Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Transl Psychiatry ; 14(1): 14, 2024 Jan 08.
Article En | MEDLINE | ID: mdl-38191622

Schizophrenia is a chronic neurodevelopmental disorder with an inflammatory/prooxidant component. N-acetylcysteine (NAC) has been evaluated in schizophrenia as an adjuvant to antipsychotics, but its role as a preventive strategy has not been sufficiently explored. We aimed to evaluate the potential of NAC administration in two-time windows before the onset of symptoms in a schizophrenia-like maternal immune stimulation (MIS) rat model. Pregnant Wistar rats were injected with Poly I:C or Saline on gestational day (GD) 15. Three different preventive approaches were evaluated: 1) NAC treatment during periadolescence in the offspring (from postnatal day [PND] 35 to 49); 2) NAC treatment during pregnancy after MIS challenge until delivery (GD15-21); and 3) NAC treatment throughout all pregnancy (GD1-21). At postnatal day (PND) 70, prepulse inhibition (PPI) and anxiety levels were evaluated. In vivo magnetic resonance (MR) imaging was acquired on PND100 to assess structural changes in gray and white matter, and brain metabolite concentrations. Additionally, inflammation and oxidative stress (IOS) markers were measured ex vivo in selected brain regions. MIS offspring showed behavioral, neuroanatomical, and biochemical alterations. Interestingly, NAC treatment during periadolescence prevented PPI deficits and partially counteracted some biochemical imbalances. Moreover, NAC treatments during pregnancy not only replicated the beneficial outcomes reported by the treatment in periadolescence, but also prevented some neuroanatomical deficits, including reductions in hippocampal and corpus callosum volumes. This study suggests that early reduction of inflammation and prooxidation could help prevent the onset of schizophrenia-like symptoms, supporting the importance of anti-IOS compounds in ameliorating this disorder.


Acetylcysteine , Schizophrenia , Female , Pregnancy , Rats , Animals , Rats, Wistar , Acetylcysteine/pharmacology , Schizophrenia/drug therapy , Schizophrenia/prevention & control , Poly I-C , Inflammation
2.
Psychiatry Res ; 331: 115643, 2024 Jan.
Article En | MEDLINE | ID: mdl-38064909

Prenatal infections and cannabis use during adolescence are well-recognized risk factors for schizophrenia. As inflammation and oxidative stress (OS) contribute to this disorder, anti-inflammatory drugs have been proposed as potential therapies. This study aimed to evaluate the association between delta-9-tetrahydrocannabinol (THC) and schizophrenia-like abnormalities in a maternal immune activation (MIA) model. Additionally, we assessed the preventive effect of cannabidiol (CBD), a non-psychotropic/anti-inflammatory cannabinoid. THC and/or CBD were administered to Saline- and MIA-offspring during periadolescence. At adulthood, THC-exposed MIA-offspring showed significant improvements in sensorimotor gating deficits. Structural and metabolic brain changes were evaluated by magnetic resonance imaging, revealing cortical shrinkage in Saline- and enlargement in MIA-offspring after THC-exposure. Additionally, MIA-offspring displayed enlarged ventricles and decreased hippocampus, which were partially reverted by both cannabinoids. CBD prevented THC-induced reduction in the corpus callosum, despite affecting white matter structure. Post-mortem studies revealed detrimental effects of THC, including increased inflammation and oxidative stress. CBD partially reverted these pro-inflammatory alterations and modulated THC's effects on the endocannabinoid system. In conclusion, contrary to expectations, THC exhibited greater behavioural and morphometric benefits, despite promoting a pro-inflammatory state that CBD partially reverted. Further research is needed to elucidate the underlying mechanisms involved in the observed benefits of THC.


Cannabidiol , Cannabinoids , Cannabis , Schizophrenia , Humans , Pregnancy , Female , Adult , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Schizophrenia/drug therapy , Schizophrenia/metabolism , Dronabinol/pharmacology , Poly I-C , Inflammation , Anti-Inflammatory Agents
3.
J Neuroinflammation ; 20(1): 198, 2023 Sep 01.
Article En | MEDLINE | ID: mdl-37658434

BACKGROUND: Most current disease-modifying therapies approved for multiple sclerosis (MS) are immunomodulatory drugs that counteract the aberrant activity of the immune system. Hence, new pharmacological interventions that drive anti-inflammatory activity and neuroprotection would represent interesting alternative therapeutic approaches or complementary strategies to treat progressive forms of MS. There is evidence of reduced noradrenaline levels and alterations to locus coeruleus (LC) noradrenergic neurons in MS patients, as well as in animal models of this disease, potentially factors contributing to the pathophysiology. Drugs that enhance noradrenaline appear to have some beneficial effects in MS, suggesting their potential to dampen the underlying pathology and disease progression. METHODS: Therefore, we explored the consequences of chronic LC noradrenergic neurons activation by chemogenetics in experimental autoimmune encephalomyelitis (EAE) mice, the most widely used experimental model of MS. LC activation from the onset or the peak of motor symptoms was explored as two different therapeutic approaches, assessing the motor and non-motor behavioral changes as EAE progresses, and studying demyelination, inflammation and glial activation in the spinal cord and cerebral cortex during the chronic phase of EAE. RESULTS: LC activation from the onset of motor symptoms markedly alleviated the motor deficits in EAE mice, as well as their anxiety-like behavior and sickness, in conjunction with reduced demyelination and perivascular infiltration in the spinal cord and glial activation in the spinal cord and prefrontal cortex (PFC). When animals exhibited severe paralysis, LC activation produced a modest alleviation of EAE motor symptoms and it enhanced animal well-being, in association with an improvement of the EAE pathology at the spinal cord and PFC level. Interestingly, the reduced dopamine beta-hydroxylase expression associated with EAE in the spinal cord and PFC was reversed through chemogenetic LC activation. CONCLUSION: Therefore, clear anti-inflammatory and neuroprotective effects were produced by the selective activation of LC noradrenergic neurons in EAE mice, having greater benefits when LC activation commenced earlier. Overall, these data suggest noradrenergic LC neurons may be targets to potentially alleviate some of the motor and non-motor symptoms in MS.


Adrenergic Neurons , Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Mice , Locus Coeruleus , Norepinephrine
4.
Front Pharmacol ; 13: 886514, 2022.
Article En | MEDLINE | ID: mdl-35959428

There is evidence that in schizophrenia, imbalances in inflammatory and oxidative processes occur during pregnancy and in the early postnatal period, generating interest in the potential therapeutic efficacy of anti-inflammatory and antioxidant compounds. Mangiferin is a polyphenolic compound abundant in the leaves of Mangifera indica L. that has robust antioxidant and anti-inflammatory properties, making it a potential candidate for preventive or co-adjuvant therapy in schizophrenia. Hence, this study set-out to evaluate the effect of mango leaf extract (MLE) in a model of schizophrenia based on maternal immune activation, in which Poly I:C (4 mg/kg) is administered intravenously to pregnant rats. Young adult (postnatal day 60-70) or adolescent (postnatal day 35-49) male offspring received MLE (50 mg/kg of mangiferin) daily, and the effects of MLE in adolescence were compared to those of risperidone, assessing behavior, brain magnetic resonance imaging (MRI), and oxidative/inflammatory and antioxidant mediators in the adult offspring. MLE treatment in adulthood reversed the deficit in prepulse inhibition (PPI) but it failed to attenuate the sensitivity to amphetamine and the deficit in novel object recognition (NOR) induced. By contrast, adolescent MLE treatment prevented the sensorimotor gating deficit in the PPI test, producing an effect similar to that of risperidone. This MLE treatment also produced a reduction in grooming behavior, but it had no effect on anxiety or novel object recognition memory. MRI studies revealed that adolescent MLE administration partially counteracted the cortical shrinkage, and cerebellum and ventricle enlargement. In addition, MLE administration in adolescence reduced iNOS mediated inflammatory activation and it promoted the expression of biomarkers of compensatory antioxidant activity in the prefrontal cortex and hippocampus, as witnessed through the reduction of Keap1 and the accumulation of NRF2 and HO1. Together, these findings suggest that MLE might be an alternative therapeutic or preventive add-on strategy to improve the clinical expression of schizophrenia in adulthood, while also modifying the time course of this disease at earlier stages in populations at high-risk.

5.
Sensors (Basel) ; 21(21)2021 Oct 26.
Article En | MEDLINE | ID: mdl-34770410

Strong evidence from studies on primates and rodents shows that changes in pupil diameter may reflect neural activity in the locus coeruleus (LC). Pupillometry is the only available non-invasive technique that could be used as a reliable and easily accessible real-time biomarker of changes in the in vivo activity of the LC. However, the application of pupillometry to preclinical research in rodents is not yet fully standardized. A lack of consensus on the technical specifications of some of the components used for image recording or positioning of the animal and cameras have been recorded in recent scientific literature. In this study, a novel pupillometry system to indirectly assess, in real-time, the function of the LC in anesthetized rodents is presented. The system comprises a deep learning SOLOv2 instance-based fast segmentation framework and a platform designed to place the experimental subject, the video cameras for data acquisition, and the light source. The performance of the proposed setup was assessed and compared to other baseline methods using a validation and an external test set. In the latter, the calculated intersection over the union was 0.93 and the mean absolute percentage error was 1.89% for the selected method. The Bland-Altman analysis depicted an excellent agreement. The results confirmed a high accuracy that makes the system suitable for real-time pupil size tracking, regardless of the pupil's size, light intensity, or any features typical of the recording process in sedated mice. The framework could be used in any neurophysiological study with sedated or fixed-head animals.


Deep Learning , Locus Coeruleus , Animals , Light , Mice , Pupil
6.
Int J Neuropsychopharmacol ; 24(9): 734-748, 2021 09 21.
Article En | MEDLINE | ID: mdl-34165516

BACKGROUND: Minocycline (MIN) is a tetracycline with antioxidant, anti-inflammatory, and neuroprotective properties. Given the likely involvement of inflammation and oxidative stress (IOS) in schizophrenia, MIN has been proposed as a potential adjuvant treatment in this pathology. We tested an early therapeutic window, during adolescence, as prevention of the schizophrenia-related deficits in the maternal immune stimulation (MIS) animal model. METHODS: On gestational day 15, Poly I:C or vehicle was injected in pregnant Wistar rats. A total 93 male offspring received MIN (30 mg/kg) or saline from postnatal day (PND) 35-49. At PND70, rats were submitted to the prepulse inhibition test. FDG-PET and T2-weighted MRI brain studies were performed at adulthood. IOS markers were evaluated in frozen brain tissue. RESULTS: MIN treatment did not prevent prepulse inhibition test behavioral deficits in MIS offspring. However, MIN prevented morphometric abnormalities in the third ventricle but not in the hippocampus. Additionally, MIN reduced brain metabolism in cerebellum and increased it in nucleus accumbens. Finally, MIN reduced the expression of iNOS (prefrontal cortex, caudate-putamen) and increased the levels of KEAP1 (prefrontal cortex), HO1 and NQO1 (amygdala, hippocampus), and HO1 (caudate-putamen). CONCLUSIONS: MIN treatment during adolescence partially counteracts volumetric abnormalities and IOS deficits in the MIS model, likely via iNOS and Nrf2-ARE pathways, also increasing the expression of cytoprotective enzymes. However, MIN treatment during this peripubertal stage does not prevent sensorimotor gating deficits. Therefore, even though it does not prevent all the MIS-derived abnormalities evaluated, our results suggest the potential utility of early treatment with MIN in other schizophrenia domains.


Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Brain Diseases, Metabolic/drug therapy , Minocycline/pharmacology , Nervous System Malformations/pathology , Neurodevelopmental Disorders/drug therapy , Oxidative Stress/drug effects , Prenatal Exposure Delayed Effects/drug therapy , Prepulse Inhibition/drug effects , Schizophrenia/drug therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Behavior, Animal/drug effects , Brain Diseases, Metabolic/etiology , Disease Models, Animal , Female , Magnetic Resonance Imaging , Male , Minocycline/administration & dosage , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/etiology , Neurodevelopmental Disorders/chemically induced , Neurodevelopmental Disorders/immunology , Positron-Emission Tomography , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/immunology , Rats , Rats, Wistar , Schizophrenia/chemically induced , Schizophrenia/immunology
7.
Eur Neuropsychopharmacol ; 46: 14-27, 2021 05.
Article En | MEDLINE | ID: mdl-33735708

The likely involvement of inflammation and oxidative stress (IOS) in mental disease has led to advocate anti-oxidant and anti-inflammatory drugs as therapeutic strategies in the treatment of schizophrenia. Since omega-3 fatty acids (ω-3) show anti-inflammatory/neuroprotective properties, we aim to evaluate whether ω-3 treatment during adolescence in the maternal immune stimulation (MIS) animal model of schizophrenia could prevent the brain and behavioural deficits described in adulthood. At gestational day 15, PolyI:C (4 mg/kg) or saline (VH) were injected to pregnant Wistar rats. Male offspring received ω-3 (800 mg/kg) or saline (Sal) daily from postnatal day (PND) 35-49, defining 4 groups: MIS-ω-3; MIS-Sal; VH-ω-3 and VH-Sal. At PND70, rats were submitted to prepulse inhibition test (PPI). FDG-PET and T2-weighted MRI brain studies were performed in adulthood and analyzed by means of SPM12. IOS markers were measured in selected brain areas. MIS-offspring showed a PPI deficit compared with VH-offspring and ω-3 treatment prevented this deficit. Also, ω-3 reduced the brain metabolism in the deep mesencephalic area and prevented the volumetric abnormalities in the hippocampus but not in the ventricles in MIS-offspring. Besides, ω-3 reduced the expression of iNOS and Keap1 and increased the activity/concentration of HO1, NQO1 and GPX. Our study demonstrates that administration of ω-3 during adolescence prevents PPI behavioural deficits and hippocampal volumetric abnormalities, and partially counteracts IOS deficits via iNOS and Nrf2-ARE pathways in the MIS model. This study highlights the need for novel strategies based on anti-inflammatory/anti-oxidant compounds to alter the disease course in high-risk populations at early stages.


Fatty Acids, Omega-3 , Prenatal Exposure Delayed Effects , Schizophrenia , Virus Diseases , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants , Disease Models, Animal , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Female , Kelch-Like ECH-Associated Protein 1 , Male , NF-E2-Related Factor 2/therapeutic use , Poly I-C , Pregnancy , Prenatal Exposure Delayed Effects/prevention & control , Rats , Rats, Wistar , Schizophrenia/diagnostic imaging , Schizophrenia/drug therapy , Schizophrenia/prevention & control , Virus Diseases/drug therapy
8.
Eur Neuropsychopharmacol ; 29(7): 880-896, 2019 07.
Article En | MEDLINE | ID: mdl-31229322

Inflammation and oxidative stress (IOS) are considered key pathophysiological elements in the development of mental disorders. Recent studies demonstrated that the antipsychotic risperidone elicits an antiinflammatory effect in the brain. We administered risperidone for 2-weeks at adolescence to assess its role in preventing brain-related IOS changes in the maternal immune stimulation (MIS) model at adulthood. We also investigated the development of volumetric and neurotrophic abnormalities in areas related to the HPA-axis. Poly I:C (MIS) or saline (Sal) were injected into pregnant Wistar rats on GD15. Male offspring received risperidone or vehicle daily from PND35-PND49. We studied 4 groups (8-15 animals/group): Sal-vehicle, MIS-vehicle, Sal-risperidone and MIS-risperidone. [18F]FDG-PET and MRI studies were performed at adulthood and analyzed using SPM12 software. IOS and neurotrophic markers were measured using WB and ELISA assays in brain tissue. Risperidone elicited a protective function of schizophrenia-related IOS deficits. In particular, risperidone elicited the following effects: reduced volume in the ventricles and the pituitary gland; reduced glucose metabolism in the cerebellum, periaqueductal gray matter, and parietal cortex; higher FDG uptake in the cingulate cortex, hippocampus, thalamus, and brainstem; reduced NFκB activity and iNOS expression; and increased enzymatic activity of CAT and SOD in some brain areas. Our study suggests that some schizophrenia-related IOS changes can be prevented in the MIS model. It also stresses the need to search for novel strategies based on anti-inflammatory compounds in risk populations at early stages in order to alter the course of the disease.


Antipsychotic Agents/pharmacology , Brain/drug effects , Oxidative Stress/drug effects , Risperidone/pharmacology , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Magnetic Resonance Imaging , Male , NF-kappa B/metabolism , Poly I-C , Positron-Emission Tomography , Rats , Rats, Wistar
9.
Neuropharmacology ; 135: 202-210, 2018 06.
Article En | MEDLINE | ID: mdl-29551688

Tapentadol is an analgesic that acts as an agonist of µ opioid receptors (MOR) and that inhibits noradrenaline reuptake. Data from healthy rats show that tapentadol inhibits neuronal activity in the locus coeruleus (LC), a nucleus regulated by both the noradrenergic and opioid systems. Thus, we set out to investigate the effect of tapentadol on LC activity in streptozotocin (STZ)-induced diabetic rats, a model of diabetic polyneuropathy, by analyzing single-unit extracellular recordings of LC neurons. Four weeks after inducing diabetes, tapentadol dose-response curves were obtained from animals pre-treated with RX821002 or naloxone (alpha2-adrenoceptors and opioid receptors antagonists, respectively). In STZ rats, the spontaneous activity of LC neurons (0.9 ±â€¯0.1 Hz) was lower than in naïve animals (1.5 ±â€¯0.1 Hz), and tapentadol's inhibitory effect was also weaker. Alpha2-adrenoceptors blockade by RX821002 (100 µg/kg i.v.) in STZ animals significantly increased the spontaneous activity (from 0.8 ±â€¯0.1 to 1.4 ±â€¯0.2 Hz) and it dampened the inhibition of LC neurons produced by tapentadol. However, opioid receptors blockade following naloxone pre-treatment (5 mg/kg i.v.) did not alter the spontaneous firing rate (0.9 ±â€¯0.2 vs 0.9 ±â€¯0.2 Hz) or the inhibitory effect of tapentadol on LC neurons in STZ animals. Thus, diabetic polyneuropathy appears to exert neuroplastic changes in LC neurotransmission, enhancing the sensitivity of alpha2-adrenoceptors and dampening opioid receptors expression. Tapentadol's activity seems to be predominantly mediated through its noradrenergic effects rather than its influence on opioid receptors in the STZ model of diabetic polyneuropathy.


Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/complications , Diabetic Neuropathies/physiopathology , Locus Coeruleus/cytology , Neural Inhibition/drug effects , Tapentadol/pharmacology , Action Potentials/drug effects , Animals , Dose-Response Relationship, Drug , Idazoxan/analogs & derivatives , Idazoxan/pharmacology , Male , Naloxone/pharmacology , Neurons/physiology , Pain Measurement/drug effects , Rats , Tapentadol/antagonists & inhibitors
10.
Brain Stimul ; 11(1): 222-230, 2018.
Article En | MEDLINE | ID: mdl-29074339

BACKGROUND: Deep Brain Stimulation (DBS) of the subgenual cingulate cortex (SCC) is a promising therapeutic alternative to treat resistant major depressive disorder. In preclinical studies, DBS of the ventromedial prefrontal cortex (vmPFC, the rodent SCC correlate) provokes an antidepressant-like effect, along with changes in noradrenaline levels at the site of stimulation. Hence, DBS appears to activate the noradrenergic-locus coeruleus (LC) system. OBJECTIVE/HYPOTHESIS: The aim of this study was to evaluate the effect of vmPFC DBS on the electrical activity of noradrenergic LC neurons, cortical oscillations and coherence between both brain areas in male rats. METHODS: The antidepressant-like effect of vmPFC DBS was evaluated through the forced swimming test. Tonic and evoked activity of LC neurons, LC activity of alpha2-adrenoceptors, local field potentials from LC and electrocorticogram signals were studied after DBS by electrophysiological recordings in anaesthetized rats. The effect of DBS on tyrosine hydroxylase (TH), noradrenaline transporters (NAT), phosphorylation of the extracellular signal-regulated kinase (ERK) and corticotropin releasing factor (CRF) expression in the LC were measured by western blot assays. RESULTS: DBS induced an antidepressant-like effect increasing climbing behaviour in the FST that was accompanied by a robust increase of TH expression in the rat LC. The tonic and evoked activity of LC neurons was enhanced by DBS, which impaired alpha2-adrenoceptors activity. DBS also promoted an increase in slow LC oscillations, as well as a shift in LC-cortical coherence. CONCLUSION: DBS of the vmPFC appears to affect the LC, producing changes that may underlie its antidepressant-like effects.


Deep Brain Stimulation , Depression/therapy , Locus Coeruleus/cytology , Locus Coeruleus/physiology , Norepinephrine/metabolism , Prefrontal Cortex/physiology , Animals , Depressive Disorder, Major/therapy , Gyrus Cinguli/physiology , Male , Rats , Rats, Wistar , Swimming
11.
Expert Opin Drug Discov ; 9(5): 567-78, 2014 May.
Article En | MEDLINE | ID: mdl-24738878

INTRODUCTION: Depression is a multifactorial mood disorder with a high prevalence worldwide. Until now, treatments for depression have focused on the inhibition of monoaminergic reuptake sites, which augment the bioavailability of monoamines in the CNS. Advances in drug discovery have widened the therapeutic options with the synthesis of so-called selective serotonin reuptake inhibitors (SSRIs), such as fluoxetine. AREAS COVERED: The aim of this case history is to describe and discuss the pharmacokinetic and pharmacodynamic profiles of fluoxetine, including its acute effects and the adaptive changes induced after long-term treatment. Furthermore, the authors review the effect of fluoxetine on neuroplasticity and adult neurogenesis. In addition, the article summarises the preclinical behavioural data available on fluoxetine's effects on depressive-like behaviour, anxiety and cognition as well as its effects on other diseases. Finally, the article describes the seminal studies validating the antidepressant effects of fluoxetine. EXPERT OPINION: Fluoxetine is the first selective SSRI that has a recognised clinical efficacy and safety profile. Since its discovery, other molecules that mimic its mechanism of action have been developed, commencing a new age in the treatment of depression. Fluoxetine has also demonstrated utility in the treatment of other disorders for which its prescription has now been approved.


Antidepressive Agents, Second-Generation/pharmacology , Antidepressive Agents, Second-Generation/pharmacokinetics , Drug Discovery/history , Fluoxetine/pharmacology , Fluoxetine/pharmacokinetics , Animals , Antidepressive Agents, Second-Generation/chemistry , Drug Evaluation, Preclinical/history , Fluoxetine/chemistry , History, 20th Century , Humans
12.
Eur Neuropsychopharmacol ; 24(6): 996-1003, 2014 Jun.
Article En | MEDLINE | ID: mdl-24491949

Depression can influence pain and vice versa, yet the biological mechanisms underlying how one influences the pathophysiology of the other remains unclear. Dysregulation of locus coeruleus-noradrenergic transmission is implicated in both conditions, although it is not known whether this effect is exacerbated in cases of co-morbid depression and chronic pain. We studied locus coeruleus activity using immunofluorescence and electrophysiological approaches in rats subjected to unpredictable chronic mild stress (CMS, an experimental model of depression) and/or chronic constriction injury (CCI, a model of chronic neuropathic pain) for 2 weeks. CCI alone had no effect on any of the locus coeruleus parameters studied, while CMS led to a slight reduction in the electrophysiological activity of the locus coeruleus. Furthermore, CMS was associated with an increase in the number of tyrosine hydroxylase-positive cells in the locus coeruleus, although they were smaller in size. Interestingly, these effects of CMS were exacerbated when combined with CCI, even though no changes in the α2-adrenoreceptors or the noradrenaline transporter were observed in any group. Together, these findings suggest that CMS triggers several modifications in locus coeruleus-noradrenergic transmission that are exacerbated by co-morbid chronic pain.


Locus Coeruleus/physiopathology , Neuralgia/physiopathology , Neurons/physiology , Stress, Psychological/physiopathology , Synaptic Transmission/physiology , Action Potentials/physiology , Animals , Cell Size , Chronic Disease , Chronic Pain/epidemiology , Chronic Pain/pathology , Chronic Pain/physiopathology , Comorbidity , Depressive Disorder/epidemiology , Depressive Disorder/pathology , Depressive Disorder/physiopathology , Disease Models, Animal , Locus Coeruleus/pathology , Male , Neuralgia/epidemiology , Neuralgia/pathology , Neurons/pathology , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/metabolism , Sciatic Nerve/injuries , Stress, Psychological/epidemiology , Stress, Psychological/pathology , Tyrosine 3-Monooxygenase/metabolism
13.
Pain ; 154(10): 2014-2023, 2013 Oct.
Article En | MEDLINE | ID: mdl-23792242

Stressful experiences seem to negatively influence pain perception through as yet unknown mechanisms. As the noradrenergic locus coeruleus (LC) nucleus coordinates many components of the stress response, as well as nociceptive transmission, we evaluated whether the sensory and affective dimension of chronic neuropathic pain worsens in situations of stress due to adaptive changes of LC neurons. Accordingly, male rats were socially isolated for 5 weeks, and in the last 2 weeks, neuropathic pain was induced by chronic constriction injury. In this situation of stress, chronic pain selectively heightened the animal's aversion to painful experiences (affective pain), as measured in the place escape/avoidance test, although no changes were observed in the sensory dimension of pain. In addition, electrophysiological recordings of LC neurons showed a low tonic but exacerbated nociceptive-evoked activity when the injured paw was stimulated. These changes were accompanied by an increase in tyrosine hydroxylase and gephyrin expression in the LC. Furthermore, intra-LC administration of bicuculline, a γ-aminobutyric acid-A receptor antagonist, attenuated the negative affective effects of pain. These data show that changes in the LC are greater than those expected from the simple summation of each independent factor (pain and stress), revealing mechanisms through which stressors may exacerbate pain perception without affecting the sensorial dimension.


Avoidance Learning/physiology , Chronic Pain/physiopathology , Locus Coeruleus/physiology , Pain Measurement/methods , Social Isolation , Stress, Psychological/physiopathology , Animals , Chronic Pain/psychology , Male , Pain Measurement/psychology , Rats , Rats, Sprague-Dawley , Social Isolation/psychology , Stress, Psychological/psychology
14.
Neuropharmacology ; 72: 250-8, 2013 Sep.
Article En | MEDLINE | ID: mdl-23664814

Tapentadol is a novel centrally acting drug that combines mu-opioid receptor (MOR) agonism and noradrenaline reuptake inhibition (NRI), producing analgesic effects in various painful conditions. We investigated the acute effects of tapentadol in the locus coeruleus (LC), a central nucleus regulated by the noradrenergic and opioid systems that is critical in pain modulation. In single-unit extracellular recordings of LC neurons from anaesthetized male Sprague-Dawley rats, tapentadol clearly inhibited the spontaneous electrophysiological activity of LC neurons in a dose-dependent manner (ED50 = 0.8 mg/kg). This inhibitory effect was reversed by RX821002 (an alpha2-adrenoceptor antagonist) and naloxone (a mu-opioid receptor antagonist) by 96.7% and 28.2%, respectively. Pretreatment with RX821002, N-ethoxycarbonyl-2-ethoxy-1-2-dihydroquinoline (EEDQ, an irreversible alpha2-adrenoceptor antagonist) or naloxone shifted the tapentadol dose-effect curve to the right (ED50 = 2.2 mg/kg, 2.0 mg/kg and 2.1 mg/kg, respectively). Furthermore, tapentadol inhibited the LC response to mechanical stimulation of the hindpaw in a dose-dependent manner. In summary, we demonstrate that acute administration of tapentadol inhibits LC neurons in vivo, mainly due to the activation of alpha2-adrenoceptors. These data suggest that both the noradrenergic and opioid systems participate in the inhibitory effect of tapentadol on LC neurons, albeit to different extents, which may account for its potent analgesic effect and mild opioidergic side-effects.


Action Potentials/drug effects , Analgesics, Opioid/pharmacology , Locus Coeruleus/cytology , Neurons/drug effects , Phenols/pharmacology , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Drug Interactions , Idazoxan/analogs & derivatives , Idazoxan/pharmacology , Male , Narcotic Antagonists/pharmacology , Neural Inhibition/drug effects , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Tapentadol
15.
Expert Opin Drug Discov ; 7(8): 745-55, 2012 Aug.
Article En | MEDLINE | ID: mdl-22680253

INTRODUCTION: Affective disorders, including major depressive disorder (MDD), are among the most severely disabling mental disorders, and in many cases are associated with poor treatment outcomes. From the emergence of the monoamine hypothesis of depression, the first-line treatment for MDD had mainly acted by inhibiting monoamine reuptake, and thereby increasing these levels in the synaptic cleft. However, in recent years, several new antidepressant drugs have appeared, including duloxetine, a dual serotonin (5-HT) and noradrenaline (NA) reuptake inhibitor recommended for the treatment of MDD. AREAS COVERED: The article reviews and discusses the biochemical and functional profile of duloxetine splitting the review into acute and long-term treatment with this dual monoamine reuptake inhibitor. In addition, the authors summarize available preclinical behavioral research data, which have demonstrated among other effects, the antidepressant-like activity of duloxetine in several animal models. The authors focus on the most recent literature on synaptic neuroplasticity modulation of this antidepressant drug. Finally, the authors briefly mention other approved indications of duloxetine. EXPERT OPINION: Duloxetine inhibits 5-HT and NA reuptake, effectively desensitizes various autoreceptors and promotes neuroplasticity. Clinically, duloxetine is an effective antidepressant that is well tolerated and has significant efficacy in the treatment of MDD.


Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Thiophenes/pharmacology , Thiophenes/therapeutic use , Animals , Drug Evaluation, Preclinical , Duloxetine Hydrochloride , Humans
16.
Curr Neuropharmacol ; 9(4): 530-52, 2011 Dec.
Article En | MEDLINE | ID: mdl-22654714

Depression is a neuropsychiatric disorder affecting a huge percentage of the active population especially in developed countries. Research has devoted much of its attention to this problematic and many drugs have been developed and are currently prescribed to treat this pathology. Yet, many patients are refractory to the available therapeutic drugs, which mainly act by increasing the levels of the monoamines serotonin and noradrenaline in the synaptic cleft. Even in the cases antidepressants are effective, it is usually observed a delay of a few weeks between the onset of treatment and remission of the clinical symptoms. Additionally, many of these patients who show remission with antidepressant therapy present a relapse of depression upon treatment cessation. Thus research has focused on other possible molecular targets, besides monoamines, underlying depression. Both basic and clinical evidence indicates that depression is associated with several structural and neurochemical changes where the levels of neurotrophins, particularly of brain-derived neurotrophic factor (BDNF), are altered. Antidepressants, as well as other therapeutic strategies, seem to restore these levels. Neuronal atrophy, mostly detected in limbic structures that regulate mood and cognition, like the hippocampus, is observed in depressed patients and in animal behavioural paradigms for depression. Moreover, chronic antidepressant treatment enhances adult hippocampal neurogenesis, supporting the notion that this event underlies antidepressants effects. Here we review some of the preclinical and clinical studies, aimed at disclosing the role of neurotrophins in the pathophysiological mechanisms of depression and the mode of action of antidepressants, which favour the neurotrophic/neurogenic hypothesis.

...