Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Exp Brain Res ; 232(6): 1793-803, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24584836

RESUMEN

The olfactory bulbectomized (OB) rat, an animal model of chronic depression with comorbid anxiety, exhibits a profound dysregulation of the brain serotonergic signalling, a neurotransmission system involved in pain transmission and modulation. We here report an increased nociceptive response of OB rats in the tail flick test which is reverted after chronic, but not acute, administration of fluoxetine. Autoradiographic studies demonstrated down-regulation of 5-HT transporters ([(3)H]citalopram binding) and decreased functionality of 5-HT1A receptors (8-OH-DPAT-stimulated [(35)S]GTPγS binding) in the dorsal horn of the lumbar spinal cord in OB rats. Acute administration of fluoxetine (5-40 mg/kg i.p.) did not modify tail flick latencies in OB rats. However, chronic fluoxetine (10 mg/kg/day s.c., 14 days; osmotic minipumps) progressively attenuated OB-associated thermal hyperalgesia, and a total normalization of the nociceptive response was achieved at the end of the treatment with the antidepressant. In these animals, autoradiographic studies revealed further down-regulation of 5-HT transporters and normalization in the functionality of 5-HT1A receptors on the spinal cord. On the other hand, acute morphine (0.5-10 mg/kg s.c.) produced a similar analgesic effect in OB and sham and OB rats, and no changes were detected in the density ([(3)H]DAMGO binding) and functionality (DAMGO-stimulated [(35)S]GTPγS binding) of spinal µ-opioid receptors in OB rats before and after chronic fluoxetine. Our findings demonstrate the participation of the spinal serotonergic system in the increased thermal nociception exhibited by the OB rat and the antinociceptive effect of chronic fluoxetine in this animal model of depression.


Asunto(s)
Depresión/complicaciones , Depresión/patología , Hiperalgesia/etiología , Serotonina/metabolismo , Médula Espinal/metabolismo , Animales , Antidepresivos/uso terapéutico , Autorradiografía , Depresión/tratamiento farmacológico , Modelos Animales de Enfermedad , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacocinética , Conducta Exploratoria/fisiología , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Hiperalgesia/tratamiento farmacológico , Masculino , Morfina/uso terapéutico , Neurotransmisores/farmacocinética , Bulbo Olfatorio/lesiones , Bulbo Olfatorio/cirugía , Dimensión del Dolor , Radiografía , Radioisótopos/farmacocinética , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1A/metabolismo , Receptores Opioides mu/metabolismo , Serotoninérgicos/farmacocinética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Médula Espinal/diagnóstico por imagen , Médula Espinal/efectos de los fármacos
2.
Neurochem Res ; 37(5): 1037-42, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22270908

RESUMEN

Lysophosphatidylinositol (LPI) is a biologically active lipid that produces a number of responses in cultured cells, and has been suggested to have neuroprotective properties in vivo. Some of the actions of LPI are mediated by G-protein coupled receptors, but it is not known whether G-protein coupled receptor-mediated responses can be seen in intact brain tissue. In consequence, in the present study, we investigated autoradiographically whether LPI increased the [(35)S]GTPγS binding level in brain tissue slices. In standard assay conditions, where as a positive control a robust response to cannabinoid receptor activation by the agonist ligand CP55,940 was seen, there was no increase in the autoradiographic density over basal produced by LPI. However, when the conditions were modified (incubation at 4°C rather than at 25°C, incubation time increased to 3 h, GDP concentration reduced from 2 to 0.1 mM), a significant increase in [(35)S]GTPγS autoradiographic density in response to 10 µM LPI was seen in the prefrontal cortex, hippocampus, and cortex at the level of the hippocampus, although the degree of increase was small and very variable. No significant increases were seen in the hypothalamus or cerebellum. It is concluded that LPI, in the right conditions, can activate a sufficient number of G-proteins in the rat prefrontal cortex and hippocampus to produce a response in the [(35)S]GTPγS autoradiographic assay of G-protein coupled receptor function.


