Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Neurosci Biobehav Rev ; 163: 105748, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38857667

RESUMEN

Corticotropin-releasing factor (CRF, corticoliberin) is a neuromodulatory peptide activating the hypothalamic-pituitary-adrenal (HPA) axis, widely distributed in the central nervous system (CNS) in mammals. In addition to its neuroendocrine effects, CRF is essential in regulating many functions under physiological and pathophysiological conditions through CRF1 and CRF2 receptors (CRF1R, CRF2R). This review aims to present selected examples of the diverse and sometimes opposite effects of CRF and its receptor ligands in various pathophysiological states, including stress/anxiety, depression, and processes associated with brain injury. It seems interesting to draw particular attention to the fact that CRF and its receptor ligands exert different effects depending on the brain structures or subregions, likely stemming from the varied distribution of CRFRs in these regions and interactions with other neurotransmitters. CRFR-mediated region-specific effects might also be related to brain site-specific ligand binding and the associated activated signaling pathways. Intriguingly, different types of CRF molecules can also influence the diverse actions of CRF in the CNS.

2.
Sci Rep ; 12(1): 19015, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36348016

RESUMEN

Eleven multiple analogs of bradykinin-a peptide that is a natural ligand of B1 and B2 receptors but does not bind or activate the B1 receptor unless Arg9 is removed from the sequence by the action of carboxypeptidase N-were synthesized. Their biological activity was examined on T-REx cell lines expressing B1 or B2 receptors using the intracellular IP1 assay. The mRNA expression of B1R and B2R in the lysate of tumor cell lines, e.g., U87-MG (human astrocytoma), SHP-77 (human small cell lung cancer), and H4 (human brain glioma), was determined. For five B1R antagonists, adsorption at the liquid/solid interface (Au nanoparticles (AuNPs) served as the solid surface) was discussed in terms of the vibrations of molecular fragments (structural factors) responsible for the biological properties of these analogs.


Asunto(s)
Bradiquinina , Nanopartículas del Metal , Humanos , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B2/genética , Oro , Factores de Transcripción
3.
Pharmacol Biochem Behav ; 219: 173452, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36030890

RESUMEN

There is still no effective treatment for central nervous system (CNS) pathologies, including cerebral ischemia, neurotrauma, and neurodegenerative diseases in which the Glu/GABA balance is disturbed with associated excitotoxicity. It is thus important to search for new efficacious therapeutic strategies. Preclinical studies on the role of metabotropic glutamate receptors (mGluRs) in neuroprotection conducted over the years show that these receptors may have therapeutic potential in these CNS disorders. However, clinical trials, especially for treating Parkinson's disease, have been unsatisfactory. This review focuses on the specific role of group III mGluRs in neuroprotection in experimental in vitro and in vivo models of excitotoxicity/neurotoxicity using neurotoxins as well as ischemia, traumatic brain injury, and neurodegenerative diseases such as Parkinson's disease, Alzheimer's diseases, and multiple sclerosis. The review highlights recent preclinical studies in which group III mGluR ligands (especially those acting at mGluR4 or mGluR7) were administered after damage, thus emphasizing the importance of the therapeutic time window in the treatment of ischemic stroke and traumatic brain injury. From a clinical standpoint, the review also highlights studies using group III mGluR agonists with favorable neuroprotective efficacy (histological and functional) in experimental ischemic stroke, including healthy normotensive and-hypertensive rats. This review also summarizes possible mechanisms underlying the neuroprotective activity of the group III mGluR ligands, which may be helpful in developing more effective and safe therapeutic strategies. Therefore, to fully assess the role of these receptors in neuroprotection, it is necessary to uncover new selective ligands, primarily those stimulating mGlu4 and mGlu7 receptors.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Accidente Cerebrovascular Isquémico , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Receptores de Glutamato Metabotrópico , Animales , Neuroprotección , Ratas , Receptores de Glutamato Metabotrópico/agonistas
4.
J Med Chem ; 64(12): 8410-8422, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34110823

RESUMEN

The article describes the application of the alanine-scanning technique used in combination with Raman, surface-enhanced Raman, attenuated total reflection Fourier transform infrared, and surface-enhanced infrared absorption (SEIRA) spectroscopies, which allowed defining the role of individual amino acid residues in the C-terminal 6-14 fragment of the bombesin chain (BN6-14) on the path of its adsorption on the surface of Ag (AgNPs) and Au nanoparticles (AuNPs). A reliable analysis of the SEIRA spectra of these peptides was possible, thanks to a curve fitting of these spectra. By combining alanine-scanning with biological activity studies using cell lines overexpressing bombesin receptors and the intracellular inositol monophosphate assay, it was possible to determine which peptide side chains play a significant role in binding a peptide to membrane-bound G protein-coupled receptors (GPCRs). Based on the analysis of spectral profiles and bioactivity results, conclusions for the specific peptide-metal and peptide-GPCR interactions were drawn and compared.


Asunto(s)
Bombesina/química , Bombesina/metabolismo , Nanopartículas del Metal/química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Receptores de Bombesina/metabolismo , Adsorción , Bombesina/genética , Oro/química , Células HEK293 , Humanos , Mutagénesis , Mutación , Fragmentos de Péptidos/genética , Unión Proteica , Plata/química , Espectrofotometría Infrarroja , Espectrometría Raman
5.
Neuropeptides ; 88: 102162, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34062382

RESUMEN

Corticotropin releasing factor (CRF) is a neuropeptide widely distributed in the brain as a hormonal modulator and neurotransmitter. The best known behavioral function of CRF is activation of stress and anxiety via the hypothalamus and limbic structures but the role of CRF in the cortex is still poorly understood. Our previous studies have shown anxiolytic-like effects of high doses of CRF injected into the Fr2 frontal cortex and involvement of CRF1 receptors (R) in that effect. These results seemed to be controversial as most other studies suggested anxiogenic and not anxiolytic effects of CRF1R stimulation. Since stress is associated with adrenergic system, in the present study, we focused on participation of alpha1 and alpha2 or beta adrenergic receptors in the anxiolytic-like effect of CRF. Moreover, we verified whether these effects of CRF in the Fr2 were really connected with CRF1R. Male Wistar rats were bilaterally microinjected with CRF in a dose of 0.2 µg/1 µl/site or with the specific agonist of CRF1R, stressin 1 (0.2-0.0125 µg/1 µl/site) into the Fr2 area. The elevated plus maze (EPM) test was performed 30 min later to assess the anxiolysis. An involvement of noradrenergic receptors in the CRF induced anxiolytic-like effect in the Fr2 was studied by pretreatment with the alpha1 antagonist prazosin, alpha2 agonist clonidine, alpha2 antagonist RS 79948 or beta antagonist propranolol, 20-30 min before CRF. The influence on anxiety was assessed in the EPM test. The results show that anxiolytic behavior after CRF microinjection into the Fr2 area seems to be mainly connected with the CRF1R activation because a similar effect was observed after stressin 1 administration and it was blocked by CRF1R antagonist. The results observed after administration of noradrenergic ligands indicated that anxiolytic effects of CRF in the Fr2 engaged the alpha1 and alpha2 adrenergic receptors but not beta receptors.


Asunto(s)
Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Hormona Liberadora de Corticotropina/farmacología , Lóbulo Frontal/efectos de los fármacos , Animales , Hormona Liberadora de Corticotropina/administración & dosificación , Isoquinolinas/farmacología , Naftiridinas/farmacología , Ratas , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos
6.
Artículo en Inglés | MEDLINE | ID: mdl-33991587

RESUMEN

There is currently no effective treatment either for neurological illnesses (ischemia and neurodegenerative diseases) or psychiatric disorders (depression), in which the Glu/GABA balance is disturbed and accompanied by significant excitotoxicity. Therefore, the search for new and effective therapeutic strategies is imperative for these disorders. Studies conducted over the last several years indicate that the neuropeptide Y (NPY)-ergic system may be a potential therapeutic target for neuroprotective or antidepressant compounds. This review focuses on the neuroprotective roles of Y2 and Y5 receptors (YRs) in neurological disorders such as ischemia, Alzheimer's disease, Parkinson's disease, Huntington's disease, and in psychiatric disorders such as depression. It summarizes current knowledge on the possible mechanisms underlying the neuroprotective or antidepressant-like actions of Y2R and Y5R ligands. The review also discusses ligands acting at Y2R and Y5R and their limitations as in vivo pharmacological tools. The results from the preclinical studies discussed here may be useful in developing effective therapeutic strategies to treat neurological diseases on the one hand and psychiatric disorders on the other, and may pave the way for the development of novel Y2R and Y5R ligands as candidate drugs for the treatment of these diseases.


Asunto(s)
Descubrimiento de Drogas , Neuropéptido Y/farmacología , Neuroprotección/efectos de los fármacos , Receptores de Neuropéptido Y/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/prevención & control , Animales , Depresión/tratamiento farmacológico , Depresión/prevención & control , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/prevención & control , Isquemia/tratamiento farmacológico , Isquemia/prevención & control , Ratones , Sinapsis/metabolismo
7.
Int J Mol Sci ; 21(13)2020 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-32640759

RESUMEN

Clinical and preclinical studies indicate that zinc (Zn) is an essential factor in the development and treatment of major depressive disorder (MDD). Conventional monoamine-based antidepressants mobilize zinc in the blood and brain of depressed patients as well as rodents. N-methyl-D-aspartate acid receptor (NMDAR) antagonists exhibit antidepressant-like activity. However, not much is known about the antidepressant efficacy of NMDAR antagonists in zinc-deficient (ZnD) animals. We evaluated the antidepressant-like activity of two NMDAR antagonists (ketamine; global NMDAR antagonist and Ro 25-6981 (Ro); selective antagonist of the GluN2B NMDAR subunit) in ZnD rats using the forced swim test (FST) and sucrose intake test (SIT). A single dose of either Ro 25-6981 or ketamine normalized depressive-like behaviors in ZnD rats; however, Ro was effective in both tests, while ketamine was only effective in the FST. Additionally, we investigated the mechanism of antidepressant action of Ro at the molecular (analysis of protein expression by Western blotting) and anatomical (density of dendritic spines by Golgi Cox-staining) levels. ZnD rats exhibited decreased phosphorylation of the p70S6K protein, and enhanced density of dendritic spines in the prefrontal cortex (PFC) compared to control rats. The antidepressant-like activity of Ro was associated with the increased phosphorylation of p70S6K and ERK in the PFC. In summary, single doses of the NMDAR antagonists ketamine and Ro exhibited antidepressant-like activity in the ZnD animal model of depression. Animals were only deprived of Zn for 4 weeks and the biochemical effects of Zn deprivation and Ro were investigated in the PFC and hippocampus. The shorter duration of dietary Zn restriction may be a limitation of the study. However, future studies with longer durations of dietary Zn restriction, as well as the investigation of multiple brain structures, are encouraged as a supplement to this study.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Trastorno Depresivo Mayor/tratamiento farmacológico , Dieta/efectos adversos , Ketamina/farmacología , Fenoles/farmacología , Piperidinas/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Zinc/deficiencia , Analgésicos/farmacología , Animales , Conducta Animal , Trastorno Depresivo Mayor/etiología , Trastorno Depresivo Mayor/patología , Trastorno Depresivo Mayor/psicología , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo
8.
Neuropharmacology ; 174: 107866, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31785263

RESUMEN

The activity of an allosteric agonist of muscarinic M1 receptor, VU0357017, and a positive allosteric modulator (PAM) of M5 receptor, VU0238429, were investigated alone or in combination with the mGlu2 receptor PAM, LY487379 using the following behavioural tests: prepulse inhibition (PPI), novel object recognition (NOR), and spatial delayed alternation (SDA). VU0357017 (10 and 20 mg/kg) and VU0238429 (5 and 10 mg/kg) reversed deficits in PPI while VU0238429 (2.5 and 5 mg/kg) was effective in SDA. The simultaneous administration of subeffective doses of M1 or M5 activators (5, 1, or 0.25 mg/kg) with LY487379 (0.5 mg/kg) induced the same effect as that observed for the active dose of each compound. Selective M1 or M5 receptor blockers antagonized the effect exerted by these combinations, and pharmacokinetic studies confirmed independent transport through the blood-brain barrier. The expression of both receptors (M1 and M5) was established in brain structures involved in cognition (neocortex, hippocampus, and entorhinal cortex) in both the rat and the mouse brains by immunofluorescence staining. Specifically, double neuronal staining of mGlu2-M1 and mGlu2-M5 receptors was observed in many areas of the rat brain, while the number of double-stained mGlu2-M1 receptors was moderate in the mouse brain with no mGlu2-M5 colocalization. Finally, the combined administration of subeffective doses of the compounds did not alter prolactin levels or motor coordination, in contrast to the compounds given alone at the highest dose or in combination with standard neuroleptics.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Maleato de Dizocilpina/toxicidad , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M5/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/toxicidad , Masculino , Ratones , Piridinas/farmacología , Ratas , Ratas Wistar , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M5/agonistas , Receptores de Glutamato Metabotrópico/agonistas , Sulfonamidas/farmacología
9.
ACS Chem Neurosci ; 10(8): 3454-3463, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31267743

RESUMEN

This paper discusses the biological and three-dimensional molecular structure of the novel, nonpeptide Y2R antagonist, SF-11 [N-(4-ethoxyphenyl)-4-(hydroxydiphenylmethyl)-1-piperidinecarbothioamide]. Pharmacokinetic studies in a rat model indicated that, following intraperitoneal dosing, SF-11 crossed the blood-brain barrier and was able to penetrate the brain, making it a suitable tool for behavioral studies. We showed for the first time that SF-11 decreased the immobility time in the forced swim test (FST) after acute peripheral administration (10 and 20 mg/kg), indicating that it has antidepressant potential. Inhibitors of the mitogen-activated protein kinase/extracellular signal regulated kinase (MAPK/ERK) and phosphatidylinositol 3-kinase (PI3K) signaling pathways blocked the anti-immobility effect of SF-11, suggesting that these pathways are involved in the antidepressant-like activity of SF-11 in the FST. The results of locomotor activity of rats indicate that the effects observed in the FST are specific and due to the antidepressant-like activity of SF-11. These findings provide further evidence for the antidepressant potential of Y2R antagonists. Also, the application of Fourier transform infrared absorption (FT-IR) and Raman spectroscopy (RS) methods combined with theoretical density functional theory (DFT) calculations allowed us to present the optimized spatial orientation of the investigated drug. Structural characterization of SF-11 based on vibrational spectroscopic data is of great importance and will aid in understanding its biological activity and pave the way for its development as a new antidepressant agent.


Asunto(s)
Antidepresivos/farmacología , Receptores de Neuropéptido Y/antagonistas & inhibidores , Animales , Conducta Animal/efectos de los fármacos , Masculino , Actividad Motora/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
10.
Prog Neuropsychopharmacol Biol Psychiatry ; 84(Pt A): 93-101, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29438731

RESUMEN

Our previous studies have shown that ACPT-I [(1S, 3R,4S)-1-aminocyclopentane-1,2,4-tricarboxylic acid], a blood-brain barrier permeable agonist of group III metabotropic glutamate (mGlu) receptors, was neuroprotective against middle cerebral artery occlusion/reperfusion (MCAO/R) in normotensive rats. Preclinical studies are typically performed on healthy animals, whereas stroke patients predominately exhibit comorbidities, such as hypertension; therefore, in the present study, we investigated the effect of ACPT-I in spontaneously hypertensive rats (SHR) after MCAO/R. We examined the potential neuroprotective action of ACPT-I (30 mg/kg) when administered during occlusion or reperfusion via the assessment of not only the brain infarction volume but also motor (CatWalk gait analysis and open field test) and sensorimotor (vibrissae-evoked forelimb-placing test) functions following MCAO/R. We determined that ACPT-I not only reduced the cortico-striatal infarction but also improved several gait parameters (run speed, run and stand durations, swing speed and stride length) and mobility when administered 30 min after the start of the occlusion or 30 min after the start of reperfusion. Moreover, the sensorimotor function was improved in hypertensive rats treated with ACPT-I during occlusion. In conclusion, the current findings provide further evidence for the neuroprotective effects of ACPT-I against ischemic damage. These findings may have clinical implications because hypertension is an important risk factor for ischemic stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Ciclopentanos/farmacología , Hipertensión Esencial/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Ácidos Tricarboxílicos/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Hipertensión Esencial/complicaciones , Hipertensión Esencial/patología , Marcha/efectos de los fármacos , Masculino , Distribución Aleatoria , Ratas Endogámicas SHR , Receptores de Glutamato Metabotrópico/agonistas , Sensación/efectos de los fármacos , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/patología
11.
Neuropharmacology ; 125: 333-342, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28802646

RESUMEN

Short and long acting NMDA receptor (NMDAR) antagonists exert their antidepressant-like effects by activating signaling pathways involved in the synthesis of synaptic proteins and formation of new synaptic connections in the prefrontal cortex (PFC) of rats. The blockade of the ERK pathway abolishes ketamine and Ro 25-6981 antidepressant potency. However, the role of ERK in the antidepressant-like activity of short acting NMDAR antagonists is still unclear. More puzzling is the fact that the precise role of ERK in the short and long lasting effects of long-acting NMDAR antagonists is unknown. In this study, we show that zinc, (Zn) a short-acting NMDAR antagonist evokes only transient ERK activation, which is observed 7 min after its administration in the PFC of rats. In contrast to Zn, the long acting NMDAR antagonist Ro 25-6981 produces persistent ERK activation lasting up to 24 h. Pretreatment with the MAPK/ERK inhibitor (U0126) totally abolished Zn and Ro 25-6981 antidepressant-like activities in the forced swim test in rats. However, when U0126 is administered 15 min after Zn or Ro 25-6981 both compounds maintain their short-lasting antidepressant-like activity. On the other hand, posttreatment with U0126 significantly attenuated the long lasting antidepressant-like activity of Ro 25-6981. These results indicate that the activation of ERK is crucial for the short- and long lasting antidepressant-like activity observed in the FST in rats.


Asunto(s)
Antidepresivos/farmacología , Ácido Aspártico/análogos & derivados , Trastorno Depresivo/tratamiento farmacológico , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Compuestos Organometálicos/farmacología , Fenoles/farmacología , Piperidinas/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Compuestos de Zinc/farmacología , Animales , Ácido Aspártico/farmacología , Butadienos/farmacología , Trastorno Depresivo/enzimología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/enzimología , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Sinaptosomas/efectos de los fármacos , Sinaptosomas/enzimología , Factores de Tiempo
12.
Neuroscience ; 344: 305-325, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28057538

RESUMEN

It was postulated that neuropeptide Y (NPY)-ergic system could be involved in the ischemic pathophysiology, however, the role of particular subtypes of NPY receptors (YRs) in neuroprotection against ischemia is still not well known. Therefore, we investigated the effect of NPY and YR ligands using in vitro and in vivo experimental ischemic stroke models. Our in vitro findings showed that NPY (0.5-1µM) and specific agonists of Y2R (0.1-1µM) and Y5R (0.5-1µM) but not that of Y1R produced neuroprotective effects against oxygen-glucose deprivation (OGD)-induced neuronal cell death, being also effective when given 30min after the end of OGD. The neuroprotective effects of Y2R and Y5R agonists were reversed by appropriate antagonists. Neuroprotection mediated by NPY, Y2R and Y5R agonists was accompanied by the inhibition of both OGD-induced calpain activation and glutamate release. Data from in vivo studies demonstrated that Y2R agonist (10µg/6µl; i.c.v.) not only diminished the infarct volume in rats subjected to transient middle cerebral artery occlusion (MCAO) but also improved selected gait parameters in CatWalk behavioral test, being also effective after delayed treatment. Moreover, we found that a Y5R agonist (10µg/6µl; i.c.v.) did not reduce MCAO-evoked brain damage but improved stride length, when it was given 30min after starting the occlusion. In conclusion, our studies indicate that Y5 and especially Y2 receptors may be promising targets for neuroprotection against ischemic damage.


Asunto(s)
Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptores de Neuropéptido Y/agonistas , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Calpaína/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Marcha/efectos de los fármacos , Marcha/fisiología , Glucosa/deficiencia , Ácido Glutámico/metabolismo , Ataque Isquémico Transitorio/tratamiento farmacológico , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/patología , Masculino , Ratones , Neuronas/metabolismo , Neuronas/patología , Neuroprotección/efectos de los fármacos , Neuroprotección/fisiología , Distribución Aleatoria , Ratas Sprague-Dawley , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/metabolismo
13.
Psychopharmacology (Berl) ; 234(4): 631-645, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27975125

RESUMEN

RATIONALE: It has recently been found that chronic treatment with the highly selective, brain-penetrating Y5 receptor antagonist, Lu AA33810 [N-[[trans-4-[(4,5-dihydro [1] benzothiepino[5,4-d] thiazol-2-yl) amino] cyclohexyl]methyl]-methanesulfonamide], produces antidepressant-like effects in the rat chronic mild stress model. OBJECTIVE: In the present study, we investigated the possible antidepressant-like activity of Lu AA33810 in rats subjected to glial ablation in the prefrontal cortex (PFC) by the gliotoxin L-AAA, which is an astroglial degeneration model of depression. RESULTS: We observed that Lu AA33810 administered intraperitoneally at a single dose of 10 mg/kg both reversed depressive-like behavioral changes in the forced swim test (FST) and prevented degeneration of astrocytes in the mPFC. The mechanism of antidepressant and glioprotective effects of Lu AA33810 has not been studied, so far. We demonstrated the contribution of the noradrenergic rather than the serotonergic pathway to the antidepressant-like action of Lu AA33810 in the FST. Moreover, we found that antidepressant-like effect of Lu AA33810 was connected with the influence on brain-derived neurotrophic factor (BDNF) protein expression. We also demonstrated the antidepressant-like effect of Lu AA33810 in the FST in rats which did not receive the gliotoxin. We found that intracerebroventricular injection of the selective MAPK/ERK inhibitor U0126 (5 µg/2 µl) and the selective PI3K inhibitor LY294002 (10 nmol/2 µl) significantly inhibited the anti-immobility effect of Lu AA33810 in the FST in rats, suggesting that MAPK/ERK and PI3K signaling pathways could be involved in the antidepressant-like effect of Lu AA33810. CONCLUSION: Our results indicate that Lu AA33810 exerts an antidepressant-like effect and suggest the Y5 receptors as a promising target for antidepressant therapy.


Asunto(s)
Antidepresivos/farmacología , Benzotiepinas/farmacología , Depresión/tratamiento farmacológico , Sulfonamidas/farmacología , Animales , Antidepresivos/uso terapéutico , Conducta Animal/efectos de los fármacos , Benzotiepinas/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cromonas/farmacología , Depresión/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Masculino , Morfolinas/farmacología , Neuropéptido Y/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Sulfonamidas/uso terapéutico , Natación
14.
Colloids Surf B Biointerfaces ; 149: 80-88, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27736725

RESUMEN

It has been suggested that the family of neuropeptide Y (NPY) peptides is a promising target for the neuroprotective therapy; therefore, knowledge of the structure of these biologically active compounds and their behavior at solid/liquid interface is important in order to design new analogues. Because there is still a lack of detailed information on the behavior of NPY and its mutated analogues at the solid/liquid interfaces, in this work surface-enhanced Raman spectroscopy (SERS) analysis was used to investigate NPY and its native NPY3-36, NPY13-36, and NPY22-36 and mutated acetyl-(Leu28,31)-NPY24-36C-terminal fragments, acting on Y2 receptors (Y2R), in order to determine their possible metal surface/molecule interactions. In these studies, colloidal gold nanoparticle surface served as a solid surface, whereas an aqueous solution was used as a liquid medium. The observed differences in the band intensities, wavenumbers, and widths allowed us to draw conclusions on an adsorption mode of NPY and on changes in this mode upon the shortening of the peptide chain and increase in solution pH (from pH 3 to pH 11). Briefly, three different species of Tyr were identified onto the colloidal gold surface depending upon the length of the peptide chain and solution pH. Tyrosine (TyrOH) is present in a basic medium. Tyrosinate (TyrO-) is present in an acidic solution, whereas phenoxyl radical (Tyr*) appears at neutral pH for peptides having relatively short peptide chain (acetyl-(Leu28,31)-NPY24-36). The elongation of the peptide chain partially (NPY13-36 and NPY22-36) or completely (NPY3-36 and NPY) protects the Tyr residue against conversion to the radical form.


Asunto(s)
Oro Coloide/química , Nanopartículas del Metal/química , Neuropéptido Y/química , Fenoles/química , Tirosina/química , Adsorción , Animales , Concentración de Iones de Hidrógeno , Microscopía de Fuerza Atómica , Modelos Moleculares , Mutación , Neuropéptido Y/genética , Soluciones , Espectrometría Raman , Propiedades de Superficie , Porcinos
15.
Neuropharmacology ; 102: 276-94, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26647070

RESUMEN

In the present study, we investigated the effect of ACPT-I [(1S, 3R,4S)-1-aminocyclopentane-1,2,4-tricarboxylic acid], a blood-brain-barrier permeable agonist of group III mGlu receptor, against oxygen-glucose deprivation (OGD)-evoked neuronal cell death in primary neuronal cell cultures and in the model of transient middle cerebral artery occlusion (MCAO) in rats. We found that ACPT-I (1-200 µM) in a concentration- and time-dependent way attenuated the OGD-induced neuronal cell damage, being also effective after a delayed application (30 min after OGD). The neuroprotective effects of ACPT-I were blocked by the group III mGlu receptor antagonist, (RS)-alpha-cyclopropyl-4-phosphonophenyl glycine (CPPG), and by the activator of cAMP-dependent PKA, 8-Bromo-cAMP, but not by an inhibitor of PI-3-K signaling pathway. Moreover, ACPT-I attenuated the OGD-induced calpain activity and glutamate release. In the in vitro study, we also demonstrated the neuroprotective potential of mGluR4 positive allosteric modulators (PAMs), PHCCC (30 µM) and VU0155041 (10 and 30 µM) and synergism in neuroprotective action of low concentrations of ACPT-I and mGluR4 PAMs suggesting an important role of mGluR4 activation in prevention of ischemic neuronal cell death. In the rat MCAO model, we demonstrated that ACPT-I (30 mg/kg) injected intraperitoneally either 30 min after starting MCAO or 30 min after beginning reperfusion not only diminished the infarction volume by about 30%, but also improved selected gait parameters (CatWalk analysis) and the mobility of animals in the open field test. In conclusion, our results indicate that ACPT-I may be not only neuroprotective against ischemic neuronal damage but may also diminish the postischemic functional deficits.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Ciclopentanos/uso terapéutico , Agonistas de Aminoácidos Excitadores/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Receptores de Glutamato Metabotrópico/agonistas , Accidente Cerebrovascular/tratamiento farmacológico , Ácidos Tricarboxílicos/uso terapéutico , Animales , Isquemia Encefálica/metabolismo , Células Cultivadas , Ciclopentanos/farmacología , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/farmacología , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/metabolismo , Ácidos Tricarboxílicos/farmacología
16.
Neuropharmacology ; 99: 517-26, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26297535

RESUMEN

The rapid antidepressant response to the N-methyl-D-aspartate (NMDA) receptor antagonists is mediated by activation of the mammalian target of the rapamycin (mTOR) signaling pathway, an increase in the synthesis of synaptic proteins and formation of new synapses in the prefrontal cortex (PFC) of rats. Zinc (Zn), which is a potent NMDA receptor antagonist, exerts antidepressant-like effects in screening tests and models of depression. We focused these studies in investigating whether activation of the mTOR signaling pathway is also a necessary mechanism of the antidepressant-like activity of Zn. We observed that a single injection of Zn (5 mg/kg) induced an increase in the phosphorylation of mTOR and p70S6K 30 min and 3 h after Zn treatment at time points when Zn produced also an antidepressant-like effect in the forced swim test (FST). Furthermore, Zn administered 3 h before the decapitation increased the level of brain derived neurotrophic factor (BDNF), GluA1 and synapsin I. An elevated level of GluA1 and synapsin I was still observed 24 h after the Zn treatment, although Zn did not produce any effects in the FST at that time point. We also observed that pretreatment with rapamycin (mTORC1 inhibitor), LY294002 (PI3K inhibitor), H-89 (PKA inhibitor) and GF109203X (PKC inhibitor) blocked the antidepressant-like effect of Zn in FST in rats and blocks Zn-induced activation of mTOR signaling proteins (analyzed 30 min after Zn administration). These studies indicated that the antidepressant-like activity of Zn depends on the activation of mTOR signaling and other signaling pathways related to neuroplasticity, which can indirectly modulate mTOR function.


Asunto(s)
Antidepresivos/farmacología , Trastorno Depresivo/tratamiento farmacológico , Corteza Prefrontal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Zinc/farmacología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Corteza Prefrontal/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Sinapsinas/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
17.
Neurochem Int ; 88: 110-23, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25576184

RESUMEN

Although numerous studies demonstrated a neuroprotective potency of unspecific group III mGluR agonists in in vitro and in vivo models of excitotoxicity, little is known about the protective role of group III mGlu receptor activation against neuronal cell injury evoked by ischemic conditions. The aim of the present study was to assess neuroprotective potential of the allosteric agonist of mGlu7 receptor, N,N'-Bis(diphenylmethyl)-1,2-ethanediamine dihydrochloride (AMN082) against oxygen-glucose deprivation (OGD)- and kainate (KA)-evoked neuronal cell damage in primary neuronal cultures, with special focus on its efficacy after delayed application. We demonstrated that in cortical neuronal cultures exposed to a 180 min OGD, AMN082 (0.01-1 µM) in a concentration- and time-dependent way attenuated the OGD-induced changes in the LDH release and MTT reduction assays. AMN082 (0.5 and 1 µM) produced also neuroprotective effects against KA-evoked neurotoxicity both in cortical and hippocampal cultures. Of particular importance was the finding that AMN082 attenuated excitotoxic neuronal injury after delayed application (30 min after OGD, or 30 min-1 h after KA). In both models of neurotoxicity, namely OGD- and KA-induced injury, the neuroprotective effects of AMN082 (1 µM) were reversed by the selective mGlu7 antagonist, 6-(4-Methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo[4,5-c]pyridin-4(5H)-one hydrochloride (MMPIP, 1 µM), suggesting the mGlu7-dependent mechanism of neuroprotective effects of AMN082. Next, we showed that AMN082 (0.5 and 1 µM) attenuated the OGD-induced increase in the number of necrotic nuclei as well inhibited the OGD-evoked calpain activation, suggesting the participation of these processes in the mechanism of AMN082-mediated protection. Additionally, we showed that protection evoked by AMN082 (1 µM) in KA model was connected with the inhibition of toxin-induced caspase-3 activity, and this effect was abolished by the mGlu7 receptor antagonist. The obtained results indicated that the activation of mGlu7 receptors may be a promising target for neuroprotection against ischemic and excitotoxic insults.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Glucosa/deficiencia , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Oxígeno/metabolismo , Receptores de Glutamato Metabotrópico/agonistas , Regulación Alostérica , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ácido Kaínico/toxicidad , Ratones , Neuronas/efectos de los fármacos
18.
J Colloid Interface Sci ; 437: 111-118, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25313473

RESUMEN

In this paper, we present spectroscopic studies of neuropeptide Y (NPY) and its native NPY(3-36), NPY(13-36), and NPY(22-36) and mutated acetyl-(Leu(28,31))-NPY(24-36)C-terminal fragments acting on Y2 receptor. Since there is some evidence for the correlation between the SERS patterns and the receptor binding ability, we performed a detailed analysis for these compounds at the metal/water interface using Raman spectroscopy (RS) and surface-enhanced Raman spectroscopy (SERS) methods. Many studies have suggested that interactions of this kind are crucial for a variety of biomedical and biochemical phenomena. The identification of amino acids in these peptide sequences by SERS allowed us to determine which molecular fragments were responsible for the interaction with the silver nanoparticle surface. Our findings demonstrated that in all of the investigated compounds, the NPY(32-36)C-terminal fragment (Thr(32)-Arg(33)-Gln(34)-Arg(35)-Tyr(36)NH2) was involved in the adsorption process onto metal substrate. The results of the present study suggest that the same molecular fragment interacts with the Y2 receptor, what proved the usefulness of the SERS method in the study of these biologically active compounds. The search for analogs acting on Y2 receptor may be important from the viewpoint of possible future clinical applications.


Asunto(s)
Neuropéptido Y/química , Receptores de Neuropéptido Y/efectos de los fármacos , Espectrometría Raman/métodos , Animales , Humanos , Neuropéptido Y/farmacología , Espectrofotometría Ultravioleta
19.
Behav Brain Res ; 273: 23-33, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25043733

RESUMEN

The glutamatergic predominance in the excitatory-inhibitory balance is postulated to be involved in the pathogenesis of depression. Such imbalance may be induced by astrocyte ablation which reduces glutamate uptake and increases glutamate level in the synaptic cleft. In the present study, we tried to ascertain whether astroglial degeneration in the prefrontal cortex could serve as an animal model of depression and whether inhibition of glutamatergic transmission by the mGluR5 antagonist MTEP could have antidepressant potential. Astrocytic toxins l-or dl-alpha-aminoadipic acid (AAA), 100µg/2µl, were microinjected, bilaterally into the rat medial prefrontal cortex (PFC) on the first and second day of experiment. MTEP (10mg/kg) or imipramine (30mg/kg) were administered on the fifth day. Following administration of MTEP or imipramine the forced swim test (FST) was performed for assessment of depressive-like behavior. The brains were taken out for analysis on day eight. The astrocytic marker, glial fibrillary acidic protein (GFAP) was quantified in PFC by Western blot method and by stereological counting of immunohistochemically stained sections. Both l-AAA and dl-AAA induced a significant increase in immobility time in the FST. This effect was reversed by imipramine, which indicates depressive-like effects of these toxins. A significant decrease in GFAP (about 50%) was found after l-AAA. Both the behavioral and GFAP level changes were prevented by MTEP injection. The obtained results indicate that the degeneration of astrocytes in the PFC by l-AAA may be a useful animal model of depression and suggest antidepressant potential of MTEP.


Asunto(s)
Antidepresivos/farmacología , Astrocitos/patología , Depresión/metabolismo , Depresión/patología , Piridinas/farmacología , Tiazoles/farmacología , Ácido 2-Aminoadípico/toxicidad , Animales , Astrocitos/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Actividad Motora/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/patología , Ratas , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5/antagonistas & inhibidores
20.
Neurotox Res ; 26(3): 255-73, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24842651

RESUMEN

A number of studies suggest that the ubiquitin-proteasome system (UPS) impairment may underlie neuronal death in Parkinson's disease. Celastrol is a neuroprotective agent with anti-inflammatory and antioxidant properties. The aim of this study was to determine whether celastrol may exert neuroprotective effects both in vitro and in vivo under conditions of the lactacystin-induced UPS inhibition. In the in vitro study, mouse primary cortical neurons and neuroblastoma SH-SY5Y cells were incubated with lactacystin for 48 h (2.5 and 10 µg/ml, respectively). The animal study was performed on male Wistar rats injected unilaterally with lactacystin (5 µg/2 µl) into the substantia nigra (SN) pars compacta. In the in vitro study, we did not found any protective effects of celastrol, given either in the pre- or co-treatment mode. Moreover, in the higher concentrations, celastrol itself reduced cell viability, and enhanced the lactacystin-induced cell death in both types of cells. In the in vivo study, none of the celastrol doses (0.3-3 mg/kg) attenuated the lactacystin-induced decrease in the level of dopamine (DA) and its metabolites or protected nigral dopaminergic neurons against the lactacystin-induced degeneration. The highest celastrol dose potentiated the lactacystin-induced decrease in the level of DA and its metabolites in the lesioned striatum, and accelerated the lactacystin-induced increase in the oxidative and total metabolism of DA. Moreover, when given alone, this dose of celastrol bilaterally decreased the number and/or density of dopaminergic neurons in the SN. Our results demonstrate that celastrol does not induce neuroprotective effects under conditions of UPS inhibition.


Asunto(s)
Acetilcisteína/análogos & derivados , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/metabolismo , Inhibidores de Proteasoma/toxicidad , Triterpenos/farmacología , Acetilcisteína/toxicidad , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Humanos , Masculino , Ratones , Neuroblastoma , Neuronas/efectos de los fármacos , Triterpenos Pentacíclicos , Ratas , Ratas Wistar , Sustancia Negra/efectos de los fármacos , Sustancia Negra/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...