefrontal cortex and c-Fos immunohistochemistry to determine the brain regions activated by DBS. One hour of DBS of the infralimbic prefrontal cortex has antidepressant-like effects in FST and NSFT, and increases prefrontal efflux of glutamate, which would activate AMPA receptors (AMPARs). This effect is specific of the infralimbic area since it is not observed after DBS of the prelimbic subregion. The activation of prefrontal AMPARs would result in a stimulation of prefrontal output to the brainstem, thus increasing serotonin, dopamine, and noradrenaline in the prefrontal cortex. Further, the activation of prefrontal AMPARs is necessary and sufficient condition for the antidepressant response of 1 h DBS.


Subject(s)
Deep Brain Stimulation/methods , Depressive Disorder/metabolism , Depressive Disorder/therapy , Prefrontal Cortex/metabolism , Receptors, AMPA/metabolism , Animals , Depressive Disorder/pathology , Disease Models, Animal , Dopamine/metabolism , Glutamic Acid/metabolism , Immunohistochemistry , Male , Microdialysis , Norepinephrine/metabolism , Prefrontal Cortex/pathology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Wistar , Serotonin/metabolism , gamma-Aminobutyric Acid/metabolism
14.
Neuropsychopharmacology ; 39(11): 2673-80, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24871546

ABSTRACT

Paradoxically, N-methyl-D-aspartate (NMDA) receptor antagonists are used to model certain aspects of schizophrenia as well as to treat refractory depression. However, the role of different subunits of the NMDA receptor in both conditions is poorly understood. Here we used biochemical and behavioral readouts to examine the in vivo prefrontal efflux of serotonin and glutamate as well as the stereotypical behavior and the antidepressant-like activity in the forced swim test elicited by antagonists selective for the GluN2A (NVP-AAM077) and GluN2B (Ro 25-6981) subunits. The effects of the non-subunit selective antagonist, MK-801; were also studied for comparison. The administration of MK-801 dose dependently increased the prefrontal efflux of serotonin and glutamate and markedly increased the stereotypy scores. NVP-AAM077 also increased the efflux of serotonin and glutamate, but without the induction of stereotypies. In contrast, Ro 25-6981 did not change any of the biochemical and behavioral parameters tested. Interestingly, the administration of NVP-AAM077 and Ro 25-6981 alone elicited antidepressant-like activity in the forced swim test, in contrast to the combination of both compounds that evoked marked stereotypies. Our interpretation of the results is that both GluN2A and GluN2B subunits are needed to induce stereotypies, which might be suggestive of potential psychotomimetic effects in humans, but the antagonism of only one of these subunits is sufficient to evoke an antidepressant response. We also propose that GluN2A receptor antagonists could have potential antidepressant activity in the absence of potential psychotomimetic effects.


Subject(s)
Depressive Disorder, Treatment-Resistant/drug therapy , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia , Animals , Antidepressive Agents/pharmacology , Depressive Disorder, Treatment-Resistant/physiopathology , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Glutamic Acid/metabolism , Male , Phenols/pharmacology , Piperidines/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Quinoxalines/pharmacology , Rats, Wistar , Schizophrenia/physiopathology , Serotonin/metabolism , Stereotyped Behavior/drug effects , Stereotyped Behavior/physiology
15.
Neuropharmacology ; 79: 49-58, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24211653

ABSTRACT

The prefrontal cortex (PFC) is involved in higher brain functions altered in schizophrenia. Classical antipsychotic drugs modulate information processing in cortico-limbic circuits via dopamine D2 receptor blockade in nucleus accumbens (NAc) whereas atypical antipsychotic drugs preferentially target cortical serotonin (5-HT) receptors. The brain networks involved in the therapeutic action of atypical drugs are not fully understood. Previous work indicated that medial PFC (mPFC) pyramidal neurons projecting to ventral tegmental area express 5-HT2A receptors suggesting that atypical antipsychotic drugs modulate dopaminergic activity distally, via 5-HT2A receptor (5-HT2A-R) blockade in PFC. Since the mPFC also projects heavily to NAc, we examined whether NAc-projecting pyramidal neurons also express 5-HT2A-R. Using a combination of retrograde tracing experiments and in situ hybridization we report that a substantial proportion of mPFC-NAc pyramidal neurons in rat brain express 5-HT2A-R mRNA in a layer- and area-specific manner (up to 68% in layer V of contralateral cingulate). The functional relevance of 5-HT2A-R to modulate mPFC-NAc projections was examined in dual-probe microdialysis experiments. The application of the preferential 5-HT2A-R agonist DOI into mPFC enhanced glutamate release locally (+66 ± 18%) and in NAc (+74 ± 12%) indicating that cortical 5-HT2A-R activation augments glutamatergic transmission in NAc. Since NAc integrates glutamatergic and dopaminergic inputs, blockade of 5-HT2A-R by atypical drugs may reduce cortical excitatory inputs onto GABAergic neurons of NAc, adding to dopamine D2 receptor blockade. Together with previous observations, the present results suggest that atypical antipsychotic drugs may control the activity of the mesolimbic pathway at cell body and terminal level.


Subject(s)
Antipsychotic Agents/pharmacology , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Amphetamines/pharmacology , Animals , Glutamic Acid/metabolism , In Situ Hybridization , Male , Microdialysis , Neural Pathways/anatomy & histology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neuroanatomical Tract-Tracing Techniques , Nucleus Accumbens/anatomy & histology , Nucleus Accumbens/drug effects , Prefrontal Cortex/anatomy & histology , Prefrontal Cortex/drug effects , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , RNA, Messenger , Rats , Rats, Wistar , Serotonin 5-HT2 Receptor Agonists/pharmacology
16.
Schizophr Bull ; 38(1): 9-14, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21965469

ABSTRACT

Several genetic, neurodevelopmental, and pharmacological animal models of schizophrenia have been established. This short review examines the validity of one of the most used pharmacological model of the illness, ie, the acute administration of N-methyl-D-aspartate (NMDA) receptor antagonists in rodents. In some cases, data on chronic or prenatal NMDA receptor antagonist exposure have been introduced for comparison. The face validity of acute NMDA receptor blockade is granted inasmuch as hyperlocomotion and stereotypies induced by phencyclidine, ketamine, and MK-801 are regarded as a surrogate for the positive symptoms of schizophrenia. In addition, the loss of parvalbumin-containing cells (which is one of the most compelling finding in postmortem schizophrenia brain) following NMDA receptor blockade adds construct validity to this model. However, the lack of changes in glutamic acid decarboxylase (GAD(67)) is at variance with human studies. It is possible that changes in GAD(67) are more reflective of the neurodevelopmental condition of schizophrenia. Finally, the model also has predictive validity, in that its behavioral and transmitter activation in rodents are responsive to antipsychotic treatment. Overall, although not devoid of drawbacks, the acute administration of NMDA receptor antagonists can be considered as a good model of schizophrenia bearing a satisfactory degree of validity.


Subject(s)
Disease Models, Animal , Ketamine/pharmacology , Phencyclidine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Schizophrenia/physiopathology , Animals , Corpus Callosum/physiopathology , Dopamine/pharmacology , Glutamic Acid/pharmacology , Humans , Parvalbumins/pharmacology , Receptor, Serotonin, 5-HT2A/drug effects , Rodentia , Serotonin/pharmacology
17.
Int J Neuropsychopharmacol ; 15(7): 945-56, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21733285

ABSTRACT

Previous studies have shown that systemic, but not unilateral intra-prefrontal cortex administration of non-competitive NMDA antagonists, increased prefrontal activity, the cortical efflux of serotonin, and induced stereotypies. In this work we used in-vivo microdialysis and immunohistochemistry to test the hypothesis as to whether MK-801 and ketamine need to act on both prefrontal cortices to reproduce these neurochemical and behavioural changes. Dialysis probes were implanted in the medial prefrontal cortex, and extracellular serotonin as well as behavioural stereotypies was measured after systemic administration of MK-801 and ketamine (1 mg/kg and 25 mg/kg, respectively), and unilateral and bilateral perfusion of both drugs (300 µm and 3 mm, respectively). Additionally, the prefrontal (glutamatergic) level of activity was measured using c-Fos immunohistochemistry. Systemic and bilateral (but not unilateral) prefrontal administration of MK-801 and ketamine increased serotonin efflux whereas only systemic administration of both drugs produced hyperlocomotion and stereotypies. The unilateral perfusion of 1 µm tetrodotoxin in the medial prefrontal cortex reduced increases of serotonin in both hemispheres, the expression of c-Fos in the contralateral side, and stereotypy scores after systemic NMDA antagonists. Our results support the hypothesis that a bilateral impairment of cortical inhibition in the medial prefrontal cortex is needed for non-competitive NMDA antagonists to induce the state of pyramidal cell hyperactivity and concurrent efflux of serotonin. Furthermore, hyperlocomotion and stereotypies produced by MK-801 and ketamine do not appear to result from changes in the activity of prefrontal cortex although this structure exerts some control over these behaviours.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Functional Laterality/drug effects , Neural Pathways/drug effects , Prefrontal Cortex/drug effects , Analysis of Variance , Anesthetics, Local/pharmacology , Animals , Dizocilpine Maleate/pharmacology , Drug Administration Routes , Gene Expression Regulation/drug effects , Ketamine/pharmacology , Male , Microdialysis , Neural Pathways/physiology , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Serotonin/metabolism , Stereotyped Behavior/drug effects , Tetrodotoxin/pharmacology , Vesicular Glutamate Transport Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...