Asunto(s)
Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Hipocampo/efectos de los fármacos , Lisofosfolípidos/farmacología , Corteza Prefrontal/efectos de los fármacos , Radioisótopos de Azufre/metabolismo , Animales , Autorradiografía , Hipocampo/metabolismo , Masculino , Corteza Prefrontal/metabolismo , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley
3.
Sci Rep ; 10(1): 4366, 2020 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-32127599

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
J Neurochem ; 108(6): 1423-33, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19183263

RESUMEN

Bilateral olfactory bulbectomy in the rat (OBX) induces behavioral, neurochemical, and structural abnormalities similar to those observed in human depression that are normalized after chronic, but not acute, treatment with antidepressants. In our study, OBX animals exhibited significant increases in both CB(1) receptor density ([(3)H]CP55490 binding) and functionality (stimulation of [(35)S]GTPgammaS binding by the cannabinoid (CB) agonist WIN 55212-2) at the prefrontal cortex (PFC). After chronic treatment with fluoxetine (10 mg/kg/day, 14 days, s.c.), OBX-induced hyperactivity in the open-field test was fully abolished. Interestingly, chronic fluoxetine fully reversed the enhanced CB(1)-receptor signaling in PFC observed following OBX. The CB agonist Delta(9)-tetrahydrocannabinol (5 mg/kg, i.p., 1 day) did not produce any behavioral effect in sham-operated animals but returned locomotor activity to control values in OBX rats. As both acute administration of Delta(9)-tetrahydrocannabinol and chronic fluoxetine elicited a similar behavioral effect in the OBX rat, it is not unlikely that the regionally selective enhancement of CB(1) receptor-signaling in the PFC could be related with the altered OBX behavior. Our findings reinforce the utility of this animal model to further investigating the implication of the endocannabinoid system in the modulation of emotional processes and its potential role in the adaptive responses to chronic antidepressants.


Asunto(s)
Antidepresivos de Segunda Generación/administración & dosificación , Depresión/patología , Fluoxetina/administración & dosificación , Corteza Prefrontal/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Autorradiografía , Ciclohexanoles/metabolismo , Depresión/tratamiento farmacológico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Dronabinol/análogos & derivados , Dronabinol/farmacología , Esquema de Medicación , Conducta Exploratoria/efectos de los fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Inmunosupresores/metabolismo , Isótopos/metabolismo , Masculino , Bulbo Olfatorio/lesiones , Corteza Prefrontal/metabolismo , Unión Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
5.
Sci Rep ; 9(1): 19344, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31852967

RESUMEN

The non-selective activation of central and peripheral opioid receptors is a major shortcoming of currently available opioids. Targeting peripheral opioid receptors is a promising strategy to preclude side effects. Recently, we showed that fentanyl-derived µ-opioid receptor (MOR) agonists with reduced acid dissociation constants (pKa) due to introducing single fluorine atoms produced injury-restricted antinociception in rat models of inflammatory, postoperative and neuropathic pain. Here, we report that a new double-fluorinated compound (FF6) and fentanyl show similar pKa, MOR affinity and [35S]-GTPγS binding at low and physiological pH values. In vivo, FF6 produced antinociception in injured and non-injured tissue, and induced sedation and constipation. The comparison of several fentanyl derivatives revealed a correlation between pKa values and pH-dependent MOR activation, antinociception and side effects. An opioid ligand's pKa value may be used as discriminating factor to design safer analgesics.


Asunto(s)
Analgésicos Opioides/efectos adversos , Analgésicos Opioides/síntesis química , Analgésicos Opioides/química , Animales , Diseño de Fármacos , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Ligandos , Masculino , Nocicepción/efectos de los fármacos , Piperidinas/efectos adversos , Piperidinas/síntesis química , Piperidinas/química , Ratas Wistar , Receptores Opioides mu/metabolismo
6.
Neuropharmacology ; 55(8): 1391-6, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18809415

RESUMEN

5-HT1A receptor-mediated signalling in rat brain was evaluated after chronic administration (14 days; s.c.) of the selective serotonin reuptake inhibitor (SRRI) fluoxetine (10 mg/kg/day) alone, or in combination with the 5-HT1A receptor antagonist WAY100635 (0.1 mg/kg/day). The density of 5-HT1A binding sites was unchanged following fluoxetine, WAY100635, or the combination of fluoxetine and WAY100635. However, the net stimulation of [35S]GTPgammaS binding induced by the 5-HT1A agonist 8-OH-DPAT was significantly attenuated in dorsal raphe nucleus (DRN), but not in hippocampus, after chronic fluoxetine. Moreover, depending of the area analysed, the basal binding of [35S]GTPgammaS was differentially affected by this treatment: increased in DRN and decreased in hippocampal dentate gyrus. Interestingly, the changes in [35S]GTPgammaS basal binding and on 5-HT1A receptors functionality were prevented by the concomitant administration of WAY100635. The inhibition of dorsal raphe firing by 8-OH-DPAT was also attenuated in fluoxetine-treated rats (ED50 = 2.12 +/- 0.32 microg/kg and 4.34 +/- 0.09 microg/kg, for vehicle and fluoxetine respectively), an effect which was also prevented by the concomitant administration of WAY100635 (ED50 = 2.10 +/- 0.58 microg/kg). Chronic administration of WAY100635 alone did not affect the 5-HT1A receptor-induced stimulation of [35S]GTPgammaS binding, nor the 8-OH-DPAT-induced inhibition of 5-HT neuron firing. These results demonstrate that the concomitant blockade of 5-HT1A receptors when administering fluoxetine prevents those adaptive changes of 5-HT1A receptor function associated with the chronic administration of this antidepressant. These findings could be relevant from the therapeutic point of view, and further support the potential benefit of treatments with a SSRI/5-HT1A receptor antagonist combination.


Asunto(s)
Encéfalo/efectos de los fármacos , Fluoxetina/farmacología , Piperazinas/farmacología , Piridinas/farmacología , Receptor de Serotonina 5-HT1A/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Antagonistas de la Serotonina/farmacología , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacocinética , Animales , Autorradiografía , Encéfalo/metabolismo , Interacciones Farmacológicas , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Unión Proteica/efectos de los fármacos , Radioisótopos/metabolismo , Ratas , Ratas Wistar , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Agonistas de Receptores de Serotonina/metabolismo , Factores de Tiempo
7.
Pain ; 159(11): 2277-2284, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29994988

RESUMEN

Recently, (±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide (NFEPP), a newly designed µ-opioid receptor (MOR) agonist with a low pKa, has been shown to produce injury-restricted analgesia in models of inflammatory and postoperative pain, without exhibiting typical opioid side effects. Here, we investigated MOR binding of NFEPP in brain and dorsal root ganglia, pH in injured tissues, and the analgesic efficacy of NFEPP compared with fentanyl in a chronic constriction injury model of neuropathic pain, and in the acetic acid-induced abdominal writhing assay in rats. Binding experiments revealed significantly lower affinity of NFEPP compared with fentanyl at pH 7.4. In vivo, pH significantly dropped both at injured nerves after chronic constriction injury and in the abdominal cavity after acetic acid administration. Intravenous NFEPP as well as fentanyl dose-dependently diminished neuropathy-induced mechanical and heat hypersensitivity, and acetic acid-induced abdominal constrictions. In both models, NFEPP-induced analgesia was fully reversed by naloxone methiodide, a peripherally restricted opioid receptor antagonist, injected at the nerve injury site or into the abdominal cavity. Our results indicate that NFEPP exerts peripheral opioid receptor-mediated analgesia exclusively in damaged tissue in models of neuropathic and abdominal pain.


Asunto(s)
Dolor Abdominal/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Piperidinas/uso terapéutico , Receptores Opioides mu/agonistas , Animales , Encéfalo/citología , Membrana Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Ganglios Espinales/citología , Concentración de Iones de Hidrógeno , Hiperalgesia/tratamiento farmacológico , Masculino , Dimensión del Dolor/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Ratas , Ratas Wistar , Receptores Opioides mu/metabolismo , Estadísticas no Paramétricas
8.
Sci Rep ; 8(1): 8965, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29895890

RESUMEN

Novel pain killers without adverse effects are urgently needed. Opioids induce central and intestinal side effects such as respiratory depression, sedation, addiction, and constipation. We have recently shown that a newly designed agonist with a reduced acid dissociation constant (pKa) abolished pain by selectively activating peripheral µ-opioid receptors (MOR) in inflamed (acidic) tissues without eliciting side effects. Here, we extended this concept in that pKa reduction to 7.22 was achieved by placing a fluorine atom at the ethylidene bridge in the parental molecule fentanyl. The new compound (FF3) showed pH-sensitive MOR affinity, [35S]-GTPγS binding, and G protein dissociation by fluorescence resonance energy transfer. It produced injury-restricted analgesia in rat models of inflammatory, postoperative, abdominal, and neuropathic pain. At high dosages, FF3 induced sedation, motor disturbance, reward, constipation, and respiratory depression. These results support our hypothesis that a ligand's pKa should be close to the pH of injured tissue to obtain analgesia without side effects.


Asunto(s)
Analgésicos , Diseño de Fármacos , Neuralgia/tratamiento farmacológico , Dolor Postoperatorio/tratamiento farmacológico , Receptores Opioides mu/agonistas , Analgésicos/síntesis química , Analgésicos/química , Analgésicos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Concentración de Iones de Hidrógeno , Masculino , Estructura Molecular , Muramidasa , Neuralgia/genética , Neuralgia/metabolismo , Neuralgia/patología , Dolor Postoperatorio/genética , Dolor Postoperatorio/metabolismo , Dolor Postoperatorio/patología , Fragmentos de Péptidos , Ratas , Ratas Wistar , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo
9.
Elife ; 62017 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-28673386

RESUMEN

Novel painkillers are urgently needed. The activation of opioid receptors in peripheral inflamed tissue can reduce pain without central adverse effects such as sedation, apnoea, or addiction. Here, we use an unprecedented strategy and report the synthesis and analgesic efficacy of the standard opioid morphine covalently attached to hyperbranched polyglycerol (PG-M) by a cleavable linker. With its high-molecular weight and hydrophilicity, this conjugate is designed to selectively release morphine in injured tissue and to prevent blood-brain barrier permeation. In contrast to conventional morphine, intravenous PG-M exclusively activated peripheral opioid receptors to produce analgesia in inflamed rat paws without major side effects such as sedation or constipation. Concentrations of morphine in the brain, blood, paw tissue, and in vitro confirmed the selective release of morphine in the inflamed milieu. Thus, PG-M may serve as prototype of a peripherally restricted opioid formulation designed to forego central and intestinal side effects.


Asunto(s)
Analgesia/métodos , Analgésicos/farmacología , Analgésicos/farmacocinética , Glicerol/farmacología , Glicerol/farmacocinética , Morfina/farmacología , Morfina/farmacocinética , Polímeros/farmacología , Polímeros/farmacocinética , Analgésicos/química , Estructuras Animales/química , Animales , Glicerol/química , Interacciones Hidrofóbicas e Hidrofílicas , Peso Molecular , Morfina/química , Polímeros/química , Ratas
10.
Neurosci Lett ; 515(2): 181-6, 2012 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-22480692

RESUMEN

Serotonin and noradrenaline reuptake inhibitors have shown to produce antinociceptive effects in several animal models of neuropathic pain. In the present work, we have analyzed the density of brain and spinal serotonin and noradrenaline transporters (5-HTT and NAT) in a rat model of neuropathic pain, the spinal nerve ligation (SNL). Quantitative autoradiography revealed a significant decrease in the density of 5-HTT ([(3)H]citalopram binding) at the level of the lumbar spinal cord following 2 weeks of neuropathic surgery (lamina V, -40%: 6.01±0.64 nCi/mg tissue in sham-animals vs 3.59±1.56 in SNL-animals; lamina X, -30%: 9.10±2.00 vs 6.40±1.93 and lamina IX, -22%: 12.01±2.41 vs 9.42±1.58). By contrast, NAT density ([(3)H]nisoxetine binding) was significantly increased (lamina I-II, +34%: 2.20±0.45 vs 2.96±0.65; lamina V, +57%: 1.34±0.28 vs 2.11±0.66; and lamina IX, +58%: 2.39±0.71 vs 3.78±1.10). At supraspinal structures, SNL induced adaptive changes only in the density of 5-HTT (septal nuclei, +33%: 10.18±2.03 vs 13.53±1.14; CA3 field of hippocampus, +18%: 6.94±1.01 vs 8.21±0.81; paraventricular thalamic nucleus, +21%: 15.18±1.88 vs 18.35±2.08; lateral hypothalamic area, +40%: 12.68±1.90 vs 17.8±2.55; ventromedial hypothalamic nuclei, +19%: 7.16±0.92 vs 8.55±0.40; and dorsal raphe nucleus, +15%: 35.22±3.88 vs 40.68±3.11). Thus, we demonstrate, in the SNL model of neuropathic pain, the existence of opposite changes in the spinal expression of 5-HTT (down-regulation) and NAT (up-regulation), and the presence of supraspinal adaptive changes (up-regulation) only on 5-HTT density. These findings may help understanding the pathogeny of neuropathic pain and the differential analgesic action of antidepressants targeting 5-HTT and/or NAT transporters.


Asunto(s)
Encéfalo/metabolismo , Hiperalgesia/metabolismo , Neuralgia/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Médula Espinal/metabolismo , Animales , Regulación hacia Abajo , Masculino , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley , Nervios Espinales/lesiones , Nervios Espinales/metabolismo , Regulación hacia Arriba
11.
PLoS One ; 7(7): e42111, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22848724

RESUMEN

Typically, approach behaviour is displayed in the context of moving towards a desired goal, while avoidance behaviour is displayed in the context of moving away from threatening or novel stimuli. In the current research, we detected three sub-populations of C57BL/6J mice that spontaneously responded with avoiding, balancing or approaching behaviours in the presence of the same conflicting stimuli. While the balancing animals reacted with balanced responses between approach and avoidance, the avoiding or approaching animals exhibited inhibitory or advance responses towards one of the conflicting inputs, respectively. Individual differences in approach and avoidance motivation might be modulated by the normal variance in the level of functioning of different systems, such as endocannabinoid system (ECS). The present research was aimed at analysing the ECS involvement on approach and avoidance behavioural processes. To this aim, in the three selected sub-populations of mice that exhibited avoiding or balancing or approaching responses in an approach/avoidance Y-maze we analysed density and functionality of CB(1) receptors as well as enzyme fatty acid amide hydrolase activity in different brain regions, including the networks functionally responsible for emotional and motivational control. The main finding of the present study demonstrates that in both approaching and avoiding animals higher CB(1) receptor density in the amygdaloidal centro-medial nuclei and in the hypothalamic ventro-medial nucleus was found when compared with the CB(1) receptor density exhibited by the balancing animals. The characterization of the individual differences to respond in a motivationally based manner is relevant to clarify how the individual differences in ECS activity are associated with differences in motivational and affective functioning.


Asunto(s)
Reacción de Prevención , Motivación/fisiología , Receptor Cannabinoide CB1/metabolismo , Amidohidrolasas/metabolismo , Animales , Autorradiografía , Encéfalo/metabolismo , Encéfalo/fisiología , Ciclohexanoles/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL
12.
Toxicol Lett ; 207(1): 1-6, 2011 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21889975

RESUMEN

It is now well established that nicotine adversely affects the integrity of the blood-brain barrier (BBB). In contrast, nicotine has been reported to increase the transendothelial electrical resistance (TEER) of CaCo2 colon cancer cells. In the present study, the effects of nicotine upon the TEER and sucrose permeability of ECV304/C6 co-cultures and, for comparative purposes, CaCo2 cells has been investigated. Neither ECV304 nor C6 cells were found to express measurable membrane levels of nicotinic acetylcholine receptors, as assessed by [³H]-epibatidine binding. Nicotine treatment (0.01-1 µM) for up to 48 h had little or no effect upon the TEER or sucrose permeability of either ECV304/C6 co-cultures or CaCo2 cells. It is concluded that in contrast to the situation for the BBB, ECV304 cells lack nicotinic acetylcholine receptors and the barrier properties of ECV304/C6 co-cultures are not affected to any important extent by nicotine. This study underlines the conclusions made by other authors that the ECV304/C6 co-culture system is of limited validity as a model of the BBB.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Sacarosa/farmacología , Animales , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Células CACO-2 , Permeabilidad de la Membrana Celular/efectos de los fármacos , Técnicas de Cocultivo , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Inmunohistoquímica , Proteínas de la Membrana/metabolismo , Ocludina , Fosfoproteínas/metabolismo , Piridinas/farmacología , Ratas , Receptores Nicotínicos/metabolismo , Proteína de la Zonula Occludens-1
13.
Curr Pharm Des ; 17(5): 521-33, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21375480

RESUMEN

Over the past five decades, the pharmacological treatment of depression has been based on the pathophysiological hypothesis of a deficiency in monoamines, mainly serotonin and noradrenaline. Antidepressant prescribed today, all of them designed to enhance central monoaminergic tone, present several important limitations, including a 2-5 weeks response lag and also a limited clinical efficacy. As it is increasingly evident that the abnormalities associated to depression go beyond monoamines, the development of better antidepressants will depend on the identification and understanding of new cellular targets. In this regard, much evidence supports a role for cellular and molecular mechanisms of neuroplasticity, including neurotrophic inputs, in mood disorders, in parallel with the biological features associated to stress conditions. In order to illustrate the possible relevance of neuroplasticity-related pathways for the therapy of depressive states, we here review the biological evidence supporting some therapeutic strategies in a very initial phase of development (modulation of the Wnt/GSK-3ß/ß-catenin pathway, potentiation of endocannabinoid activity, agonism of 5-HT(4) receptors), which involve modulation of downstream mechanisms and neuroplasticity circuits. These strategies also show the existence of mixed mechanisms of action, constituting a nexus between the "classic" aminergic theory and the "new" neuroplasticity hypothesis.


Asunto(s)
Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Encéfalo/efectos de los fármacos , Depresión/tratamiento farmacológico , Descubrimiento de Drogas/métodos , Plasticidad Neuronal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Encéfalo/fisiología , Moduladores de Receptores de Cannabinoides/fisiología , Depresión/fisiopatología , Humanos , Modelos Biológicos , Plasticidad Neuronal/fisiología , Receptores de Serotonina 5-HT4/fisiología , Agonistas del Receptor de Serotonina 5-HT4/farmacología , Agonistas del Receptor de Serotonina 5-HT4/uso terapéutico , Transducción de Señal/fisiología
14.
Exp Neurol ; 224(1): 37-47, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20353772

RESUMEN

There is now a large volume of data indicating that compounds activating cannabinoid CB(1) receptors, either directly or indirectly by preventing the breakdown of endogenous cannabinoids, can protect against neuronal damage produced by a variety of neuronal "insults". Given that such neurodegenerative stimuli result in increased endocannabinoid levels and that animals with genetic deletions of CB(1) receptors are more susceptible to the deleterious effects of such stimuli, a case can be made for an endogenous neuroprotective role of endocannabinoids. However, this is an oversimplification of the current literature, since (a) compounds released together with the endocannabinoids can contribute to the neuroprotective effect; (b) other proteins, such as TASK-1 and PPARalpha, are involved; (c) the CB(1) receptor antagonist/inverse agonist rimonabant has also been reported to have neuroprotective properties in a number of animal models of neurodegenerative disorders. Furthermore, the CB(2) receptor located on peripheral immune cells and activated microglia are potential targets for novel therapies. In terms of the clinical usefulness of targeting the endocannabinoid system for the treatment of neurodegenerative disorders, data are emerging, but important factors to be considered are windows of opportunity (for acute situations such as trauma and ischemia) and the functionality of the target receptors (for chronic neurodegenerative disorders such as Alzheimer's disease).


Asunto(s)
Encéfalo/metabolismo , Moduladores de Receptores de Cannabinoides/metabolismo , Citoprotección/fisiología , Endocannabinoides , Neuronas/metabolismo , Animales , Humanos , Degeneración Nerviosa/metabolismo , Fármacos Neuroprotectores/metabolismo , Receptores de Cannabinoides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA