Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.308
Filtrar
1.
Behav Pharmacol ; 35(4): 185-192, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38563661

RESUMEN

LY-404,039 is an orthosteric agonist at metabotropic glutamate 2 and 3 (mGlu 2/3 ) receptors, with a possible additional agonist effect at dopamine D 2 receptors. LY-404,039 and its pro-drug, LY-2140023, have previously been tested in clinical trials for psychiatric indications and could therefore be repurposed if they were shown to be efficacious in other conditions. We have recently demonstrated that the mGlu 2/3 orthosteric agonist LY-354,740 alleviated L-3,4-dihydroxyphenylalanine (L-DOPA)-induced abnormal involuntary movements (AIMs) in the 6-hydroxydopamine (6-OHDA)-lesioned rat without hampering the anti-parkinsonian action of L-DOPA. Here, we seek to take advantage of a possible additional D 2 -agonist effect of LY-404,039 and see if an anti-parkinsonian benefit might be achieved in addition to the antidyskinetic effect of mGlu 2/3 activation. To this end, we have administered LY-404,039 (vehicle, 0.1, 1 and 10 mg/kg) to 6-OHDA-lesioned rats, after which the severity of axial, limbs and oro-lingual (ALO) AIMs was assessed. The addition of LY-404,039 10 mg/kg to L-DOPA resulted in a significant reduction of ALO AIMs over 60-100 min (54%, P  < 0.05). In addition, LY-404,039 significantly enhanced the antiparkinsonian effect of L-DOPA, assessed through the cylinder test (76%, P  < 0.01). These results provide further evidence that mGlu 2/3 orthosteric stimulation may alleviate dyskinesia in PD and, in the specific case of LY-404,039, a possible D 2 -agonist effect might also make it attractive to address motor fluctuations. Because LY-404,039 and its pro-drug have been administered to humans, they could possibly be advanced to Phase IIa trials rapidly for the treatment of motor complications in PD.


Asunto(s)
Discinesia Inducida por Medicamentos , Levodopa , Oxidopamina , Trastornos Parkinsonianos , Receptores de Glutamato Metabotrópico , Animales , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/metabolismo , Ratas , Trastornos Parkinsonianos/tratamiento farmacológico , Masculino , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Oxidopamina/farmacología , Levodopa/farmacología , Antiparkinsonianos/farmacología , Aminoácidos/farmacología , Relación Dosis-Respuesta a Droga , Modelos Animales de Enfermedad , Ratas Sprague-Dawley , Compuestos Bicíclicos con Puentes/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Ratas Wistar
2.
Physiol Behav ; 280: 114564, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38657747

RESUMEN

Although salivation is essential during eating behavior, little is known about the brainstem centers that directly control the salivary glands. With regard to the inferior salivatory nucleus (ISN), the site of origin of the parasympathetic preganglionic cell bodies that innervate the parotid glands, previous anatomical studies have located it within the rostrodorsal medullary reticular formation. However, to date there is no functional data that shows the secretory nature of the somas grouped in this region. To activate only the somas and rule out the activation of the efferent fibers from and the afferent fibers to the ISN, in exp. 1, NMDA neurotoxin was administered to the rostrodorsal medullary region and the secretion of saliva was recorded during the following hour. Results showed an increased secretion of parotid saliva but a total absence of submandibular-sublingual secretion. In exp. 2, results showed that the hypersecretion of parotid saliva after NMDA microinjection was completely blocked by the administration of atropine (a cholinergic blocker) but not after administration of dihydroergotamine plus propranolol (α and ß-adrenergic blockers, respectively). These findings suggest that the somata of the rostrodorsal medulla are secretory in nature, controlling parotid secretion via a cholinergic pathway. The data thus functionally supports the idea that these cells constitute the ISN.


Asunto(s)
N-Metilaspartato , Glándula Parótida , Receptores de N-Metil-D-Aspartato , Animales , Masculino , Ratas , Glándula Parótida/metabolismo , Glándula Parótida/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , N-Metilaspartato/farmacología , N-Metilaspartato/metabolismo , Ratas Wistar , Salivación/efectos de los fármacos , Salivación/fisiología , Bulbo Raquídeo/metabolismo , Bulbo Raquídeo/efectos de los fármacos , Saliva/metabolismo , Agonistas de Aminoácidos Excitadores/farmacología , Atropina/farmacología , Propranolol/farmacología , Antagonistas Adrenérgicos beta/farmacología , Microinyecciones , Sialorrea
3.
J Med Chem ; 67(2): 1314-1326, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38170918

RESUMEN

Metabotropic glutamate (Glu) receptors (mGlu receptors) play a key role in modulating excitatory neurotransmission in the central nervous system (CNS). In this study, we report the structure-based design and pharmacological evaluation of densely functionalized, conformationally restricted glutamate analogue (1S,2S,3S)-2-((S)-amino(carboxy)methyl)-3-(carboxymethyl)cyclopropane-1-carboxylic acid (LBG30300). LBG30300 was synthesized in a stereocontrolled fashion in nine steps from a commercially available optically active epoxide. Functional characterization of all eight mGlu receptor subtypes showed that LBG30300 is a picomolar agonist at mGlu2 with excellent selectivity over mGlu3 and the other six mGlu receptor subtypes. Bioavailability studies on mice (IV administration) confirm CNS exposure, and an in silico study predicts a binding mode of LBG30300 which induces a flipping of Tyr144 to allow for a salt bridge interaction of the acetate group with Arg271. The Tyr144 residue now prevents Arg271 from interacting with Asp146, which is a residue of differentiation between mGlu2 and mGlu3 and thus could explain the observed subtype selectivity.


Asunto(s)
Sistema Nervioso Central , Receptores de Glutamato Metabotrópico , Ratones , Animales , Sistema Nervioso Central/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Ciclopropanos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Glutamatos , Ácidos Carboxílicos
4.
Neurosci Lett ; 820: 137595, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38096972

RESUMEN

The current study was designed to examine the role of glutamate NMDA receptors of the mediodorsal thalamus (MD) in scopolamine-induced memory impairment. Adult male rats were bilaterally cannulated into the MD. According to the results, intraperitoneal (i.p.) administration of scopolamine (1.5 mg/kg) immediately after the training phase (post-training) impaired memory consolidation. Bilateral microinjection of the glutamate NMDA receptors agonist, N-Methyl-D-aspartic acid (NMDA; 0.05 µg/rat), into the MD significantly improved scopolamine-induced memory consolidation impairment. Co-administration of D-AP5, a glutamate NMDA receptor antagonist (0.001-0.005 µg/rat, intra-MD) potentiated the response of an ineffective dose of scopolamine (0.5 mg/kg, i.p.) to impair memory consolidation, mimicking the response of a higher dose of scopolamine. Noteworthy, post-training intra-MD microinjections of the same doses of NMDA or D-AP5 alone had no effect on memory consolidation. Moreover, the blockade of the glutamate NMDA receptors by 0.003 ng/rat of D-AP5 prevented the improving effect of NMDA on scopolamine-induced amnesia. Thus, it can be concluded that the MD glutamatergic system may be involved in scopolamine-induced memory impairment via the NMDA receptor signaling pathway.


Asunto(s)
N-Metilaspartato , Escopolamina , Ratas , Masculino , Animales , Escopolamina/farmacología , N-Metilaspartato/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Ácido Glutámico/metabolismo , Ratas Wistar , Amnesia/inducido químicamente , Trastornos de la Memoria/inducido químicamente , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Tálamo/metabolismo , Reacción de Prevención
5.
Biol Psychiatry ; 94(2): 164-173, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-36958998

RESUMEN

BACKGROUND: Patients with schizophrenia show reduced NMDA glutamate receptor-dependent auditory plasticity, which is rate limiting for auditory cognitive remediation (AudRem). We evaluate the utility of behavioral and neurophysiological pharmacodynamic target engagement biomarkers, using a d-serine+AudRem combination. METHODS: Forty-five participants with schizophrenia or schizoaffective disorder were randomized to 3 once-weekly AudRem visits + double-blind d-serine (80, 100, or 120 mg/kg) or placebo in 3 dose cohorts of 12 d-serine and 3 placebo-treated participants each. In AudRem, participants indicated which paired tone was higher in pitch. The primary outcome was plasticity improvement, operationalized as change in pitch threshold between AudRem tones [(test tone Hz - reference tone Hz)/reference tone Hz] between the initial plateau pitch threshold (mean of trials 20-30 of treatment visit 1) to pitch threshold at the end of visit(s). Target engagement was assessed by electroencephalography outcomes, including mismatch negativity (pitch primary). RESULTS: There was a significant overall treatment effect for plasticity improvement (p = .014). Plasticity improvement was largest within the 80 and 100 mg/kg groups (p < .001, d > 0.67), while 120 mg/kg and placebo-treated participants showed nonsignificant within-group changes. Plasticity improvement was seen after a single treatment and was sustained on subsequent treatments. Target engagement was demonstrated by significantly larger mismatch negativity (p = .049, d = 1.0) for the 100 mg/kg dose versus placebo. CONCLUSIONS: Our results demonstrate sufficient proof of principle for continued development of both the d-serine+AudRem combination and our target engagement methodology. The ultimate utility is dependent on the results of an ongoing larger, longer study of the combination for clinically relevant outcomes.


Asunto(s)
Antipsicóticos , Esquizofrenia , Humanos , Esquizofrenia/tratamiento farmacológico , Serina , Receptores de N-Metil-D-Aspartato , N-Metilaspartato/farmacología , N-Metilaspartato/uso terapéutico , Agonistas de Aminoácidos Excitadores/farmacología , Agonistas de Aminoácidos Excitadores/uso terapéutico , Ácido Glutámico/farmacología , Método Doble Ciego , Plasticidad Neuronal , Antipsicóticos/uso terapéutico
6.
Biol Reprod ; 107(4): 916-927, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-35746896

RESUMEN

Free amino acids are present in the natural environment of the preimplantation embryo, and their availability can influence early embryo development. Glutamic acid is one of the amino acids with the highest concentrations in female reproductive fluids, and we investigated whether glutamic acid/glutamate can affect preimplantation embryo development by acting through cell membrane receptors. Using reverse transcription-polymerase chain reaction, we detected 15 ionotropic glutamate receptor transcripts and 8 metabotropic glutamate receptor transcripts in mouse ovulated oocytes and/or in vivo developed blastocysts. Using immunohistochemistry, we detected the expression of two α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunits, three kainate receptor subunits, and member 5 metabotropic glutamate receptor protein in blastocysts. Extracellular concentrations of glutamic acid starting at 5 mM impaired mouse blastocyst development, and this fact may be of great practical importance since glutamic acid and its salts (mainly monosodium glutamate) are widely used as food additives. Experiments with glutamate receptor agonists (in combination with gene expression analysis) revealed that specific AMPA receptors (formed from glutamate receptor, ionotropic, AMPA3 [GRIA3] and/or glutamate receptor, ionotropic, AMPA4 [GRIA4] subunits), kainate receptors (formed from glutamate receptor, ionotropic, kainate 3 [GRIK3] and glutamate receptor, ionotropic, kainate 4 [GRIK4] or glutamate receptor, ionotropic, kainate 5 [GRIK5] subunits), and member 5 metabotropic glutamate receptor (GRM5) were involved in this effect. The glutamic acid-induced effects were prevented or reduced by pretreatment of blastocysts with AMPA, kainate, and GRM5 receptor antagonists, further confirming the involvement of these receptor types. Our results show that glutamic acid can act as a signaling molecule in preimplantation embryos, exerting its effects through the activation of cell membrane receptors.


Asunto(s)
Receptores de Ácido Kaínico , Receptores de Glutamato Metabotrópico , Animales , Blastocisto/metabolismo , Agonistas de Aminoácidos Excitadores/farmacología , Femenino , Aditivos Alimentarios , Glutamatos , Ácido Kaínico/farmacología , Ratones , Receptores AMPA/genética , Receptores AMPA/metabolismo , Receptores de Ácido Kaínico/genética , Receptores de Ácido Kaínico/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Sales (Química)/metabolismo , Glutamato de Sodio , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
7.
J Neurophysiol ; 127(1): 56-85, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34731070

RESUMEN

Thalamic stroke leads to ataxia if the cerebellum-receiving ventrolateral thalamus (VL) is affected. The compensation mechanisms for this deficit are not well understood, particularly the roles that single neurons and specific neuronal subpopulations outside the thalamus play in recovery. The goal of this study was to clarify neuronal mechanisms of the motor cortex involved in mitigation of ataxia during locomotion when part of the VL is inactivated or lesioned. In freely ambulating cats, we recorded the activity of neurons in layer V of the motor cortex as the cats walked on a flat surface and horizontally placed ladder. We first reversibly inactivated ∼10% of the VL unilaterally using glutamatergic transmission antagonist CNQX and analyzed how the activity of motor cortex reorganized to support successful locomotion. We next lesioned 50%-75% of the VL bilaterally using kainic acid and analyzed how the activity of motor cortex reorganized when locomotion recovered. When a small part of the VL was inactivated, the discharge rates of motor cortex neurons decreased, but otherwise the activity was near normal, and the cats walked fairly well. Individual neurons retained their ability to respond to the demand for accuracy during ladder locomotion; however, most changed their response. When the VL was lesioned, the cat walked normally on the flat surface but was ataxic on the ladder for several days after lesion. When ladder locomotion normalized, neuronal discharge rates on the ladder were normal, and the shoulder-related group was preferentially active during the stride's swing phase.NEW & NOTEWORTHY This is the first analysis of reorganization of the activity of single neurons and subpopulations of neurons related to the shoulder, elbow, or wrist, as well as fast- and slow-conducting pyramidal tract neurons in the motor cortex of animals walking before and after inactivation or lesion in the thalamus. The results offer unique insights into the mechanisms of spontaneous recovery after thalamic stroke, potentially providing guidance for new strategies to alleviate locomotor deficits after stroke.


Asunto(s)
Ataxia/fisiopatología , Corteza Motora/fisiopatología , Plasticidad Neuronal/fisiología , Tractos Piramidales/fisiopatología , Recuperación de la Función/fisiología , Núcleos Talámicos Ventrales/fisiopatología , Caminata/fisiología , Animales , Conducta Animal/fisiología , Gatos , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Núcleos Talámicos Ventrales/efectos de los fármacos , Núcleos Talámicos Ventrales/patología
8.
J Med Chem ; 65(1): 734-746, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34918931

RESUMEN

NMDA receptors mediate glutamatergic neurotransmission and are therapeutic targets due to their involvement in a variety of psychiatric and neurological disorders. Here, we describe the design and synthesis of a series of (R)-3-(5-furanyl)carboxamido-2-aminopropanoic acid analogues 8a-s as agonists at the glycine (Gly) binding site in the GluN1 subunit, but not GluN3 subunits, of NMDA receptors. These novel analogues display highly variable potencies and agonist efficacies among the NMDA receptor subtypes (GluN1/2A-D) in a manner dependent on the GluN2 subunit. Notably, compound 8p is identified as a potent partial agonist at GluN1/2C (EC50 = 0.074 µM) with an agonist efficacy of 28% relative to activation by Gly and virtually no agonist activity at GluN1/2A, GluN1/2B, and GluN1/2D. Thus, these novel agonists can modulate the activity of specific NMDA receptor subtypes by replacing the full endogenous agonists Gly or d-serine (d-Ser), thereby providing new opportunities in the development of novel therapeutic agents.


Asunto(s)
Proteínas Portadoras/agonistas , Agonistas de Aminoácidos Excitadores/síntesis química , Agonistas de Aminoácidos Excitadores/farmacología , Glicina/efectos de los fármacos , Proteínas de la Membrana/agonistas , Proteínas del Tejido Nervioso/agonistas , Receptores de N-Metil-D-Aspartato/agonistas , Animales , Humanos , Modelos Moleculares , Relación Estructura-Actividad , Xenopus , Xenopus laevis
9.
Neurochem Int ; 152: 105244, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34826530

RESUMEN

Excitotoxicity refers to the ability of excessive extracellular excitatory amino acids to damage neurons via receptor activation. It is a crucial pathogenetic process in neurodegenerative diseases. TP53 is confirmed to be involved in excitotoxicity. It is demonstrated that TP53 induced glycolysis and apoptotic regulator (TIGAR)-regulated metabolic pathway can protect against neuronal injury. However, the role of TIGAR in excitotoxicity and specific mechanisms is still unknown. In this study, an in vivo excitotoxicity model was constructed via stereotypical kainic acid (KA) injection into the striatum of mice. KA reduced TIGAR expression levels, neuroinflammatory responses and mitochondrial dysfunction. TIGAR overexpression could reverse KA-induced neuronal injury by reducing neuroinflammation and improving mitochondrial function, thereby exerting neuroprotective effects. Therefore, this study could provide a potential therapeutic target for neurodegenerative diseases.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Mitocondrias/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Neuroprotección/efectos de los fármacos , Monoéster Fosfórico Hidrolasas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Kaínico/farmacología , Ratones Transgénicos , Mitocondrias/metabolismo , Enfermedades Neuroinflamatorias/inducido químicamente , Fármacos Neuroprotectores/farmacología
10.
Cell Rep ; 37(9): 110076, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34852231

RESUMEN

A core network of widely expressed proteins within the glutamatergic post-synapse mediates activity-dependent synaptic plasticity throughout the brain, but the specific proteomic composition of synapses differs between brain regions. Here, we address the question, how does proteomic composition affect activity-dependent protein-protein interaction networks (PINs) downstream of synaptic activity? Using quantitative multiplex co-immunoprecipitation, we compare the PIN response of in vivo or ex vivo neurons derived from different brain regions to activation by different agonists or different forms of eyeblink conditioning. We report that PINs discriminate between incoming stimuli using differential kinetics of overlapping and non-overlapping PIN parameters. Further, these "molecular logic rules" differ by brain region. We conclude that although the PIN of the glutamatergic post-synapse is expressed widely throughout the brain, its activity-dependent dynamics show remarkable stimulus-specific and brain-region-specific diversity. This diversity may help explain the challenges in developing molecule-specific drug therapies for neurological disorders.


Asunto(s)
Parpadeo/efectos de los fármacos , Encéfalo/metabolismo , Metoxihidroxifenilglicol/análogos & derivados , N-Metilaspartato/farmacología , Mapas de Interacción de Proteínas , Proteoma/metabolismo , Sinapsis/metabolismo , Animales , Encéfalo/efectos de los fármacos , Condicionamiento Palpebral , Agonistas de Aminoácidos Excitadores/farmacología , Femenino , Masculino , Metoxihidroxifenilglicol/farmacología , Ratones , Plasticidad Neuronal , Proteoma/análisis , Sinapsis/efectos de los fármacos
11.
Cell Rep ; 37(5): 109950, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34731619

RESUMEN

Evidence for prefrontal cortical (PFC) GABAergic dysfunction is one of the most consistent findings in schizophrenia and may contribute to cognitive deficits. Recent studies suggest that the mGlu1 subtype of metabotropic glutamate receptor regulates cortical inhibition; however, understanding the mechanisms through which mGlu1 positive allosteric modulators (PAMs) regulate PFC microcircuit function and cognition is essential for advancing these potential therapeutics toward the clinic. We report a series of electrophysiology, optogenetic, pharmacological magnetic resonance imaging, and animal behavior studies demonstrating that activation of mGlu1 receptors increases inhibitory transmission in the prelimbic PFC by selective excitation of somatostatin-expressing interneurons (SST-INs). An mGlu1 PAM reverses cortical hyperactivity and concomitant cognitive deficits induced by N-methyl-d-aspartate (NMDA) receptor antagonists. Using in vivo optogenetics, we show that prelimbic SST-INs are necessary for mGlu1 PAM efficacy. Collectively, these findings suggest that mGlu1 PAMs could reverse cortical GABAergic deficits and exhibit efficacy in treating cognitive dysfunction in schizophrenia.


Asunto(s)
Antipsicóticos/farmacología , Conducta Animal/efectos de los fármacos , Cognición/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Agonistas de Aminoácidos Excitadores/farmacología , Glicina/análogos & derivados , Interneuronas/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Receptores de Glutamato Metabotrópico/agonistas , Resorcinoles/farmacología , Esquizofrenia/tratamiento farmacológico , Psicología del Esquizofrénico , Somatostatina/metabolismo , Animales , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/psicología , Modelos Animales de Enfermedad , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Glicina/farmacología , Interneuronas/metabolismo , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibición Neural/efectos de los fármacos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Somatostatina/genética
12.
Neurochem Int ; 150: 105191, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34547325

RESUMEN

Spinal cord ischemia-reperfusion injury (SCIRI) can cause dramatic neuron loss and lead to paraplegia in patients. In this research, the role of mGluR5, a member of the metabotropic glutamate receptors (mGluRs) family, was investigated both in vitro and in vivo to explore a possible method to treat this complication. In vitro experiment, after activating mGluR5 via pretreating cells with (RS)-2-Chloro-5-hydroxyphenylglycine (CHPG) and 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl) benzamide (CDPPB), excitotoxicity induced by glutamate (Glu) was attenuated in primary spinal cord neurons, evidenced by higher neuron viability, decreased lactate dehydrogenase (LDH) release and less detected TUNEL-positive cells. According to Western Blot (WB) results, Glu treatment resulted in a high level of large-conductance Ca2+- and voltage-activated K+ (BK) channels, with activation relying on the mGluR5-IP3R (inositol triphosphate) pathway. In vivo part, a rat model of SCIRI was built to further investigate the role of mGluR5. After pretreating them with CHPG and CDPPB, the rats showed markedly lower spinal water content, attenuated motor neuron injury in the spinal cord of L4 segments, and better neurological function. This effect could be partially reversed by paxilline, a blocker of BK channels. In addition, activating BK channels alone using specific openers: NS1619 or NS11021 can protect spinal cord neurons from injury induced by either SCIRI or Glu. In conclusion, in this research, we proved that mGluR5 exerts a protective role in SCIRI, and this effect partially works via IP3R-mediated activation of BK channels.


Asunto(s)
Adenosilhomocisteinasa/biosíntesis , Canales de Potasio de Gran Conductancia Activados por el Calcio/biosíntesis , Neuroprotección/fisiología , Receptor del Glutamato Metabotropico 5/biosíntesis , Daño por Reperfusión/metabolismo , Isquemia de la Médula Espinal/metabolismo , Animales , Benzamidas/farmacología , Células Cultivadas , Agonistas de Aminoácidos Excitadores/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Neuroprotección/efectos de los fármacos , Paxillin/farmacología , Pirazoles/farmacología , Ratas , Receptor del Glutamato Metabotropico 5/agonistas , Daño por Reperfusión/prevención & control , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Isquemia de la Médula Espinal/prevención & control
13.
Cell Rep ; 36(9): 109648, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34469715

RESUMEN

Metabotropic glutamate receptors (mGluRs) are dimeric G-protein-coupled receptors activated by the main excitatory neurotransmitter, L-glutamate. mGluR activation by agonists binding in the venus flytrap domain is regulated by positive (PAM) or negative (NAM) allosteric modulators binding to the 7-transmembrane domain (7TM). We report the cryo-electron microscopy structures of fully inactive and intermediate-active conformations of mGlu5 receptor bound to an antagonist and a NAM or an agonist and a PAM, respectively, as well as the crystal structure of the 7TM bound to a photoswitchable NAM. The agonist induces a large movement between the subunits, bringing the 7TMs together and stabilizing a 7TM conformation structurally similar to the inactive state. Using functional approaches, we demonstrate that the PAM stabilizes a 7TM active conformation independent of the conformational changes induced by agonists, representing an alternative mode of mGlu activation. These findings provide a structural basis for different mGluR activation modes.


Asunto(s)
Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Receptor del Glutamato Metabotropico 5/agonistas , Receptor del Glutamato Metabotropico 5/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Microscopía por Crioelectrón , Cristalografía por Rayos X , Agonistas de Aminoácidos Excitadores/metabolismo , Antagonistas de Aminoácidos Excitadores/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Subunidades de Proteína , Receptor del Glutamato Metabotropico 5/metabolismo , Receptor del Glutamato Metabotropico 5/ultraestructura , Relación Estructura-Actividad
14.
Int J Neuropsychopharmacol ; 24(11): 907-919, 2021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34363482

RESUMEN

BACKGROUND: Enhancement of N-methyl-D-aspartate (NMDA) receptor function using glycine-site agonist D-cycloserine is known to facilitate fear extinction, providing a means to augment cognitive behavioral therapy in anxiety disorders. A novel class of glycine-site agonists has recently been identified, and we have found that the prototype, AICP, is more effective than D-cycloserine in modulating neuronal function. METHODS: Using novel glycine-site agonist AICP, local infusion studies, and genetic models, we elucidated the role of GluN2C-containing receptors in fear extinction. RESULTS: We tested the effect of intracerebroventricular injection of AICP on fear extinction and found a robust facilitation of fear extinction. This effect was dependent on GluN2C subunit, consistent with superagonist action of AICP at GluN2C-containing receptors. Local infusion studies in wild-type and GluN2C knockout mice suggested that AICP produces its effect via GluN2C-containing receptors in the basolateral amygdala (BLA). Furthermore, consistent with astrocytic expression of GluN2C subunit in the amygdala, we found that AICP did not facilitate fear extinction in mice with conditional deletion of obligatory GluN1 subunit from astrocytes. Importantly, chemogenetic activation of astrocytes in the basolateral amygdala facilitated fear extinction. Acutely, AICP was found to facilitate excitatory neurotransmission in the BLA via presynaptic GluN2C-dependent mechanism. Immunohistochemical studies suggest that AICP-mediated facilitation of fear extinction involves synaptic insertion of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor GluA1 subunit. CONCLUSION: These results identify a unique role of astrocytic NMDA receptors composed of GluN2C subunit in extinction of conditioned fear memory and demonstrate that further development of recently identified superagonists of GluN2C-containing receptors may have utility for anxiety disorders.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Astrocitos/metabolismo , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Complejo Nuclear Basolateral/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Cicloserina/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Ratones , Receptores AMPA/metabolismo , Transmisión Sináptica/efectos de los fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo
15.
Cells ; 10(8)2021 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-34440722

RESUMEN

Human stem cell-derived neurons are increasingly considered powerful models in drug discovery and disease modeling, despite limited characterization of their molecular properties. Here, we have conducted a detailed study of the properties of a commercial human induced Pluripotent Stem Cell (iPSC)-derived neuron line, iCell [GABA] neurons, maintained for up to 3 months in vitro. We confirmed that iCell neurons display neurite outgrowth within 24 h of plating and label for the pan-neuronal marker, ßIII tubulin within the first week. Our multi-electrode array (MEA) recordings clearly showed neurons generated spontaneous, spike-like activity within 2 days of plating, which peaked at one week, and rapidly decreased over the second week to remain at low levels up to one month. Extracellularly recorded spikes were reversibly inhibited by tetrodotoxin. Patch-clamp experiments showed that iCell neurons generated spontaneous action potentials and expressed voltage-gated Na and K channels with membrane capacitances, resistances and membrane potentials that are consistent with native neurons. Our single neuron recordings revealed that reduced spiking observed in the MEA after the first week results from development of a dominant inhibitory tone from GABAergic neuron circuit maturation. GABA evoked concentration-dependent currents that were inhibited by the convulsants, bicuculline and picrotoxin, and potentiated by the positive allosteric modulators, diazepam, chlordiazepoxide, phenobarbital, allopregnanolone and mefenamic acid, consistent with native neuronal GABAA receptors. We also show that glycine evoked robust concentration-dependent currents that were inhibited by the neurotoxin, strychnine. Glutamate, AMPA, Kainate and NMDA each evoked concentration-dependent currents in iCell neurons that were blocked by their selective antagonists, consistent with the expression of ionotropic glutamate receptors. The NMDA currents required the presence of the co-agonist glycine and were blocked in a highly voltage-dependent manner by Mg2+ consistent with the properties of native neuronal NMDA receptors. Together, our data suggest that such human iPSC-derived neurons may have significant value in drug discovery and development and may eventually largely replace the need for animal tissues in human biomedical research.


Asunto(s)
Descubrimiento de Drogas , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Neuronas GABAérgicas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Canales Iónicos/efectos de los fármacos , Canales Iónicos/metabolismo , Potenciales de la Membrana , Células-Madre Neurales/metabolismo , Fenotipo
16.
Behav Brain Res ; 415: 113522, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34391797

RESUMEN

BACKGROUND: Motor cortex stimulation (MCS) is proper as a non-pharmacological therapy for patients with chronic and neuropathic pain (NP). AIMS: This work aims to investigate if the MCS in the primary motor cortex (M1) produces analgesia and how the MCS could interfere in the MCS-induced analgesia. Also, to elucidate if the persistent activation of N-methyl-d-aspartic acid receptor (NMDAr) in the periaqueductal grey matter (PAG) can contribute to central sensitisation of the NP. METHODS: Male Wistar rats were submitted to the von Frey test to evaluate the mechanical allodynia after 21 days of chronic constriction injury (CCI) of the sciatic nerve. The MCS was performed with low-frequency (20 µA, 100 Hz) currents during 15 s by a deep brain stimulation (DBS) device. Moreover, the effect of M1-treatment with an NMDAr agonist (at 2, 4, and 8 nmol) was investigated in CCI rats. The PAG dorsomedial column (dmPAG) was pretreated with the NMDAr antagonist LY 235959 (at 8 nmol), followed by MCS. RESULTS: The MCS decreased the mechanical allodynia in rats with chronic NP. The M1-treatment with an NMDA agonist at 2 and 8 nmol reduced the mechanical allodynia in CCI rats. In addition, dmPAG-pretreatment with LY 235959 at 8 nmol attenuated the mechanical allodynia evoked by MCS. CONCLUSION: The M1 cortex glutamatergic system is involved in the modulation of chronic NP. The analgesic effect of MCS may depend on glutamate signaling recruitting NMDAr located on PAG neurons in rodents with chronic NP.


Asunto(s)
Dolor Crónico/terapia , Estimulación Encefálica Profunda , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Corteza Motora/efectos de los fármacos , Neuralgia/terapia , Sustancia Gris Periacueductal/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/fisiología , Analgesia , Animales , Modelos Animales de Enfermedad , Isoquinolinas/farmacología , Masculino , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
17.
J Neurophysiol ; 126(4): 1209-1220, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34406887

RESUMEN

Presympathetic neurons in the paraventricular nucleus of the hypothalamus (PVN) play a key role in cardiovascular regulation. We have previously shown that brain-derived neurotrophic factor (BDNF), acting in the PVN, increases sympathetic activity and blood pressure and serves as a key regulator of stress-induced hypertensive responses. BDNF is known to alter glutamatergic and GABA-ergic signaling broadly in the central nervous system, but whether BDNF has similar actions in the PVN remains to be investigated. Here, we tested the hypothesis that increased BDNF expression in the PVN elevates blood pressure by enhancing N-methyl-d-aspartate (NMDA) receptor (NMDAR)- and inhibiting GABAA receptor (GABAAR)-mediated signaling. Sprague-Dawley rats received bilateral PVN injections of AAV2 viral vectors expressing green fluorescent protein (GFP) or BDNF. Three weeks later, cardiovascular responses to PVN injections of NMDAR and GABAAR agonists and antagonists were recorded under α-chloralose-urethane anesthesia. In addition, expressions of excitatory and inhibitory signaling components in the PVN were assessed using immunofluorescence. Our results showed that NMDAR inhibition led to a greater decrease in blood pressure in the BDNF vs. GFP group, while GABAAR inhibition led to greater increases in blood pressure in the GFP group compared to BDNF. Conversely, GABAAR activation decreased blood pressure significantly more in GFP vs. BDNF rats. In addition, immunoreactivity of NMDAR1 was upregulated, while GABAAR-α1 and K+/Cl- cotransporter 2 were downregulated by BDNF overexpression in the PVN. In summary, our findings indicate that hypertensive actions of BDNF within the PVN are mediated, at least in part, by augmented NMDAR and reduced GABAAR signaling.NEW & NOTEWORTHY We have shown that BDNF, acting in the PVN, elevates blood pressure in part by augmenting NMDA receptor-mediated excitatory input and by diminishing GABAA receptor-mediated inhibitory input to PVN neurons. In addition, we demonstrate that elevated BDNF expression in the PVN upregulates NMDA receptor immunoreactivity and downregulates GABAA receptor as well as KCC2 transporter immunoreactivity.


Asunto(s)
Presión Sanguínea/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Fenómenos Electrofisiológicos/fisiología , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sistema Nervioso Simpático/fisiología , Animales , Presión Sanguínea/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/farmacología , Fenómenos Electrofisiológicos/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Agonistas de Receptores de GABA-A/farmacología , Antagonistas de Receptores de GABA-A/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sistema Nervioso Simpático/efectos de los fármacos
18.
Behav Brain Res ; 413: 113443, 2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34216648

RESUMEN

The present study investigated the pharmacological mechanisms of the antidepressant-like effects of amantadine in mice and their influence on hippocampal neurogenesis. To improve the translational validity of preclinical results, reproducibility across laboratories and replication in other animal models and species are crucial. Single amantadine administration at doses of 50 and 75 mg/kg resulted in antidepressant-like effects in mice in the tail suspension test (TST), reflected by an increase in immobility time. The effects of amantadine were seen at doses that did not alter locomotor activity. The tyrosine hydroxylase inhibitor α-methyl-ρ-tyrosine did not influence the anti-immobility effect of amantadine in the TST. Pretreatment with the α1 adrenergic receptor antagonist prazosin, ß adrenergic receptor antagonist propranolol, α2 adrenergic receptor antagonist yohimbine, and α2 adrenergic receptor agonist clonidine did not alter the antidepressant-like effect of amantadine. However, amantadine's effect was blocked by the dopamine D2 receptor antagonist haloperidol and glutamate receptor agonist N-methyl-D-aspartate (NMDA). Repeated amantadine administration (50 mg/kg) also exerted an antidepressant-like effect, paralleled by an increase in hippocampal neurogenesis. The present results demonstrate that the antidepressant-like effects of amantadine may be mediated by its actions on D2 and NMDA receptors and likely involve hippocampal neurogenesis.


Asunto(s)
Agonistas Adrenérgicos/farmacología , Antagonistas Adrenérgicos/farmacología , Amantadina/farmacología , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Receptores de Dopamina D2/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Amantadina/administración & dosificación , Animales , Antidepresivos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Hipocampo/efectos de los fármacos , Masculino , Ratones , Neurogénesis/efectos de los fármacos , alfa-Metiltirosina/farmacología
19.
Bull Exp Biol Med ; 171(3): 327-332, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34297297

RESUMEN

We studied the prolonged action of kainic acid on glutamatergic neurons in the dorsal hippocampus and the endocannabinoid-dependent protection against neurodegeneration. The pyramidal neurons of the CA3 field of the hippocampus, as well as granular and mossy cells of the dentate gyrus were examined. Light and electron microscopy revealed substantial damage to the components of the protein-synthesizing (rough endoplasmic reticulum, Golgi apparatus, and polyribosomes) and catabolic (lysosomes, autophagosomes, multivesicular structures, and lipofuscin formations) systems in all cells. Pyramidal and mossy neurons die mainly by the necrotic pathway. The death of granular cells occurred through both apoptosis and necrosis. The most vulnerable cells are mossy neurons located in the hilus. Activation of the endocannabinoid system induced by intracerebral injection of URB597, an inhibitor of degradation of endocannabinoid anandamide, protected the normal structure of the hippocampus and prevented neuronal damage and death induced by KA.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Endocannabinoides/metabolismo , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Kaínico/farmacología , Degeneración Nerviosa/patología , Alcamidas Poliinsaturadas/metabolismo , Células Piramidales/efectos de los fármacos , Estado Epiléptico/patología , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Benzamidas/farmacología , Región CA3 Hipocampal/efectos de los fármacos , Región CA3 Hipocampal/metabolismo , Región CA3 Hipocampal/patología , Carbamatos/farmacología , Giro Dentado/efectos de los fármacos , Giro Dentado/metabolismo , Giro Dentado/patología , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/ultraestructura , Masculino , Microscopía Electrónica , Necrosis/metabolismo , Necrosis/patología , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/metabolismo , Células Piramidales/metabolismo , Células Piramidales/patología , Ratas , Ratas Wistar , Estado Epiléptico/inducido químicamente , Estado Epiléptico/metabolismo
20.
J Neurochem ; 158(5): 1083-1093, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34293825

RESUMEN

Kainate (KA) receptors (KARs) are important modulators of synaptic transmission. We studied here the role of KARs on glutamatergic synaptic transmission in the CA2 region of the hippocampus where the actions of these receptors are unknown. We observed that KA depresses glutamatergic synaptic transmission at Schaffer collateral-CA2 synapses; an effect that was antagonized by NBQX (a KA/AMPA receptors antagonist) under condition where AMPA receptors were previously blocked. The study of paired-pulse facilitation ratio, miniature responses, and fluctuation analysis indicated a presynaptic locus of action for KAR. Additionally, we determined the action mechanism for this depression of glutamate release mediated by the activation of KARs. We found that inhibition of protein kinase A suppressed the effect of KAR activation on evoked excitatory post-synaptic current, an effect that was not suppressed by protein kinase C inhibitors. Furthermore, in the presence of Pertussis toxin, the depression of glutamate release mediated by KAR activation was not present, invoking the participation of a Gi/o protein in this modulation. Finally, the KAR-mediated depression of glutamate release was not suppressed by treatments that affect calcium entry trough voltage-dependent calcium channels or calcium release from intracellular stores. We conclude that KARs present at these synapses mediate a depression of glutamate release through a mechanism that involves the activation of G protein and protein kinase A.


Asunto(s)
Región CA2 Hipocampal/metabolismo , Ácido Glutámico/metabolismo , Receptores de Ácido Kaínico/agonistas , Receptores de Ácido Kaínico/metabolismo , Transmisión Sináptica/fisiología , Animales , Región CA2 Hipocampal/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Femenino , Ácido Kaínico/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Transmisión Sináptica/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA