Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 13: 999685, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36438799

RESUMEN

Background: Phosphodiesterase 10A (PDE10A) is expressed almost exclusively in the striatum and its inhibition is suggested to offer potential treatment in disorders associated with basal ganglia. We evaluated the selectivity, cytotoxicity, genotoxicity, pharmacokinetics and potential adverse effects of a novel PDE10A inhibitor, CPL500036, in vivo. Methods: The potency of CPL500036 was demonstrated by microfluidic technology, and selectivity was investigated in a radioligand binding assay against 44 targets. Cardiotoxicity in vitro was evaluated in human ether-a-go-go related gene (hERG)-potassium channel-overexpressing cells by the patch-clamp method and by assessing key parameters in 3D cardiac spheroids. Cytotoxicity was determined in H1299, HepG2 and SH-SY5Y cell lines. The Ames test was used for genotoxicity analyses. During in vivo studies, CPL500036 was administered by oral gavage. CPL500036 exposure were determined by liquid chromatography-tandem mass spectrometry and plasma protein binding was assessed. The bar test was employed to assess catalepsy. Prolactin and glucose levels in rat blood were measured by ELISAs and glucometers, respectively. Cardiovascular safety in vivo was investigated in dogs using a telemetry method. Results: CPL500036 inhibited PDE10A at an IC50 of 1 nM, and interacted only with the muscarinic M2 receptor as a negative allosteric modulator with an IC50 of 9.2 µM. Despite inhibiting hERG tail current at an IC25 of 3.2 µM, cardiovascular adverse effects were not observed in human cardiac 3D spheroids or in vivo. Cytotoxicity in vitro was observed only at > 60 µM and genotoxicity was not recorded during the Ames test. CPL500036 presented good bioavailability and penetration into the brain. CPL500036 elicited catalepsy at 0.6 mg/kg, but hyperprolactinemia or hyperglycemic effects were not observed in doses up to 3 mg/kg. Conclusion: CPL500036 is a potent, selective and orally bioavailable PDE10A inhibitor with a good safety profile distinct from marketed antipsychotics. CPL500036 may be a compelling drug candidate.

2.
Pharmaceuticals (Basel) ; 14(8)2021 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-34451801

RESUMEN

TrkB is a tyrosine kinase receptor that is activated upon binding to brain-derived neurotrophic factor (BDNF). To date, the search for low-molecular-weight molecules mimicking BDNF's action has been unsuccessful. Several molecules exerting antidepressive effects in vivo, such as 7,8-DHF, have been suggested to be TrkB agonists. However, more recent publications question this hypothesis. In this study, we developed a set of experimental procedures including the evaluation of direct interactions, dimerization, downstream signaling, and cytoprotection in parallel with physicochemical and ADME methods to verify the pharmacology of 7,8-DHF and other potential reference compounds, and perform screening for novel TrkB agonists. 7,8 DHF bound to TrkB with Kd = 1.3 µM; however, we were not able to observe any other activity against the TrkB receptor in SN56 T48 and differentiated SH-SY5Y cell lines. Moreover, the pharmacokinetic and pharmacodynamic effects of 7,8-DHF at doses of 1 and 50 mg/kg were examined in mice after i.v and oral administration, respectively. The poor pharmacokinetic properties and lack of observed activation of TrkB-dependent signaling in the brain confirmed that 7,8-DHF is not a relevant tool for studying TrkB activation in vivo. The binding profile for 133 molecular targets revealed a significant lack of selectivity of 7,8-DHF, suggesting a distinct functional profile independent of interaction with TrkB. Additionally, a compound library was screened in search of novel low-molecular-weight orthosteric TrkB agonists; however, we were not able to identify reliable drug candidates. Our results suggest that published reference compounds including 7,8-DHF do not activate TrkB, consistent with canonical dogma, which indicates that the reported pharmacological activity of these compounds should be interpreted carefully in a broad functional context.

3.
Sci Rep ; 7: 43135, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28230157

RESUMEN

Internally quenched fluorescent (IQF) peptide substrates originating from FRET (Förster Resonance Energy Transfer) are powerful tool for examining the activity and specificity of proteases, and a variety of donor/acceptor pairs are extensively used to design individual substrates and combinatorial libraries. We developed a highly sensitive and adaptable donor/acceptor pair that can be used to investigate the substrate specificity of cysteine proteases, serine proteases and metalloproteinases. This novel pair comprises 7-amino-4-carbamoylmethylcoumarin (ACC) as the fluorophore and 2,4-dinitrophenyl-lysine (Lys(DNP)) as the quencher. Using caspase-3, caspase-7, caspase-8, neutrophil elastase, legumain, and two matrix metalloproteinases (MMP2 and MMP9), we demonstrated that substrates containing ACC/Lys(DNP) exhibit 7 to 10 times higher sensitivity than conventional 7-methoxy-coumarin-4-yl acetic acid (MCA)/Lys(DNP) substrates; thus, substantially lower amounts of substrate and enzyme can be used for each assay. We therefore propose that the ACC/Lys(DNP) pair can be considered a novel and sensitive scaffold for designing substrates for any group of endopeptidases. We further demonstrate that IQF substrates containing unnatural amino acids can be used to investigate protease activities/specificities for peptides containing post-translationally modified amino acids. Finally, we used IQF substrates to re-investigate the P1-Asp characteristic of caspases, thus demonstrating that some human caspases can also hydrolyze substrates after glutamic acid.


Asunto(s)
Proteasas de Cisteína/metabolismo , Fluorometría/métodos , Metaloproteasas/metabolismo , Serina Proteasas/metabolismo , Especificidad por Sustrato , Cumarinas/metabolismo , Humanos , Lisina/análogos & derivados , Lisina/metabolismo , Sensibilidad y Especificidad
4.
Sci Rep ; 6: 27757, 2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-27282248

RESUMEN

Activity-dependent proteolysis at a synapse has been recognized as a pivotal factor in controlling dynamic changes in dendritic spine shape and function; however, excessive proteolytic activity is detrimental to the cells. The exact mechanism of control of these seemingly contradictory outcomes of protease activity remains unknown. Here, we reveal that dendritic spine maturation is strictly controlled by the proteolytic activity, and its inhibition by the endogenous inhibitor (Tissue inhibitor of matrix metalloproteinases-1 - TIMP-1). Excessive proteolytic activity impairs long-term potentiation of the synaptic efficacy (LTP), and this impairment could be rescued by inhibition of protease activity. Moreover LTP is altered persistently when the ability of TIMP-1 to inhibit protease activity is abrogated, further demonstrating the role of such inhibition in the promotion of synaptic plasticity under well-defined conditions. We also show that dendritic spine maturation involves an intermediate formation of elongated spines, followed by their conversion into mushroom shape. The formation of mushroom-shaped spines is accompanied by increase in AMPA/NMDA ratio of glutamate receptors. Altogether, our results identify inhibition of protease activity as a critical regulatory mechanism for dendritic spines maturation.


Asunto(s)
Matriz Extracelular/metabolismo , Plasticidad Neuronal , Péptido Hidrolasas/metabolismo , Animales , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Humanos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Modelos Biológicos , Plasticidad Neuronal/efectos de los fármacos , Proteolisis/efectos de los fármacos , Ratas Transgénicas , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Recombinantes/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
5.
J Neurosci ; 35(13): 5171-9, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25834043

RESUMEN

Kainate receptors (KARs) are found ubiquitously in the CNS and are present presynaptically and postsynaptically regulating synaptic transmission and excitability. Functional studies have proven that KARs act as ion channels as well as potentially activating G-proteins, thus indicating the existance of a dual signaling system for KARs. Nevertheless, it is not clear how these ion channels activate G-proteins and which of the KAR subunits is involved. Here we performed a proteomic analysis to define proteins that interact with the C-terminal domain of GluK1 and we identified a variety of proteins with many different functions, including a Go α subunit. These interactions were verified through distinct in vitro and in vivo assays, and the activation of the Go protein by GluK1 was validated in bioluminescence resonance energy transfer experiments, while the specificity of this association was confirmed in GluK1-deficient mice. These data reveal components of the KAR interactome, and they show that GluK1 and Go proteins are natural partners, accounting for the metabotropic effects of KARs.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteómica , Receptores de Ácido Kaínico/química , Receptores de Ácido Kaínico/metabolismo , Animales , Encéfalo/metabolismo , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Células HEK293 , Humanos , Ácido Kaínico/farmacología , Masculino , Ratones , Ratones Noqueados , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Subunidades de Proteína , Receptores de Ácido Kaínico/genética
6.
PLoS One ; 9(6): e99789, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24918931

RESUMEN

DP-b99 is a membrane-activated chelator of zinc and calcium ions, recently proposed as a therapeutic agent. Matrix metalloproteinases (MMPs) are zinc-dependent extracellularly operating proteases that might contribute to synaptic plasticity, learning and memory under physiological conditions. In excessive amounts these enzymes contribute to a number of neuronal pathologies ranging from the stroke to neurodegeneration and epileptogenesis. In the present study, we report that DP-b99 delays onset and severity of PTZ-induced seizures in mice, as well as displays neuroprotective effect on kainate excitotoxicity in hippocampal organotypic slices and furthermore blocks morphological reorganization of the dendritic spines evoked by a major neuronal MMP, MMP-9. Taken together, our findings suggest that DP-b99 may inhibit neuronal plasticity driven by MMPs, in particular MMP-9, and thus may be considered as a therapeutic agent under conditions of aberrant plasticity, such as those subserving epileptogenesis.


Asunto(s)
Ácido Egtácico/análogos & derivados , Metaloproteinasa 9 de la Matriz/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Animales , Quelantes/farmacología , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/metabolismo , Ácido Egtácico/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ácido Kaínico , Masculino , Ratones , Ratones Endogámicos C57BL , Convulsiones/dietoterapia , Convulsiones/metabolismo
7.
Biomaterials ; 35(5): 1402-10, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24290700

RESUMEN

A genetically encoded Förster Resonance Energy Transfer (FRET)-based biosensor that continuously monitors matrix metalloproteinase 9 (MMP-9) activity was developed. MMP-9 is an extracellularly acting endopeptidase with a prominent role in development, learning and memory, cancer metastasis, and stroke. To assess the biological function of the protease, determining the precise kinetics and localization of MMP-9 activity is required. The nontoxic, genetically encoded FRET biosensor presented herein is anchored in the cellular membrane and thus provides an important advantage over currently employed probes. The biosensor allows the study of the proteolytic activity of MMP-9 with high temporal and subcellular resolution at the precise region of MMP-9 action on the cell. The applicability of the biosensor both in vitro and in living cells was demonstrated by ratiometrically analyzing the cleavage of the biosensor by a purified auto-activating mutant of MMP-9 and endogenously secreted protease in cultured tumor and neuronal cells. The precise kinetics of endogenous MMP-9 activity was measured, which demonstrates in a straight-forward manner the applicability of the biosensor concept.


Asunto(s)
Técnicas Biosensibles , Transferencia Resonante de Energía de Fluorescencia , Metaloproteinasa 9 de la Matriz/metabolismo , Secuencia de Bases , Membrana Celular/metabolismo , Cartilla de ADN , Células HEK293 , Humanos , Cinética , Metaloproteinasa 9 de la Matriz/genética , Proteolisis
8.
Biochim Biophys Acta ; 1804(4): 891-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20056171

RESUMEN

Mass spectrometry is used to probe the kinetics of hydrogen-deuterium exchange in lysozyme in pH 5, 6 and 7.4. An analysis based on a Verhulst growth model is proposed and effectively applied to the kinetics of the hydrogen exchange. The data are described by a power-like function which is based on a time-dependence of the exchange rate. Experimental data ranging over many time scales is considered and accurate fits of a power-like function are obtained. Results of fittings show correlation between faster hydrogen-deuterium exchange and increase of pH. Furthermore a model is presented that discriminates between easily exchangeable hydrogens (located in close proximity to the protein surface) and those protected from the exchange (located in the protein interior). A possible interpretation of the model and its biological significance are discussed.


Asunto(s)
Muramidasa/química , Animales , Pollos , Medición de Intercambio de Deuterio , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Cinética , Espectrometría de Masas , Modelos Biológicos , Modelos Químicos , Simulación de Dinámica Molecular , Muramidasa/metabolismo , Protones
9.
Biochemistry ; 47(9): 2710-20, 2008 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-18220364

RESUMEN

Recognition of the 5' cap by the eukaryotic initiation factor 4E (eIF4E) is the rate-limiting step in the ribosome recruitment to mRNAs. The regular cap consists of 7-monomethylguanosine (MMG) linked by a 5'-5' triphosphate bridge to the first transcribed nucleoside, while some primitive eukaryotes possess a N (2), N (2),7-trimethylguanosine (TMG) cap structure as a result of trans splicing. Mammalian eIF4E is highly specific to the MMG form of the cap in terms of association constants and thermodynamic driving force. We have investigated conformational changes of eIF4E induced by interaction with two cap analogues, 7-methyl-GTP and N (2), N (2),7-trimethyl-GTP. Hydrogen-deuterium exchange and electrospray mass spectrometry were applied to probe local dynamics of murine eIF4E in the apo and cap-bound forms. The data show that the cap binding induces long-range conformational changes in the protein, not only in the cap-binding pocket but also in a distant region of the 4E-BP/eIF4G binding site. Formation of the complex with 7-methyl-GTP makes the eIF4E structure more compact, while binding of N (2), N (2),7-trimethyl-GTP leads to higher solvent accessibility of the protein backbone in comparison with the apo form. The results suggest that the additional double methylation at the N (2)-amino group of the cap causes sterical effects upon binding to mammalian eIF4E which influence the overall solution dynamics of the protein, thus precluding formation of a tight complex.


Asunto(s)
Factor 4E Eucariótico de Iniciación/química , Guanosina/análogos & derivados , Análogos de Caperuza de ARN/química , ARN Mensajero/química , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Factor 4E Eucariótico de Iniciación/metabolismo , Guanosina/química , Guanosina/metabolismo , Datos de Secuencia Molecular , Unión Proteica , Análogos de Caperuza de ARN/metabolismo , Proteínas de Unión a Caperuzas de ARN/química , Proteínas de Unión a Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...