Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-36209771

RESUMEN

The NMDA antagonist ketamine demonstrated a fast antidepressant activity in treatment-resistant depression. Pre-clinical studies suggest that de novo synthesis of the brain-derived neurotrophic factor (BDNF) in the PFC might be involved in the rapid antidepressant action of ketamine. Applying a genetic model of impaired glutamate release, this study aims to further identify the molecular mechanisms that could modulate antidepressant action and resistance to treatment. To that end, mice knocked-down for the vesicular glutamate transporter 1 (VGLUT1+/-) were used. We analyzed anhedonia and helpless behavior as well as the expression of the proteins linked to glutamate transmission in the PFC of mice treated with ketamine or the reference antidepressant reboxetine. Moreover, we analyzed the acute effects of ketamine in VGLUT1+/- mice pretreated with chronic reboxetine or those that received a PFC rescue expression of VGLUT1. Chronic reboxetine rescued the depressive-like phenotype of the VGLUT1+/- mice. In addition, it enhanced the expression of the proteins linked to the AMPA signaling pathway as well as the immature form of BDNF (pro-BDNF). Unlike WT mice, ketamine had no effect on anhedonia or pro-BDNF expression in VGLUT1+/- mice; it also failed to decrease phosphorylated eukaryote elongation factor 2 (p-eEF2). Nevertheless, we found that reboxetine administered as pretreatment or PFC overexpression of VGLUT1 did rescue the antidepressant-like activity of acute ketamine in the mice. Our results strongly suggest that not only do PFC VGLUT1 levels modulate the rapid-antidepressant action of ketamine, but also highlight a possible mechanism for antidepressant resistance in some patients.


Asunto(s)
Ketamina , Proteína 1 de Transporte Vesicular de Glutamato , Animales , Ratones , Anhedonia , Antidepresivos/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Depresión/metabolismo , Modelos Animales de Enfermedad , Ácido Glutámico/metabolismo , Ketamina/farmacología , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Reboxetina/farmacología , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
2.
Neurourol Urodyn ; 41(6): 1482-1488, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35771361

RESUMEN

AIMS: Urethral closure function is essential for urinary continence in women and decreased urethral pressure is associated with stress urinary incontinence (SUI). For decades, the effects of serotonergic drugs on central neural control of urethral closure have been investigated and discussed. Epidemiological studies suggest that the use of selective serotonin reuptake inhibitors (SSRIs), such as citalopram, is associated with SUI. However, the literature findings are conflicting. This study aimed to evaluate citalopram's effect on opening urethral pressure (OUP) in healthy women. METHODS: We conducted a randomized, double-blind, placebo- and active-controlled crossover study in 24 healthy women. On three study days, which were separated by 8 days of washout, the subjects received single doses of either 40 mg citalopram (and placeboreboxetine ), 8 mg reboxetine (and placebocitalopram ), or two placebos. Study drugs were administered at a 1-h interval due to a difference in estimated time to peak plasma concentration (tmax ). We measured OUP with urethral pressure reflectometry under both resting and squeezing conditions of the pelvic floor at estimated tmax for both study drugs (one timepoint). RESULTS: Compared to placebo, citalopram increased OUP by 6.6 cmH2 0 (95% confidence interval [CI] 0.04-13.1, p = 0.048) in resting condition. In squeezing condition, OUP increased by 7.1 cmH2 0 (95% CI: 1.3-12.9, p = 0.01). Reboxetine increased OUP by 30.0 cmH2 0 in resting condition compared to placebo (95% CI: 23.5-36.5, p < 0.001), and 27.0 cmH2 0 (95% CI: 21.2-32.8, p < 0.001) in squeezing condition. CONCLUSION: Citalopram increased OUP slightly compared to placebo suggesting that SSRI treatment does not induce or aggravate SUI.


Asunto(s)
Citalopram , Incontinencia Urinaria de Esfuerzo , Citalopram/efectos adversos , Estudios Cruzados , Método Doble Ciego , Femenino , Humanos , Reboxetina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/efectos adversos , Uretra , Incontinencia Urinaria de Esfuerzo/tratamiento farmacológico
3.
Behav Brain Res ; 416: 113535, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34416301

RESUMEN

Reboxetine, the first selective norepinephrine (NA) reuptake inhibitor used in the treatment of depression, mainly acts by binding to the NA transporter and blocking reuptake of extracellular NA. Recently, some other pharmacological targets beyond the NA transporter are being demonstrated for reboxetine. Peroxisome proliferator activated receptor α (PPARα) is a member of the nuclear hormone receptor family of ligand-dependent transcription factors. Previous reports have demonstrated the role of hippocampal PPARα in the pathophysiology of depression. Here we assume that hippocampal PPARα may participate in the antidepressant mechanism of reboxetine. Therefore, the chronic social defeat stress (CSDS) model of depression, various behavioral tests, the western blotting and adenovirus associated virus (AAV)-mediated genetic knockdown methods were used together in the present study. Our results showed that repeated reboxetine treatment markedly restored the decreasing effects of CSDS on the expression of hippocampal PPARα, brain-derived neurotrophic factor (BDNF) and phosphorylated cAMP response element binding protein (pCREB). Pharmacological blockade of PPARα notably prevented the antidepressant-like effects of reboxetine in the CSDS model. Furthermore, genetic knockdown of hippocampal PPARα also fully abolished the antidepressant-like effects of reboxetine in the CSDS model. Taken together, promoting the hippocampal PPARα expression participates in the antidepressant mechanism of reboxetine.


Asunto(s)
Inhibidores de Captación Adrenérgica/farmacología , Depresión/tratamiento farmacológico , Hipocampo/metabolismo , PPAR alfa/metabolismo , Reboxetina/farmacología , Animales , Conducta Animal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL
4.
Behav Brain Res ; 418: 113642, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-34755639

RESUMEN

Vocal deficits and anxiety are common, co-occurring, and interacting signs of Parkinson Disease (PD) that have a devastating impact on quality of life. Both manifest early in the disease process. Unlike hallmark motor signs of PD, neither respond adequately to dopamine replacement therapies, suggesting that their disease-specific mechanisms are at least partially extra-dopaminergic. Because noradrenergic dysfunction is also a defining feature of PD, especially early in the disease progression, drug therapies targeting norepinephrine are being trialed for treatment of motor and non-motor impairments in PD. Research assessing the effects of noradrenergic manipulation on anxiety and vocal impairment in PD, however, is sparse. In this pre-clinical study, we quantified the influence of pharmacologic manipulation of norepinephrine on vocal impairment and anxiety in Pink1-/- rats, a translational model of PD that demonstrates both vocal deficits and anxiety. Ultrasonic vocalization acoustics, anxiety behavior, and limb motor activity were tested twice for each rat: after injection of saline and after one of three drugs. We hypothesized that norepinephrine reuptake inhibitors (atomoxetine and reboxetine) and a ß receptor antagonist (propranolol) would decrease vocal impairment and anxiety compared to saline, without affecting spontaneous motor activity. Our results demonstrated that atomoxetine and reboxetine decreased anxiety behavior. Atomoxetine also modulated ultrasonic vocalization acoustics, including an increase in vocal intensity, which is almost always reduced in animal models and patients with PD. Propranolol did not affect anxiety or vocalization. Drug condition did not influence spontaneous motor activity. These studies demonstrate relationships among vocal impairment, anxiety, and noradrenergic systems in the Pink1-/- rat model of PD.


Asunto(s)
Ansiedad , Norepinefrina/farmacología , Enfermedad de Parkinson/fisiopatología , Vocalización Animal/efectos de los fármacos , Inhibidores de Captación Adrenérgica/farmacología , Animales , Clorhidrato de Atomoxetina/farmacología , Modelos Animales de Enfermedad , Humanos , Masculino , Proteínas Quinasas/genética , Ratas , Ratas Long-Evans , Reboxetina/farmacología
5.
Neuropharmacology ; 196: 108702, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34246685

RESUMEN

A growing body of literature implicates noradrenergic (NE) signaling in the modulation of ethanol consumption. However, relatively few studies have detailed specific brain pathways that mediate NE-associated binge-like ethanol consumption. To begin to fill this gap in the literature, male and female C57BL6/J and TH-ires-cre mice underwent pharmacological and chemogenetic testing, respectively, in combination with "drinking in the dark" procedures to model binge-like consumption of ethanol or sucrose solutions. First, we showed that intraperitoneal administration of the NE reuptake inhibitor, reboxetine, blunted binge-like ethanol intake in C57BL6/J mice. Chemogenetic activation of locus coeruleus (LC) tyrosine hydroxylase (TH)-expressing neurons blunted binge-like ethanol intake regardless of sex. Chemogenetic activation of LC projections to the lateral hypothalamus (LH), a region implicated in ethanol consumption, blunted binge-like ethanol drinking without altering sucrose intake in ethanol-experienced or ethanol-naïve mice. In C57BL/6 J mice, LH-targeted microinfusion of an α1-adrenergic receptor (AR) agonist blunted binge-like ethanol intake across both sexes, while LH infusion of a ß-AR agonist blunted binge-like ethanol intake in females exclusively. Finally, in mice with high baseline ethanol intake both an α1- AR agonist and an α-2 AR antagonist blunted binge-like ethanol intake. The present results provide novel evidence that increased NE tone in a circuit arising from the LC and projecting to the LH reduces binge-like ethanol drinking in mice, and may represent a novel approach to treating binge or heavy drinking prior to the development of dependence. This article is part of the special Issue on "Neurocircuitry Modulating Drug and Alcohol Abuse".


Asunto(s)
Inhibidores de Captación Adrenérgica/farmacología , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Depresores del Sistema Nervioso Central/administración & dosificación , Etanol/administración & dosificación , Área Hipotalámica Lateral/metabolismo , Locus Coeruleus/metabolismo , Norepinefrina/metabolismo , Reboxetina/farmacología , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Consumo Excesivo de Bebidas Alcohólicas/fisiopatología , Femenino , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/fisiopatología , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/fisiopatología , Masculino , Ratones , Vías Nerviosas , Tirosina 3-Monooxigenasa
6.
Int J Mol Sci ; 22(9)2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-34062902

RESUMEN

Currently utilized antidepressants have limited effectiveness and frequently incur undesired effects. Most antidepressants are thought to act via the inhibition of monoamine reuptake; however, direct binding to monoaminergic receptors has been proposed to contribute to both their clinical effectiveness and their side effects, or lack thereof. Among the target receptors of antidepressants, α1­adrenergic receptors (ARs) have been implicated in depression etiology, antidepressant action, and side effects. However, differences in the direct effects of antidepressants on signaling from the three subtypes of α1-ARs, namely, α1A-, α1B- and α1D­ARs, have been little explored. We utilized cell lines overexpressing α1A-, α1B- or α1D-ARs to investigate the effects of the antidepressants imipramine (IMI), desipramine (DMI), mianserin (MIA), reboxetine (REB), citalopram (CIT) and fluoxetine (FLU) on noradrenaline-induced second messenger generation by those receptors. We found similar orders of inhibition at α1A-AR (IMI < DMI < CIT < MIA < REB) and α1D­AR (IMI = DMI < CIT < MIA), while the α1B-AR subtype was the least engaged subtype and was inhibited with low potency by three drugs (MIA < IMI = DMI). In contrast to their direct antagonistic effects, prolonged incubation with IMI and DMI increased the maximal response of the α1B-AR subtype, and the CIT of both the α1A- and the α1B-ARs. Our data demonstrate a complex, subtype-specific modulation of α1-ARs by antidepressants of different groups.


Asunto(s)
Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Receptores Adrenérgicos alfa 1/genética , Animales , Antidepresivos/clasificación , Citalopram/farmacología , Depresión/etiología , Depresión/genética , Depresión/patología , Desipramina/farmacología , Fluoxetina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Imipramina/farmacología , Mianserina/farmacología , Ratones , Células PC12 , Ratas , Reboxetina/farmacología , Transducción de Señal/efectos de los fármacos
7.
Int J Neuropsychopharmacol ; 24(6): 490-498, 2021 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-33617635

RESUMEN

BACKGROUND: Noradrenaline has an important role as a neuromodulator of the central nervous system. Noradrenergic enhancement was recently shown to enhance glutamate-dependent cortical facilitation and long term potentiation-like plasticity. As cortical excitability and plasticity are closely linked to various cognitive processes, here we aimed to explore whether these alterations are associated with respective cognitive performance changes. Specifically, we assessed the impact of noradrenergic enhancement on motor learning (serial reaction time task), attentional processes (Stroop interference task), and working memory performance (n-back letter task). METHODS: The study was conducted in a cross-over design. Twenty-five healthy humans performed the respective cognitive tasks after a single dose of the noradrenaline reuptake inhibitor reboxetine or placebo administration. RESULTS: The results show that motor learning, attentional processes, and working memory performance in healthy participants were improved by reboxetine application compared with placebo. CONCLUSIONS: The results of the present study thus suggest that noradrenergic enhancement can improve memory formation and executive functions in healthy humans. The respective changes are in line with related effects of noradrenaline on cortical excitability and plasticity.


Asunto(s)
Inhibidores de Captación Adrenérgica/farmacología , Atención/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Nootrópicos/farmacología , Norepinefrina , Reboxetina/farmacología , Inhibidores de Captación Adrenérgica/administración & dosificación , Adulto , Estudios Cruzados , Femenino , Humanos , Masculino , Nootrópicos/administración & dosificación , Reboxetina/administración & dosificación , Adulto Joven
8.
Neurobiol Dis ; 152: 105295, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33549722

RESUMEN

Noradrenergic neurotransmission may play an important role in tremor modulation through its innervation of key structures of the central tremor circuits. Here, Parkinson's disease (PD) patients with (PDT+) or without (PDT-) rest tremor had 11C-methylreboxetine(11C-MeNER) positron emission tomography (PET) to test the hypothesis that noradrenaline terminal function was relatively preserved in PDT+ compared to PDT-. METHODS: Sixty-five PD patients and 28 healthy controls (HC) were scanned with 11C-MeNER PET. Patients were categorized as PDT+ if subscores in UPDRS-III item 3 or MDS-UPDRS-III item 17 was ≥2; remaining were categorized as PDT-. Simplified reference tissue model 2 distribution volume ratios (DVR) for 11C-MeNER were calculated for thalamus, dorsal and median raphe, locus coeruleus (LC) and red nucleus using time activity curves (TACs) obtained from volumes of interest (VOI). Data were statistically interrogated with a general linear mixed model using 'region', and 'group' as factors and the interaction of 'region x group' was examined. RESULTS: Tremor positive PD patients had a significantly higher mean 11C-MeNER DVR compared to PDT- in LC and thalamus. The PDT+ mean LC DVR was similar to that of HC. PDT+ mean 11C-MeNER DVRs were significantly lower than HC in the dorsal raphe while the PDT- group showed significantly lower mean 11C-MeNER DVR across all regions compared to HC. CONCLUSION: While both PD T+ and PD T- groups showed a significant loss of noradrenaline terminal function compared to controls, noradrenergic neurons were relatively preserved in PDT+ in LC and thalamus. The greater loss of noradrenergic transporters in PDT- in LC and thalamus compared with PDT+ is in line with earlier in-vitro studies and could potentially contribute to their tremor negative phenotype.


Asunto(s)
Neuronas Adrenérgicas/patología , Encéfalo/patología , Enfermedad de Parkinson/patología , Terminales Presinápticos/patología , Temblor/patología , Neuronas Adrenérgicas/metabolismo , Anciano , Encéfalo/diagnóstico por imagen , Radioisótopos de Carbono/farmacología , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacología , Reboxetina/farmacología , Temblor/diagnóstico por imagen , Temblor/etiología
9.
Nat Prod Res ; 35(6): 1042-1045, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31135223

RESUMEN

Armillaria mellea, also known as Hazel mushroom, is a delicious food material and traditional herbal medicine in East Asia. Protoilludane sesquiterpenoid aromatic esters from A. mellea (PSAM) are the main active components with antibacterial and anticancer activities. This study explored the antidepressant-like activities of PSAM and its possible mechanisms of action using the open field test (OFT), tail suspension test (TST) and forced swimming test (FST) in mice for the first time. The results revealed that PSAM (1 mg/kg, i.p.) exhibited markedly antidepressant-like activity, which could be reversed by pretreatment with haloperidol (a non-selective D2 receptor antagonist), bicuculline (a competitive GABA antagonist), NMDA (an agonist at the glutamate site). Meanwhile, PSAM also effectively increased the hippocampus dopamine (DA) and γ-aminobutyric acid (GABA) and decreased the hippocampus glutamate (Glu) levels of mice, indicating that the antidepressant-like effect of PSAM might be mediated by the DAergic, GABAergic and Gluergic systems.


Asunto(s)
Antidepresivos/uso terapéutico , Armillaria/química , Ésteres/uso terapéutico , Sesquiterpenos Policíclicos/uso terapéutico , Sesquiterpenos/uso terapéutico , Animales , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Depresión/tratamiento farmacológico , Depresión/fisiopatología , Dopamina/metabolismo , Fluoxetina/farmacología , Fluoxetina/uso terapéutico , Ácido Glutámico/metabolismo , Suspensión Trasera , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Prueba de Campo Abierto , Sesquiterpenos Policíclicos/farmacología , Reboxetina/farmacología , Reboxetina/uso terapéutico , Natación , Ácido gamma-Aminobutírico/metabolismo
10.
Addict Biol ; 26(3): e12957, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32815666

RESUMEN

Major depression (MD) is a frequent comorbidity in alcohol use disorder (AUD) patients. Antidepressant prescription is often limited by poor clinical outcomes or unwanted side effects in comorbid AUD-MD patients. Recent studies suggest that abrupt cessation of selective serotonin reuptake inhibitors antidepressant treatment increases alcohol consumption after an alcohol deprivation period in rats. However, the appearance of this effect after the treatment with selective noradrenaline reuptake inhibitors (SNRIs) is not known. Here, we report that interruption of subchronic (14 days) treatment with the SNRIs reboxetine (15 mg/kg/day intraperitoneally) resulted in escalation of ethanol intake when the animals resume alcohol self-administration. This effect of reboxetine treatment cessation was associated with a profound deactivation of the endocannabinoid/acylethanolamide signaling system in the prefrontal cortex but not in the dorsal hippocampus, as reflected by the decrease in the protein expression of the cannabinoid CB1 receptor, the PPARα receptor, the 2-arachidonoylglycerol synthesizing enzymes DAGLα and DGALß, and the endocanabinoid degrading enzyme MAGL. This was associated with dysregulation of the expression of glutamic acid receptors GluN1, GluA1, and mGlu5 in the medial prefrontal cortex and the dorsal hippocampus of the animals exposed to reboxetine. The present results further support the idea that abrupt cessation of antidepressant therapy along alcohol deprivation time can boost alcohol intake after relapse through mechanisms associated with endocannabinoid/glutamate signaling dysregulation. This finding might be relevant for patients suffering AUD/MD comorbidity where antidepressant therapy must be monitored with caution for avoiding unwanted side effects if adherence to the treatment is not fully achieved.


Asunto(s)
Alcoholismo/fisiopatología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Endocannabinoides/metabolismo , Ácido Glutámico/metabolismo , Reboxetina/farmacología , Animales , Hipocampo/efectos de los fármacos , Masculino , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Wistar , Autoadministración , Transducción de Señal/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/fisiopatología
11.
Exp Neurol ; 333: 113428, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32745472

RESUMEN

Chronic pain is one of the most challenging and debilitating symptoms to manage after traumatic brain injury (TBI), yet the underlying mechanisms remain elusive. The disruption of normal endogenous pain control mechanisms has been linked to several forms of chronic pain and may play a role in pain after TBI. We hypothesized therefore that dysfunctional descending noradrenergic and serotonergic pain control circuits may contribute to the loss of diffuse noxious inhibitory control (DNIC), a critical endogenous pain control mechanism, weeks to months after TBI. For these studies, the rat lateral fluid percussion model of mild TBI was used along with a DNIC paradigm involving a capsaicin-conditioning stimulus. We observed sustained failure of the DNIC response up to 180-days post injury. We confirmed, that descending α2 adrenoceptor-mediated noradrenergic signaling was critical for endogenous pain inhibition in uninjured rats. However, augmenting descending noradrenergic signaling using reboxetine, a selective noradrenaline reuptake inhibitor, failed to restore DNIC after TBI. Furthermore, blocking serotonin-mediated descending signaling using selective spinal serotonergic fiber depletion with 5, 7-dihydroxytryptamine was also unsuccessful at restoring endogenous pain modulation after TBI. Unexpectedly, increasing descending serotonergic signaling using the selective serotonin reuptake inhibitor escitalopram and the serotonin-norepinephrine reuptake inhibitor duloxetine restored the DNIC response in TBI rats at both 49- and 180- days post injury. Consistent with these observations, spinal serotonergic fiber depletion with 5, 7-dihydroxytryptamine eliminated the effects of escitalopram. Intact α2 adrenoceptor signaling, however, was not required for the serotonin-mediated restoration of DNIC after TBI. These results suggest that TBI causes maladaptation of descending nociceptive signaling mechanisms and changes in the function of both adrenergic and serotonergic circuits. Such changes could predispose those with TBI to chronic pain.


Asunto(s)
Lesiones Traumáticas del Encéfalo/fisiopatología , Control Inhibidor Nocivo Difuso/efectos de los fármacos , 5,7-Dihidroxitriptamina/farmacología , Inhibidores de Captación Adrenérgica/farmacología , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Capsaicina/farmacología , Dolor Crónico/etiología , Clorhidrato de Duloxetina/farmacología , Masculino , Vías Nerviosas/fisiopatología , Norepinefrina , Dimensión del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reboxetina/farmacología , Receptores Adrenérgicos alfa 2 , Serotonina , Serotoninérgicos/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
12.
Psychopharmacology (Berl) ; 237(4): 1131-1145, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31912189

RESUMEN

RATIONALE: Current data indicate that the noradrenergic system plays a critical role in neuropathic pain treatment. Notably, drugs that directly affect this system may have curative potential in neuropathy-associated pain. OBJECTIVES: The aim of this study was to evaluate the potential therapeutic efficacy of reboxetine, a potent and selective noradrenaline reuptake inhibitor, on hyperalgesia and allodynia responses in rats with experimental diabetes. Furthermore, mechanistic studies were performed to elucidate the possible mode of actions. METHODS: Experimental diabetes was induced by a single dose of streptozotocin. Mechanical hyperalgesia, mechanical allodynia, thermal hyperalgesia, and thermal allodynia responses in diabetic rats were evaluated by Randall-Selitto, dynamic plantar, Hargreaves, and warm plate tests, respectively. RESULTS: Reboxetine treatment (8 and 16 mg/kg for 2 weeks) demonstrated an effect comparable to that of the reference drug, pregabalin, improving the hyperalgesic and allodynic responses secondary to diabetes mellitus. Pretreatment with phentolamine, metoprolol, SR 59230A, and atropine did not alter the abovementioned effects of reboxetine; however, the administration of α-methyl-para-tyrosine methyl ester, propranolol, ICI-118,551, SCH-23390, sulpiride, and naltrindole significantly inhibited these effects. Moreover, reboxetine did not induce a significant difference in the rat plasma glucose levels. CONCLUSIONS: Our findings indicate that the antihyperalgesic and antiallodynic effects of reboxetine are mediated by the catecholaminergic system; ß2-adrenoceptors; D1-, D2/D3-dopaminergic receptors; and δ-opioid receptors. The results suggest that this analgesic effect of reboxetine, besides its neutral profile on glycemic control, may be advantageous in the pharmacotherapy of diabetic neuropathy-induced pain.


Asunto(s)
Inhibidores de Captación Adrenérgica/uso terapéutico , Neuropatías Diabéticas/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Reboxetina/uso terapéutico , Receptores Opioides delta/metabolismo , Inhibidores de Captación Adrenérgica/farmacología , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/tratamiento farmacológico , Neuropatías Diabéticas/sangre , Neuropatías Diabéticas/inducido químicamente , Hiperalgesia/sangre , Hiperalgesia/inducido químicamente , Masculino , Neuralgia/sangre , Neuralgia/inducido químicamente , Ratas , Ratas Sprague-Dawley , Reboxetina/farmacología , Receptores Adrenérgicos beta 2/metabolismo , Receptores Dopaminérgicos/metabolismo , Estreptozocina/toxicidad
13.
Mol Neurobiol ; 56(12): 8628-8642, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31297718

RESUMEN

The reduction of brain noradrenaline levels is associated to the initiation of Alzheimer's disease and contributes to its progression. This seems to be due mainly to the anti-neuroinflammatory actions of noradrenaline. The analysis of noradrenaline effects on brain cells demonstrates that it also regulates the production of the chemokine CCL2. In the present study, we analyzed the effect of the selective noradrenaline reuptake inhibitor, reboxetine, on the inflammatory and neurodegenerative alterations present in 5xFAD mice, and how the genetic removal of CCL2 affects reboxetine actions. We observed that the removal of CCL2 reduced the memory impairments in 5xFAD mice as well as the neuroinflammatory response, the accumulation of amyloid beta plaques, and the degeneration of neurons in the brain cortex. The administration of reboxetine with osmotic pumps for 28 days also resulted in anti-inflammatory and neuroprotective changes in 5xFAD mice, even in the absence of CCL2. Yet, 6-month-old CCL2KO mice presented a significant degree of neuroinflammation and neuronal damage. These findings indicate that reboxetine treatment prevents the brain alterations caused by prolonged overproduction of amyloid beta, being these effects independent of CCL2, which is a mediator of the damage caused by amyloid beta in the brain cortex, but necessary for the prevention of the development of neurodegeneration in normal healthy conditions.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Quimiocina CCL2/metabolismo , Inflamación/tratamiento farmacológico , Degeneración Nerviosa/tratamiento farmacológico , Reboxetina/uso terapéutico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Apoptosis/efectos de los fármacos , Axones/efectos de los fármacos , Axones/metabolismo , Axones/patología , Biomarcadores/metabolismo , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Inflamación/complicaciones , Inflamación/genética , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/tratamiento farmacológico , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/patología , Degeneración Nerviosa/complicaciones , Degeneración Nerviosa/genética , Reboxetina/farmacología
14.
Mol Neurobiol ; 56(4): 3038-3052, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30088175

RESUMEN

Current pharmacological treatments for major depressive disorder (MDD) are severely compromised by both slow action and limited efficacy. RNAi strategies have been used to evoke antidepressant-like effects faster than classical drugs. Using small interfering RNA (siRNA), we herein show that TASK3 potassium channel knockdown in monoamine neurons induces antidepressant-like responses in mice. TASK3-siRNAs were conjugated to cell-specific ligands, sertraline (Ser) or reboxetine (Reb), to promote their selective accumulation in serotonin (5-HT) and norepinephrine (NE) neurons, respectively, after intranasal delivery. Following neuronal internalization of conjugated TASK3-siRNAs, reduced TASK3 mRNA and protein levels were found in the brainstem 5-HT and NE cell groups. Moreover, Ser-TASK3-siRNA induced robust antidepressant-like behaviors, enhanced the hippocampal plasticity, and potentiated the fluoxetine-induced increase on extracellular 5-HT. Similar responses, yet of lower magnitude, were detected for Reb-TASK3-siRNA. These findings provide substantial support for TASK3 as a potential target, and RNAi-based strategies as a novel therapeutic approach to treat MDD.


Asunto(s)
Depresión/tratamiento farmacológico , Técnicas de Silenciamiento del Gen , Neuronas/metabolismo , Canales de Potasio/metabolismo , Reboxetina/administración & dosificación , Sertralina/administración & dosificación , Administración Intranasal , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Conducta Animal , Regulación hacia Abajo/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Norepinefrina/metabolismo , ARN Interferente Pequeño/metabolismo , Reboxetina/farmacología , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Sertralina/farmacología
15.
Behav Brain Res ; 359: 9-16, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30312638

RESUMEN

The aim of the present study was to investigate and compare the ability of three novel 5-HT6 and/or 5-HT7 receptor antagonists as follows: PZ-668-a preferential 5-HT6 antagonist; PZ-1433-a preferential 5-HT7 antagonist; and ADN-1184-a monoaminergic ligand with potent 5HT6/7 antagonist properties, to augment the effect of antidepressant drugs with different mechanisms of action (escitalopram, reboxetine, and bupropion) in the forced swim test in rats. In neurochemical ex vivo experiments, the influence of the tested compounds on levels of monoamines and their metabolites were determined in the rat frontal cortex, in addition to behavioral experiments. The results of our investigations revealed the differences in action of the tested compounds. PZ-668 strongly affected dopaminergic and faintly noradrenergic system, PZ-1433 induced a significant elevation in dopamine, noradrenaline, serotonin, and their metabolite levels, while ADN-1184 appeared to act mostly through dopaminergic transmission. The agent with 5-HT6 antagonistic properties (PZ-668) revealed an anti-immobility action of bupropion (primarily) and reboxetine in interaction studies. PZ-1433, the 5-HT7 preferential antagonist facilitated antidepressant effects of escitalopram and, to a lesser extent, bupropion, while ADN-1184, a multireceptor ligand, potentiated the effectiveness of escitalopram, reboxetine, and bupropion. The presented findings may contribute to further investigations of more effective and safer antidepressant drugs, and may help selecting optimal augmentation therapy in treatment-resistant depression.


Asunto(s)
Antidepresivos/farmacología , Monoaminas Biogénicas/metabolismo , Encéfalo/efectos de los fármacos , Trastorno Depresivo/tratamiento farmacológico , Actividad Motora/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , Animales , Encéfalo/metabolismo , Bupropión/farmacología , Citalopram/farmacología , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Isoxazoles/farmacología , Masculino , Actividad Motora/fisiología , Ratas Wistar , Reboxetina/farmacología , Receptores de Serotonina/metabolismo , Sulfonamidas/farmacología
16.
Sleep ; 42(4)2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30590857

RESUMEN

STUDY OBJECTIVES: Recent findings indicate that noradrenergic and antimuscarinic processes are crucial for sleep-related reductions in pharyngeal muscle activity. However, there are few human studies. Accordingly, this study aimed to determine if a combined noradrenergic and antimuscarinic intervention increases pharyngeal dilator muscle activity and improves airway function in sleeping humans. METHODS: Genioglossus (GG) and tensor palatini electromyography (EMG), pharyngeal pressure, upper airway resistance, and breathing parameters were acquired in 10 healthy adults (5 female) during two overnight sleep studies after 4 mg of reboxetine (REB) plus 20 mg of hyoscine butylbromide (HBB) or placebo using a double-blind, placebo-controlled, randomized, cross-over design. RESULTS: Compared with placebo, peak and tonic GG EMG were lower (Mean ± SD: 83 ± 73 vs. 130 ± 75, p = 0.021 and 102 ± 102 vs. 147 ± 123 % wakefulness, p = 0.021, respectively) but the sleep-related reduction in tensor palatini was less (Median [25th, 75th centiles]: 53[45, 62] vs. 34[28, 38] % wakefulness, p = 0.008) with the drug combination during nonrapid eye movement (non-REM) sleep. These changes were accompanied by improved upper airway function including reduced pharyngeal pressure swings, airway resistance, respiratory load compensation, and increased breathing frequency during N2. REB and HBB significantly reduced rapid eye movement sleep compared with placebo (0.6 ± 1.1 vs. 14.5 ± 6.8 % total sleep time, p < 0.001). CONCLUSIONS: Contrary to our hypothesis, GG muscle activity (% wakefulness) during non-REM sleep was lower with REB and HBB. However, sleep-related reductions in tensor palatini activity were less and upper airway function improved. These findings provide mechanistic insight into the role of noradrenergic and antimuscarinic processes on upper airway function in humans and have therapeutic potential for obstructive sleep apnea. CLINICAL TRIAL REGISTRATION: Australian New Zealand Clinical Trials Registry, https://www.anzctr.org.au, trial ID: ACTRN12616000469415.


Asunto(s)
Bromuro de Butilescopolamonio/farmacología , Parasimpatolíticos/farmacología , Músculos Faríngeos/fisiología , Reboxetina/farmacología , Apnea Obstructiva del Sueño/tratamiento farmacológico , Sueño REM/fisiología , Adulto , Resistencia de las Vías Respiratorias/fisiología , Australia , Estudios Cruzados , Método Doble Ciego , Electromiografía , Femenino , Humanos , Masculino , Nariz , Faringe/fisiopatología , Polisomnografía , Presión , Respiración , Escopolamina/farmacología , Sueño/fisiología , Apnea Obstructiva del Sueño/fisiopatología , Lengua/fisiopatología , Vigilia/fisiología
17.
Eur J Pharmacol ; 841: 57-66, 2018 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-30268663

RESUMEN

Center arena activity in open field (OF) test is associated with risk-taking behaviors. Noradrenaline and serotonin (5-HT) are neurotransmitters involved in both center arena activity and risk-based decision-making. However, the effects of noradrenergic/serotonergic systems on risk-based decision-making in rats with different center arena activity levels have not been clearly characterized. In this study, we explored the effects of the noradrenergic and serotonergic systems on risk-based decision-making in long center-time (LCT group) and short center-time (SCT group) rats. The two groups were formed based on performance in OF test. Then we tested their risk-based decision-making using probability discounting task (PDT); rats had to choose between "small/certain" lever that always delivered one pellet and "large/risky" lever that delivered four pellets in a probabilistic manner (100%, 50%, 25%, 12.5%). The results showed the SCT group chose the large/risky lever less often in 12.5% block and were more sensitive to loss than the SCT group. α2-adrenergic receptor agonist dexmedetomidine (0.01 mg/kg) decreased the frequency of risky choice, while the noradrenaline reuptake inhibitor (NRI) reboxetine (10 mg/kg) had the opposite effect only in the SCT group. Serotonin-noradrenaline reuptake inhibitor (SNRI) duloxetine (5 mg/kg) decreased preference for the large/risky option only in the SCT group. In contrast, pharmacological manipulations of the serotonin system did not affect the frequency of risky choices. These results suggest that noradrenergic system may be involved in weighing gains and losses for probabilistic discounting. Our findings also provide a better understanding of the involvement of center arena activity in risk-taking.


Asunto(s)
Toma de Decisiones , Norepinefrina/metabolismo , Asunción de Riesgos , Serotonina/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Conducta de Elección/efectos de los fármacos , Citalopram/farmacología , Toma de Decisiones/efectos de los fármacos , Dexmedetomidina/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Reboxetina/farmacología , Recompensa
18.
Psychopharmacology (Berl) ; 235(10): 2831-2846, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30091005

RESUMEN

RATIONALE: Antidepressant action has been linked to increased synaptic plasticity in which epigenetic mechanisms such as histone posttranslational acetylation could be involved. Interestingly, the histone deacetylases HDAC5 and SIRT2 are oppositely regulated by stress and antidepressants in mice prefrontal cortex (PFC). Besides, the neuroblastoma SH-SY5Y line is an in vitro neuronal model reliable to study drug effects with clear advantages over animals. OBJECTIVES: We aimed to characterize in vitro the role of HDAC5 and SIRT2 in antidepressant regulation of neuroplasticity. METHODS: SH-SY5Y cultures were incubated with imipramine, fluoxetine, and reboxetine (10 µM, 2 and 24 h) as well as the selective HDAC5 (MC3822, 5 µM, 24 h) or SIRT2 (33i, 5 µM, 24 h) inhibitors. The regulation of the brain-derived neurotrophic factor (BDNF), the vesicular glutamate transporter 1 (VGLUT1), the acetylated histones 3 (AcH3) and 4 (AcH4), HDAC5, and SIRT2 was studied. Comparatively, the long-term effects of these antidepressants (21 days, i.p.) in the mice (C57BL6, 8 weeks) PFC were studied. RESULTS: Antidepressants increased both in vitro and in vivo expression of BDNF, VGLUT1, AcH3, and AcH4. Moreover, imipramine and reboxetine increased the phosphorylated form of HDAC5 (P-HDAC5), mediating its cytoplasmic export. Further, SIRT2 was downregulated by all antidepressants. Finally, specific inhibition of HDAC5 and SIRT2 increased neuroplasticity markers. CONCLUSIONS: This study supports the validity of the SH-SY5Y model for studying epigenetic changes linked to synaptic plasticity induced by antidepressants as well as the effect of selective HDAC inhibitors. Particularly, nucleocytoplasmic export of HDAC5 and SIRT2 downregulation mediated by antidepressants could enhance synaptic plasticity markers leading to antidepressant action.


Asunto(s)
Antidepresivos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Histona Desacetilasas/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Sirtuina 2/metabolismo , Animales , Regulación hacia Abajo/fisiología , Epigénesis Genética/fisiología , Fluoxetina/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Imipramina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Distribución Aleatoria , Reboxetina/farmacología
19.
Med Sci Sports Exerc ; 50(8): 1579-1587, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29570538

RESUMEN

PURPOSE: To determine the role of noradrenergic modulation in the control of motor output, we compared the acute effect of reboxetine (REB), a noradrenaline reuptake inhibitor, to a placebo (PLA) on knee extensors motor performance and cortical and spinal excitability. METHODS: Eleven young men took part in two randomized experiments during which they received either 8 mg of REB or a PLA. The torque produced during a maximal voluntary contraction (MVC) and its variability (i.e., coefficient of variation) during submaximal contractions ranging from 5% to 50% MVC were measured. Paired electrical (PES) and transcranial magnetic stimulation (TMS) were used to assess changes in voluntary activation during MVC, and corticospinal (motor-evoked potential (MEP)) and spinal excitability (Hoffmann (H) reflex) during contraction at 20% MVC. RESULTS: MVC torque and torque steadiness increased respectively by 9.5% and 24% on average in REB compared with PLA condition (P < 0.001). Voluntary activation tested by TMS and PES was greater (~3%; P < 0.05) in REB than PLA condition. The increase in voluntary activation in REB condition was significantly correlated with subjects' initial voluntary activation level when tested by TMS (r = -0.62; P = 0.048) and PES (r = -0.86; P < 0.01). The maximal amplitudes of H reflex and MEP and the slope of their recruitment curves were enhanced by REB (P < 0.05). The ratio between the TMS-induced EMG silent period and the corresponding MEP (silent period/MEP) was reduced in REB condition (P < 0.01). CONCLUSIONS: The present findings indicate that voluntary activation and accuracy in force control can be increased by an enhanced level of noradrenaline concentration. This improvement in motor performance is accompanied by changes located at both cortical and spinal levels.


Asunto(s)
Rodilla/fisiología , Músculo Esquelético/inervación , Músculo Esquelético/fisiología , Norepinefrina/metabolismo , Tractos Piramidales/fisiología , Adulto , Estimulación Eléctrica , Electromiografía , Potenciales Evocados Motores/efectos de los fármacos , Reflejo H/efectos de los fármacos , Humanos , Contracción Isométrica/efectos de los fármacos , Masculino , Músculo Esquelético/efectos de los fármacos , Tractos Piramidales/efectos de los fármacos , Reboxetina/farmacología , Inhibidores de Captación de Serotonina y Norepinefrina/farmacología , Torque , Estimulación Magnética Transcraneal , Adulto Joven
20.
ChemMedChem ; 13(5): 453-463, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29451362

RESUMEN

In this work, we present label-free, mass-spectrometry-based binding assays (MS Binding Assays), targeting the human dopamine, norepinephrine, and serotonin transporters (hDAT, hNET, and hSERT) in simultaneous binding experiments. Using a validated LC-ESI-MS/MS method for quantification of the selective dopamine transporter inhibitor (R,R)-4-(2-benzhydryloxyethyl)-1-(4-fluorobenzyl)piperidin-3-ol ((R,R)-D-84), the selective norepinephrine transporter inhibitor (S,S)-reboxetine, and the selective serotonin reuptake inhibitor (S)-citalopram, binding affinities at the three monoamine transporters could be characterized simultaneously in a single binding experiment. The performed simultaneous saturation and competition experiments yielded results that are in good accordance with those determined in MS Binding Assays addressing the monoamine transporters individually. The results obtained from this study underscore the potential of MS Binding Assays for simultaneous affinity determination at different targets, which is difficult to accomplish with conventional radioligand binding assays.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Citalopram/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/antagonistas & inhibidores , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/antagonistas & inhibidores , Piperidinas/farmacología , Reboxetina/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Sitios de Unión/efectos de los fármacos , Cromatografía Liquida , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Humanos , Estructura Molecular , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA