Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cell Reports ; 13(2): 380-393, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31378672

RESUMEN

Here, we have used patient-derived induced pluripotent stem cell (iPSC) and gene-editing technology to study the cardiac-related molecular and functional consequences of mutations in GLA causing the lysosomal storage disorder Fabry disease (FD), for which heart dysfunction is a major cause of mortality. Our in vitro model recapitulated clinical data with FD cardiomyocytes accumulating GL-3 and displaying an increased excitability, with altered electrophysiology and calcium handling. Quantitative proteomics enabled the identification of >5,500 proteins in the cardiomyocyte proteome and secretome, and revealed accumulation of the lysosomal protein LIMP-2 and secretion of cathepsin F and HSPA2/HSP70-2 in FD. Genetic correction reversed these changes. Overexpression of LIMP-2 directly induced the secretion of cathepsin F and HSPA2/HSP70-2, implying causative relationship, and led to massive vacuole accumulation. In summary, our study has revealed potential new cardiac biomarkers for FD, and provides valuable mechanistic insight into the earliest pathological events in FD cardiomyocytes.


Asunto(s)
Enfermedad de Fabry/patología , Proteínas de Membrana de los Lisosomas/metabolismo , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Receptores Depuradores/metabolismo , Potenciales de Acción , Biomarcadores/metabolismo , Catepsina F/metabolismo , Edición Génica , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/fisiología , Mutación Puntual , Mapas de Interacción de Proteínas , Proteómica , Vacuolas/metabolismo , alfa-Galactosidasa/genética
2.
Allergy ; 74(3): 549-559, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-29987849

RESUMEN

BACKGROUND: Chronic rhinosinusitis (CRS) with nasal polyps (CRSwNP) is a chronic inflammatory disease often accompanied by impairment of sense of smell. This symptom has been somewhat overlooked, and its relationship to inflammatory cytokines, tissue compression, neuronal loss, and neurogenesis is still unclear. METHODS: In order to elucidate potential mechanisms leading to CRS in humans, we have established a type 2/T helper type 2 cell (Th2)-mediated allergic CRS mouse model, based on house dust mite (HDM) and Staphylococcus aureus enterotoxin B (SEB) sensitization. The inflammatory status of the olfactory epithelium (OE) was assessed using histology, biochemistry, and transcriptomics. The sense of smell was evaluated by studying olfactory behavior and recording electro-olfactograms (EOGs). RESULTS: After 22 weeks, a typical type 2/Th2-mediated inflammatory profile was obtained, as demonstrated by increased interleukin (IL)-4, IL-5, and IL-13 in the OE. The number of mast cells and eosinophils was increased, and infiltration of these cells into the olfactory mucosa was also observed. In parallel, transcriptomic and histology analyses indicated a decreased number of immature olfactory neurons, possibly due to decreased renewal. However, the number of mature sensory neurons was not affected and neither the EOG nor olfactory behavior was impaired. CONCLUSION: Our mouse model of CRS displayed an allergic response to HDM + SEB administration, including the type 2/Th2 inflammatory profile characteristic of human eosinophilic CRSwNP. Although the sense of smell did not appear to be altered in these conditions, the data reveal the influence of chronic inflammation on olfactory neurogenesis, suggesting that factors unique to humans may be involved in CRSwNP-associated anosmia.


Asunto(s)
Neurogénesis , Mucosa Olfatoria/metabolismo , Rinitis/etiología , Rinitis/metabolismo , Sinusitis/etiología , Sinusitis/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Animales , Biomarcadores , Enfermedad Crónica , Modelos Animales de Enfermedad , Ratones , Neurogénesis/genética , Neurogénesis/inmunología , Mucosa Olfatoria/fisiopatología , Neuronas Receptoras Olfatorias/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Rinitis/fisiopatología , Sinusitis/fisiopatología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
3.
Curr Protoc Pharmacol ; 75: 9.21.1-9.21.35, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27960031

RESUMEN

Over the last six decades, voltage-gated sodium (Nav ) channels have attracted a great deal of scientific and pharmaceutical interest, driving fundamental advances in both biology and technology. The structure and physiological function of these channels have been extensively studied; clinical and genetic data have uncovered their implication in diseases such as epilepsy, arrhythmias, and pain, bringing them into focus as current and future drug targets. While different techniques have been established to record the activity of Nav channels, proper determination of their properties still presents serious challenges, depending upon the experimental conditions and the desired subtype of channel to be characterized. The aim of this unit is to review the characteristics of Nav channels, their properties, the cells in which they can be studied, and the currently available techniques. Topics covered include the determination of Nav -channel biophysical properties as well as the use of toxins to discriminate between subtypes using electrophysiological or optical methods. Perspectives on the development of high-throughput screening assays with their advantages and limitations are also discussed to allow a better understanding of the challenges encountered in voltage-gated sodium channel preclinical drug discovery. © 2016 by John Wiley & Sons, Inc.


Asunto(s)
Descubrimiento de Drogas/métodos , Canales de Sodio/metabolismo , Animales , Línea Celular , Fenómenos Electrofisiológicos , Ensayos Analíticos de Alto Rendimiento , Humanos , Potenciales de la Membrana , Ratones , Ratas , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/genética
4.
Pharmacol Biochem Behav ; 91(1): 47-58, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18621075

RESUMEN

On native human, rat and mouse glycine transporter-1(GlyT1), SSR130800 behaves as a selective inhibitor with IC50 values of 1.9, 5.3 and 6.8 nM, respectively. It reversibly blocked glycine uptake in mouse brain cortical homogenates, increased extracellular levels of glycine in the rat prefrontal cortex, and potentiated NMDA-mediated excitatory postsynaptic currents in rat hippocampal slices. SSR103800 (30 mg/kg, p.o.) decreased MK-801- and PCP-induced locomotor hyperactivity in rodents. SSR103800 (1 and 10 mg/kg, p.o.) attenuated social recognition deficit in adult rats induced by neonatal injections of PCP (10 mg/kg, s.c., on post-natal day 7, 9 and 11). SSR103800 (3 mg/kg, p.o.) counteracted the deficit in short-term visual episodic-like memory induced by a low challenge dose of PCP (1 mg/kg, i.p.), in PCP-sensitized rats (10 mg/kg, i.p.). SSR103800 (30 mg/kg, i.p.) increased the prepulse inhibition of the startle reflex in DBA/1J mice. SSR103800 decreased defensive- and despair-related behaviors in the tonic immobility test in gerbils (10 and 30 mg/kg, p.o.) and in the forced-swimming procedure in rats (1 and 3 mg/kg, p.o.), respectively. These findings suggest that SSR103800 may have a therapeutic potential in the management of the core symptoms of schizophrenia and comorbid depression states.


Asunto(s)
Antipsicóticos/farmacología , Proteínas de Transporte de Glicina en la Membrana Plasmática/antagonistas & inhibidores , Esquizofrenia/tratamiento farmacológico , Animales , Antidepresivos/farmacología , Discriminación en Psicología/efectos de los fármacos , Maleato de Dizocilpina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Gerbillinae , Glicina/metabolismo , Masculino , Ratones , Actividad Motora/efectos de los fármacos , N-Metilaspartato/fisiología , Fenciclidina/farmacología , Reconocimiento en Psicología/efectos de los fármacos , Reflejo de Sobresalto/efectos de los fármacos , Estereoisomerismo , Natación/psicología
5.
Neuropsychopharmacology ; 32(1): 1-16, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17019409

RESUMEN

In this paper, we report on the pharmacological and functional profile of SSR180711 (1,4-Diazabicyclo[3.2.2]nonane-4-carboxylic acid, 4-bromophenyl ester), a new selective alpha7 acetylcholine nicotinic receptor (n-AChRs) partial agonist. SSR180711 displays high affinity for rat and human alpha7 n-AChRs (K(i) of 22+/-4 and 14+/-1 nM, respectively). Ex vivo (3)[H]alpha-bungarotoxin binding experiments demonstrate that SSR180711 rapidly penetrates into the brain (ID(50)=8 mg/kg p.o.). In functional studies performed with human alpha7 n-AChRs expressed in Xenopus oocytes or GH4C1 cells, the compound shows partial agonist effects (intrinsic activity=51 and 36%, EC(50)=4.4 and 0.9 microM, respectively). In rat cultured hippocampal neurons, SSR180711 induced large GABA-mediated inhibitory postsynaptic currents and small alpha-bungarotoxin sensitive currents through the activation of presynaptic and somato-dendritic alpha7 n-AChRs, respectively. In mouse hippocampal slices, the compound increased the amplitude of both glutamatergic (EPSCs) and GABAergic (IPSCs) postsynaptic currents evoked in CA1 pyramidal cells. In rat and mouse hippocampal slices, a concentration of 0.3 muM of SSR180711 increased long-term potentiation (LTP) in the CA1 field. Null mutation of the alpha7 n-AChR gene totally abolished SSR180711-induced modulation of EPSCs, IPSCs and LTP in mice. Intravenous administration of SSR180711 strongly increased the firing rate of single ventral pallidum neurons, extracellularly recorded in anesthetized rats. In microdialysis experiments, administration of the compound (3-10 mg/kg i.p.) dose-dependently increased extracellular acetylcholine (ACh) levels in the hippocampus and prefrontal cortex of freely moving rats. Together, these results demonstrate that SSR180711 is a selective and partial agonist at human, rat and mouse alpha7 n-AChRs, increasing glutamatergic neurotransmission, ACh release and LTP in the hippocampus.


Asunto(s)
Agonistas Nicotínicos/farmacología , Agonistas Nicotínicos/farmacocinética , Receptores Nicotínicos/fisiología , Animales , Animales Recién Nacidos , Sitios de Unión/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Hipocampo/citología , Humanos , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/fisiología , Agonistas Nicotínicos/química , Antagonistas Nicotínicos/farmacología , Oocitos/fisiología , Técnicas de Placa-Clamp/métodos , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/fisiología , Ratas , Ratas Sprague-Dawley , Receptores Nicotínicos/deficiencia , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Receptor Nicotínico de Acetilcolina alfa 7 , Ácido gamma-Aminobutírico/farmacología
6.
Neuropsychopharmacology ; 30(11): 1963-85, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15956994

RESUMEN

Noncompetitive N-methyl-D-aspartate (NMDA) blockers induce schizophrenic-like symptoms in humans, presumably by impairing glutamatergic transmission. Therefore, a compound potentiating this neurotransmission, by increasing extracellular levels of glycine (a requisite co-agonist of glutamate), could possess antipsychotic activity. Blocking the glycine transporter-1 (GlyT1) should, by increasing extracellular glycine levels, potentiate glutamatergic neurotransmission. SSR504734, a selective and reversible inhibitor of human, rat, and mouse GlyT1 (IC50=18, 15, and 38 nM, respectively), blocked reversibly the ex vivo uptake of glycine (mouse cortical homogenates: ID50: 5 mg/kg i.p.), rapidly and for a long duration. In vivo, it increased (minimal efficacious dose (MED): 3 mg/kg i.p.) extracellular levels of glycine in the rat prefrontal cortex (PFC). This resulted in an enhanced glutamatergic neurotransmission, as SSR504734 potentiated NMDA-mediated excitatory postsynaptic currents (EPSCs) in rat hippocampal slices (minimal efficacious concentration (MEC): 0.5 microM) and intrastriatal glycine-induced rotations in mice (MED: 1 mg/kg i.p.). It normalized activity in rat models of hippocampal and PFC hypofunctioning (through activation of presynaptic CB1 receptors): it reversed the decrease in electrically evoked [3H]acetylcholine release in hippocampal slices (MEC: 10 nM) and the reduction of PFC neurons firing (MED: 0.3 mg/kg i.v.). SSR504734 prevented ketamine-induced metabolic activation in mice limbic areas and reversed MK-801-induced hyperactivity and increase in EEG spectral energy in mice and rats, respectively (MED: 10-30 mg/kg i.p.). In schizophrenia models, it normalized a spontaneous prepulse inhibition deficit in DBA/2 mice (MED: 15 mg/kg i.p.), and reversed hypersensitivity to locomotor effects of d-amphetamine and selective attention deficits (MED: 1-3 mg/kg i.p.) in adult rats treated neonatally with phencyclidine. Finally, it increased extracellular dopamine in rat PFC (MED: 10 mg/kg i.p.). The compound showed additional activity in depression/anxiety models, such as the chronic mild stress in mice (10 mg/kg i.p.), ultrasonic distress calls in rat pups separated from their mother (MED: 1 mg/kg s.c.), and the increased latency of paradoxical sleep in rats (MED: 30 mg/kg i.p.). In conclusion, SSR504734 is a potent and selective GlyT1 inhibitor, exhibiting activity in schizophrenia, anxiety and depression models. By targeting one of the primary causes of schizophrenia (hypoglutamatergy), it is expected to be efficacious not only against positive but also negative symptoms, cognitive deficits, and comorbid depression/anxiety states.


Asunto(s)
Benzamidas/farmacología , Química Encefálica/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas de Transporte de Glicina en la Membrana Plasmática/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Piperidinas/farmacología , Acetilcolina/metabolismo , Potenciales de Acción/efectos de los fármacos , Anfetamina/farmacología , Análisis de Varianza , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Isótopos de Carbono/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Ritmo Circadiano/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/química , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Glicina/metabolismo , Hipocampo/citología , Humanos , Técnicas In Vitro , Concentración 50 Inhibidora , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp/métodos , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...