Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Pharmaceutics ; 14(11)2022 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-36365220

RESUMEN

Dual functionalized liposomes were developed to cross the blood−brain barrier (BBB) and to release their cargo in a pathological matrix metalloproteinase (MMP)-rich microenvironment. Liposomes were surface-functionalized with a modified peptide deriving from the receptor-binding domain of apolipoprotein E (mApoE), known to promote cargo delivery to the brain across the BBB in vitro and in vivo; and with an MMP-sensitive moiety for an MMP-triggered drug release. Different MMP-sensitive peptides were functionalized at both ends with hydrophobic stearate tails to yield MMP-sensitive lipopeptides (MSLPs), which were assembled into mApoE liposomes. The resulting bi-functional liposomes (i) displayed a < 180 nm diameter with a negative ζ-potential; (ii) were able to cross an in vitro BBB model with an endothelial permeability of 3 ± 1 × 10−5 cm/min; (iii) when exposed to functional MMP2 or 9, efficiently released an encapsulated fluorescein dye; (iv) showed high biocompatibility when tested in neuronal cultures; and (v) when loaded with glibenclamide, a drug candidate with poor aqueous solubility, reduced the release of proinflammatory cytokines from activated microglial cells.

2.
J Med Chem ; 64(12): 8333-8353, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34097384

RESUMEN

Acid-sensitive ion channels (ASICs) are sodium channels partially permeable to Ca2+ ions, listed among putative targets in central nervous system (CNS) diseases in which a pH modification occurs. We targeted novel compounds able to modulate ASIC1 and to reduce the progression of ischemic brain injury. We rationally designed and synthesized several diminazene-inspired diaryl mono- and bis-guanyl hydrazones. A correlation between their predicted docking affinities for the acidic pocket (AcP site) in chicken ASIC1 and their inhibition of homo- and heteromeric hASIC1 channels in HEK-293 cells was found. Their activity on murine ASIC1a currents and their selectivity vs mASIC2a were assessed in engineered CHO-K1 cells, highlighting a limited isoform selectivity. Neuroprotective effects were confirmed in vitro, on primary rat cortical neurons exposed to oxygen-glucose deprivation followed by reoxygenation, and in vivo, in ischemic mice. Early lead 3b, showing a good selectivity for hASIC1 in human neurons, was neuroprotective against focal ischemia induced in mice.


Asunto(s)
Bloqueadores del Canal Iónico Sensible al Ácido/uso terapéutico , Canales Iónicos Sensibles al Ácido/metabolismo , Guanidinas/uso terapéutico , Hidrazonas/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Bloqueadores del Canal Iónico Sensible al Ácido/síntesis química , Bloqueadores del Canal Iónico Sensible al Ácido/metabolismo , Canales Iónicos Sensibles al Ácido/química , Animales , Sitios de Unión , Células CHO , Pollos , Cricetulus , Diseño de Fármacos , Guanidinas/síntesis química , Guanidinas/metabolismo , Células HEK293 , Humanos , Hidrazonas/síntesis química , Hidrazonas/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Estructura Molecular , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/metabolismo , Unión Proteica , Ratas , Relación Estructura-Actividad
3.
J Nanobiotechnology ; 17(1): 49, 2019 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-30943991

RESUMEN

BACKGROUND: The increasing use of gold nanoparticles (AuNPs) in the field of neuroscience instilled hope for their rapid translation to the clinical practice. AuNPs can be engineered to carry therapeutics or diagnostics in the diseased brain, possibly providing greater cell specificity and low toxicity. Although there is a general enthusiasm for these tools, we are in early stages of their development. Overall, their brain penetrance, stability and cell specificity are critical issues that must be addressed to drive AuNPs to the clinic. RESULTS: We studied the kinetic, distribution and stability of PEG-coated AuNPs in mice receiving a single injection into the cisterna magna of the 4th ventricle. AuNPs were conjugated with the fluorescent tag Cy5.5 (Cy5.5-AuNPs) to track their in vivo distribution. Fluorescence levels from such particles were detected in mice for weeks. In situ analysis of brains by immunofluorescence and electron microscopy revealed that Cy5.5-AuNPs penetrated the brain parenchyma, spreading in the CNS parenchyma beneath the 4th ventricle. Cy5.5-AuNPs were preferentially found in neurons, although a subset of resting microglia also entrapped these particles. CONCLUSIONS: Our results suggest that the ICM route for delivering gold particles allows the targeting of neurons. This approach might be pursued to carry therapeutics or diagnostics inside a diseased brain with a surgical procedure that is largely used in gene therapy approaches. Furthermore, this approach could be used for radiotherapy, enhancing the agent's efficacy to kill brain cancer cells.


Asunto(s)
Encéfalo/metabolismo , Oro/química , Nanopartículas del Metal/química , Polietilenglicoles/química , Animales , Carbocianinas/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cisterna Magna , Estabilidad de Medicamentos , Colorantes Fluorescentes/química , Humanos , Ratones , Neuronas/citología , Neuronas/efectos de los fármacos , Permeabilidad , Distribución Tisular
4.
ACS Med Chem Lett ; 10(4): 627-632, 2019 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-30996808

RESUMEN

Acid-sensing ion channels (ASICs) are a family of ion channels permeable to cations and largely responsible for the onset of acid-evoked ion currents both in neurons and in different types of cancer cells, thus representing a potential target for drug discovery. Owing to the limited attention ASIC2 has received so far, an exploratory program was initiated to identify ASIC2 inhibitors using diminazene, a known pan-ASIC inhibitor, as a chemical starting point for structural elaboration. The performed exploration enabled the identification of a novel series of ASIC2 inhibitors. In particular, compound 2u is a brain penetrant ASIC2 inhibitor endowed with an optimal pharmacokinetic profile. This compound may represent a useful tool to validate in animal models in vivo the role of ASIC2 in different neurodegenerative central nervous system pathologies.

5.
Neuropharmacology ; 148: 366-376, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30716415

RESUMEN

Acid-sensing ion channels (ASICs) are proton-activated, sodium-permeable channels, highly expressed in both central and peripheral nervous systems. ASIC1a is the most abundant isoform in the central nervous system and is credited to be involved in several neurological disorders including stroke, multiple sclerosis, and epilepsy. Interestingly, the affinity of ASIC1a for two antagonists, diminazene and amiloride, has recently been proposed to be voltage sensitive. Based on this evidence, it is expected that the pharmacology of ASIC1cannot be properly characterized by single-cell voltage-clamp, an experimental condition in which membrane potential is maintained close to resting values. In particular, these measurements do not take into account the influence of the membrane potential depolarization induced by ASIC1a activation during acidosis or neuronal activity. We show here the voltage-dependence of some small molecules antagonists (diminazene, amiloride and a new patented drug from Merck), but not of Psalmotoxin 1, a peptide binding to regions other than the pore. We also demonstrate that the opening of ASIC1a induced by moderate acidosis determines a depolarization sufficient to change the affinity of small molecule antagonists. The characterization of this mechanism was performed on CHO-K1 expressing ASIC1a and further confirmed in hippocampal neurons in culture. Finally, perforated-patch experiments indicate that intracellular modulations do not play a role in the voltage-dependent binding of small molecules. Since ASIC1a activation promotes a membrane depolarization that may influence the binding of small molecules, we propose to adopt experimental methods that do not interfere with the membrane potential for the drug screening of ASIC1a modulators.


Asunto(s)
Bloqueadores del Canal Iónico Sensible al Ácido/farmacología , Canales Iónicos Sensibles al Ácido/fisiología , Potenciales de la Membrana/fisiología , Acidosis/fisiopatología , Amilorida/farmacología , Animales , Células Cultivadas , Cricetinae , Diminazeno/farmacología , Hipocampo/fisiología , Neuronas/fisiología , Venenos de Araña/farmacología
6.
Cell Death Dis ; 9(6): 698, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29899471

RESUMEN

The pathological cascade leading from primary storage to neural cell dysfunction and death in metachromatic leukodystrophy (MLD) has been poorly elucidated in human-derived neural cell systems. In the present study, we have modeled the progression of pathological events during the differentiation of patient-specific iPSCs to neuroepithelial progenitor cells (iPSC-NPCs) and mature neurons, astrocytes, and oligodendrocytes at the morphological, molecular, and biochemical level. We showed significant sulfatide accumulation and altered sulfatide composition during the differentiation of MLD iPSC-NPCs into neuronal and glial cells. Changes in sulfatide levels and composition were accompanied by the expansion of the lysosomal compartment, oxidative stress, and apoptosis. The neuronal and glial differentiation capacity of MLD iPSC-NPCs was significantly impaired. We showed delayed appearance and/or reduced levels of oligodendroglial and astroglial markers as well as reduced number of neurons and disorganized neuronal network. Restoration of a functional Arylsulfatase A (ARSA) enzyme in MLD cells using lentiviral-mediated gene transfer normalized sulfatide levels and composition, globally rescuing the pathological phenotype. Our study points to MLD iPSC-derived neural progeny as a useful in vitro model to assess the impact of ARSA deficiency along NPC differentiation into neurons and glial cells. In addition, iPSC-derived neural cultures allowed testing the impact of ARSA reconstitution/overexpression on disease correction and, importantly, on the biology and functional features of human NPCs, with important therapeutic implications.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Leucodistrofia Metacromática/patología , Modelos Biológicos , Células-Madre Neurales/patología , Neuroglía/patología , Neuronas/patología , Apoptosis , Glicoesfingolípidos/biosíntesis , Humanos , Lisosomas/metabolismo , Degeneración Nerviosa/patología , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Sulfoglicoesfingolípidos/metabolismo
7.
Sci Rep ; 6: 37540, 2016 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-27857203

RESUMEN

The CRISPR/Cas9 system is a rapid and customizable tool for gene editing in mammalian cells. In particular, this approach has widely opened new opportunities for genetic studies in neurological disease. Human neurons can be differentiated in vitro from hPSC (human Pluripotent Stem Cells), hNPCs (human Neural Precursor Cells) or even directly reprogrammed from fibroblasts. Here, we described a new platform which enables, rapid and efficient CRISPR/Cas9-mediated genome targeting simultaneously with three different paradigms for in vitro generation of neurons. This system was employed to inactivate two genes associated with neurological disorder (TSC2 and KCNQ2) and achieved up to 85% efficiency of gene targeting in the differentiated cells. In particular, we devised a protocol that, combining the expression of the CRISPR components with neurogenic factors, generated functional human neurons highly enriched for the desired genome modification in only 5 weeks. This new approach is easy, fast and that does not require the generation of stable isogenic clones, practice that is time consuming and for some genes not feasible.


Asunto(s)
Diferenciación Celular/genética , Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/clasificación , Sistemas CRISPR-Cas/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Silenciador del Gen , Vectores Genéticos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo
8.
J Biomol Screen ; 21(4): 372-80, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26597957

RESUMEN

Acid-sensing ion channel 1a (ASIC1a) is involved in several pathologies, including neurodegenerative and neuroinflammatory disorders, stroke, epilepsy, and inflammatory pain. ASIC1a has been the subject of intense drug discovery programs devoted to the development of new pharmacological tools for its modulation. However, these efforts to generate new compounds have faced the lack of an efficient screening procedure. In the past decades, improvements in screening technologies and fluorescent sensors for the study of ion channels have provided new opportunities in this field. Unfortunately, ASIC1a is mainly a Na(+) permeable channel and undergoes desensitization after its activation, two features that make the use of the available screening procedures problematic. We propose here a novel screening approach for the study of ASIC1a activity in full automation. Our method is based on the stimulation of ASIC1a-expressing cells by protons and the use of electrochromic fluorescent voltage sensors as a readout of ion channel activation. This method will prove to be useful for drug screening programs aimed at ASIC1a modulation.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Ensayos Analíticos de Alto Rendimiento , Neuronas/efectos de los fármacos , Piranos/metabolismo , Compuestos de Piridinio/metabolismo , Estirenos/metabolismo , Imagen de Colorante Sensible al Voltaje/métodos , Canales Iónicos Sensibles al Ácido/genética , Animales , Automatización de Laboratorios , Células CHO , Cationes Monovalentes , Línea Celular , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Cricetulus , Diminazeno/análogos & derivados , Diminazeno/farmacología , Colorantes Fluorescentes/metabolismo , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/metabolismo , Técnicas de Placa-Clamp , Péptidos/farmacología , Cultivo Primario de Células , Sodio/metabolismo , Venenos de Araña/farmacología , Transgenes
9.
Biotechnol Bioeng ; 113(2): 403-13, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26301335

RESUMEN

Two binding requirements for in vitro studies on long-term neuronal networks dynamics are (i) finely controlled environmental conditions to keep neuronal cultures viable and provide reliable data for more than a few hours and (ii) parallel operation on multiple neuronal cultures to shorten experimental time scales and enhance data reproducibility. In order to fulfill these needs with a Microelectrode Arrays (MEA)-based system, we designed a stand-alone device that permits to uninterruptedly monitor neuronal cultures activity over long periods, overcoming drawbacks of existing MEA platforms. We integrated in a single device: (i) a closed chamber housing four MEAs equipped with access for chemical manipulations, (ii) environmental control systems and embedded sensors to reproduce and remotely monitor the standard in vitro culture environment on the lab bench (i.e. in terms of temperature, air CO2 and relative humidity), and (iii) a modular MEA interface analog front-end for reliable and parallel recordings. The system has been proven to assure environmental conditions stable, physiological and homogeneos across different cultures. Prolonged recordings (up to 10 days) of spontaneous and pharmacologically stimulated neuronal culture activity have not shown signs of rundown thanks to the environmental stability and have not required to withdraw the cells from the chamber for culture medium manipulations. This system represents an effective MEA-based solution to elucidate neuronal network phenomena with slow dynamics, such as long-term plasticity, effects of chronic pharmacological stimulations or late-onset pathological mechanisms.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Microelectrodos , Neuronas/fisiología , Dióxido de Carbono/análisis , Células Cultivadas , Humedad , Temperatura
10.
Brain Behav Immun ; 45: 263-76, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25499583

RESUMEN

Local acidosis is associated with neuro-inflammation and can have significant effects in several neurological disorders, including multiple sclerosis, brain ischemia, spinal cord injury and epilepsy. Despite local acidosis has been implicated in numerous pathological functions, very little is known about the modulatory effects of pathological acidosis on the activity of neuronal networks and on synaptic structural properties. Using non-invasive MRI spectroscopy we revealed protracted extracellular acidosis in the CNS of Experimental Autoimmune Encephalomyelitis (EAE) affected mice. By multi-unit recording in cortical neurons, we established that acidosis affects network activity, down-sizing firing and bursting behaviors as well as amplitudes. Furthermore, a protracted acidosis reduced the number of presynaptic terminals, while it did not affect the postsynaptic compartment. Application of the diarylamidine Diminazene Aceturate (DA) during acidosis significantly reverted both the loss of neuronal firing and bursting and the reduction of presynaptic terminals. Finally, in vivo DA delivery ameliorated the clinical disease course of EAE mice, reducing demyelination and axonal damage. DA is known to block acid-sensing ion channels (ASICs), which are proton-gated, voltage-insensitive, Na(+) permeable channels principally expressed by peripheral and central nervous system neurons. Our data suggest that ASICs activation during acidosis modulates network electrical activity and exacerbates neuro-degeneration in EAE mice. Therefore pharmacological modulation of ASICs in neuroinflammatory diseases could represent a new promising strategy for future therapies aimed at neuro-protection.


Asunto(s)
Bloqueadores del Canal Iónico Sensible al Ácido/farmacología , Canales Iónicos Sensibles al Ácido/metabolismo , Acidosis/metabolismo , Encéfalo/metabolismo , Diminazeno/análogos & derivados , Encefalomielitis Autoinmune Experimental/metabolismo , Vaina de Mielina/metabolismo , Neuronas/metabolismo , Terminales Presinápticos/metabolismo , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Axones/patología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Diminazeno/farmacología , Concentración de Iones de Hidrógeno , Espectroscopía de Resonancia Magnética , Ratones , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Terminales Presinápticos/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos
11.
PLoS One ; 8(12): e83899, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24386305

RESUMEN

It is known that cell density influences the maturation process of in vitro neuronal networks. Neuronal cultures plated with different cell densities differ in number of synapses per neuron and thus in single neuron synaptic transmission, which results in a density-dependent neuronal network activity. Although many authors provided detailed information about the effects of cell density on neuronal culture activity, a dedicated report of density and age influence on neuronal hippocampal culture activity has not yet been reported. Therefore, this work aims at providing reference data to researchers that set up an experimental study on hippocampal neuronal cultures, helping in planning and decoding the experiments. In this work, we analysed the effects of both neuronal density and culture age on functional attributes of maturing hippocampal cultures. We characterized the electrophysiological activity of neuronal cultures seeded at three different cell densities, recording their spontaneous electrical activity over maturation by means of MicroElectrode Arrays (MEAs). We had gather data from 86 independent hippocampal cultures to achieve solid statistic results, considering the high culture-to-culture variability. Network activity was evaluated in terms of simple spiking, burst and network burst features. We observed that electrical descriptors were characterized by a functional peak during maturation, followed by a stable phase (for sparse and medium density cultures) or by a decrease phase (for high dense neuronal cultures). Moreover, 900 cells/mm(2) cultures showed characteristics suitable for long lasting experiments (e.g. chronic effect of drug treatments) while 1800 cells/mm(2) cultures should be preferred for experiments that require intense electrical activity (e.g. to evaluate the effect of inhibitory molecules). Finally, cell cultures at 3600 cells/mm(2) are more appropriate for experiments in which time saving is relevant (e.g. drug screenings). These results are intended to be a reference for the planning of in vitro neurophysiological and neuropharmacological experiments with MEAs.


Asunto(s)
Hipocampo/citología , Red Nerviosa/citología , Neuronas/citología , Animales , Recuento de Células , Células Cultivadas , Fenómenos Electrofisiológicos , Hipocampo/fisiología , Ratones , Red Nerviosa/fisiología
12.
Biomicrofluidics ; 6(2): 24106-2410610, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22655017

RESUMEN

Spatially and temporally resolved delivery of soluble factors is a key feature for pharmacological applications. In this framework, microfluidics coupled to multisite electrophysiology offers great advantages in neuropharmacology and toxicology. In this work, a microfluidic device for biochemical stimulation of neuronal networks was developed. A micro-chamber for cell culturing, previously developed and tested for long term neuronal growth by our group, was provided with a thin wall, which partially divided the cell culture region in two sub-compartments. The device was reversibly coupled to a flat micro electrode array and used to culture primary neurons in the same microenvironment. We demonstrated that the two fluidically connected compartments were able to originate two parallel neuronal networks with similar electrophysiological activity but functionally independent. Furthermore, the device allowed to connect the outlet port to a syringe pump and to transform the static culture chamber in a perfused one. At 14 days invitro, sub-networks were independently stimulated with a test molecule, tetrodotoxin, a neurotoxin known to block action potentials, by means of continuous delivery. Electrical activity recordings proved the ability of the device configuration to selectively stimulate each neuronal network individually. The proposed microfluidic approach represents an innovative methodology to perform biological, pharmacological, and electrophysiological experiments on neuronal networks. Indeed, it allows for controlled delivery of substances to cells, and it overcomes the limitations due to standard drug stimulation techniques. Finally, the twin network configuration reduces biological variability, which has important outcomes on pharmacological and drug screening.

13.
Biotechnol Bioeng ; 109(1): 166-75, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21858786

RESUMEN

In vitro recording of neuronal electrical activity is a widely used technique to understand brain functions and to study the effect of drugs on the central nervous system. The integration of microfluidic devices with microelectrode arrays (MEAs) enables the recording of networks activity in a controlled microenvironment. In this work, an integrated microfluidic system for neuronal cultures was developed, reversibly coupling a PDMS microfluidic device with a commercial flat MEA through magnetic forces. Neurons from mouse embryos were cultured in a 100 µm channel and their activity was followed up to 18 days in vitro. The maturation of the networks and their morphological and functional characteristics were comparable with those of networks cultured in macro-environments and described in literature. In this work, we successfully demonstrated the ability of long-term culturing of primary neuronal cells in a reversible bonded microfluidic device (based on magnetism) that will be fundamental for neuropharmacological studies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Microelectrodos , Técnicas Analíticas Microfluídicas/métodos , Red Nerviosa/fisiología , Neuronas/fisiología , Animales , Células Cultivadas , Magnetismo , Ratones , Factores de Tiempo
14.
Nature ; 476(7359): 224-7, 2011 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-21725324

RESUMEN

Transplantation of dopaminergic neurons can potentially improve the clinical outcome of Parkinson's disease, a neurological disorder resulting from degeneration of mesencephalic dopaminergic neurons. In particular, transplantation of embryonic-stem-cell-derived dopaminergic neurons has been shown to be efficient in restoring motor symptoms in conditions of dopamine deficiency. However, the use of pluripotent-derived cells might lead to the development of tumours if not properly controlled. Here we identified a minimal set of three transcription factors--Mash1 (also known as Ascl1), Nurr1 (also known as Nr4a2) and Lmx1a--that are able to generate directly functional dopaminergic neurons from mouse and human fibroblasts without reverting to a progenitor cell stage. Induced dopaminergic (iDA) cells release dopamine and show spontaneous electrical activity organized in regular spikes consistent with the pacemaker activity featured by brain dopaminergic neurons. The three factors were able to elicit dopaminergic neuronal conversion in prenatal and adult fibroblasts from healthy donors and Parkinson's disease patients. Direct generation of iDA cells from somatic cells might have significant implications for understanding critical processes for neuronal development, in vitro disease modelling and cell replacement therapies.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Dopamina/metabolismo , Fibroblastos/citología , Neuronas/citología , Neuronas/metabolismo , Potenciales de Acción , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Reprogramación Celular/genética , Reprogramación Celular/fisiología , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Proteínas con Homeodominio LIM , Ratones , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Enfermedad de Parkinson/patología , Técnicas de Placa-Clamp , Medicina Regenerativa , Piel/citología , Factores de Transcripción
15.
Brain Behav Immun ; 25(5): 947-56, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20940040

RESUMEN

Synaptic dysfunction triggers neuronal damage in experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). While excessive glutamate signaling has been reported in the striatum of EAE, it is still uncertain whether GABA synapses are altered. Electrophysiological recordings showed a reduction of spontaneous GABAergic synaptic currents (sIPSCs) recorded from striatal projection neurons of mice with MOG((35-55))-induced EAE. GABAergic sIPSC deficits started in the acute phase of the disease (20-25days post immunization, dpi), and were exacerbated at later time-points (35, 50, 70 and 90dpi). Of note, in slices they were independent of microglial activation and of release of TNF-α. Indeed, sIPSC inhibition likely involved synaptic inputs arising from GABAergic interneurons, because EAE preferentially reduced sIPSCs of high amplitude, and was associated with a selective loss of striatal parvalbumin (PV)-positive GABAergic interneurons, which contact striatal projection neurons in their somatic region, giving rise to more efficient synaptic inhibition. Furthermore, we found also that the chronic persistence of pro-inflammatory cytokines were able, per se, to produce profound alterations of electrophysiological network properties, that were reverted by GABA administration. The results of the present investigation indicate defective GABA transmission in MS models depending from alteration of PV cells number and, in part, deriving from the effects of a chronic inflammation, and suggest that pharmacological agents potentiating GABA signaling might be considered to limit neuronal damage in MS patients.


Asunto(s)
Cuerpo Estriado/fisiopatología , Encefalomielitis Autoinmune Experimental/fisiopatología , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Western Blotting , Células Cultivadas , Cuerpo Estriado/metabolismo , Citocinas/fisiología , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/fisiología
16.
J Biomol Screen ; 15(9): 1132-43, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20834010

RESUMEN

The use of engineered mouse embryonic stem (mES) cells in high-throughput screening (HTS) can offer new opportunities for studying complex targets in their native environment, increasing the probability of discovering more meaningful hits. The authors have generated and developed a mouse embryonic stem cell line called c-Photina mES stably expressing a Ca(2+)-activated photoprotein as a reporter gene. This reporter cell line retains the ability to differentiate into any cell lineage and can be used for miniaturized screening processes in 384-well microplates. The c-Photina mES cell line is particularly well suited for the study of the pharmacological modulation of target genes that induce Ca(2+) mobilization. The authors differentiated this mES reporter cell line into neuronal cells and screened the LOPAC(1280) library monitoring the agonistic or antagonistic activities of compounds. They also demonstrate the possibility to generate and freeze bulk preparations of cells at an intermediate stage of differentiation and enriched in neural precursor cells, which retain the ability to form fully functional neural networks once thawed. The proposed cell model is of high value for HTS purposes because it offers a more physiological environment to the targets of interest and the possibility of using frozen batches of neural precursor cells.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Células Madre Embrionarias/citología , Ensayos Analíticos de Alto Rendimiento/métodos , Células-Madre Neurales/citología , Neuronas/citología , Animales , Bioensayo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Ratones , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Compuestos de Piridinio/metabolismo , Compuestos de Amonio Cuaternario/metabolismo , Bibliotecas de Moléculas Pequeñas/análisis , Bibliotecas de Moléculas Pequeñas/farmacología
17.
PLoS One ; 5(1): e8882, 2010 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-20111708

RESUMEN

Exogenous expression of pharmacological targets in transformed cell lines has been the traditional platform for high throughput screening of small molecules. However, exogenous expression in these cells is limited by aberrant dosage, or its toxicity, the potential lack of interaction partners, and alterations to physiology due to transformation itself. Instead, primary cells or cells differentiated from precursors are more physiological, but less amenable to exogenous expression of reporter systems. To overcome this challenge, we stably expressed c-Photina, a Ca(2+)-sensitive photoprotein, driven by a ubiquitous promoter in a mouse embryonic stem (mES) cell line. The same embryonic stem cell line was also used to generate a transgenic mouse that expresses c-Photina in most tissues. We show here that these cells and mice provide an efficient source of primary cells, cells differentiated from mES cells, including cardiomyocytes, neurons, astrocytes, macrophages, endothelial cells, pancreatic islet cells, stably and robustly expressing c-Photina, and may be exploited for miniaturized high throughput screening. Moreover, we provide evidence that the transgenic mice may be suitable for ex-vivo bioimaging studies in both cells and tissues.


Asunto(s)
Calcio/metabolismo , Células Madre Embrionarias/metabolismo , Proteínas Luminiscentes/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Ratones , Ratones Transgénicos
18.
J Cell Sci ; 121(Pt 22): 3757-69, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18940911

RESUMEN

The growth-cone plasma membrane constantly reconfigures during axon navigation and upon target recognition. The identity and regulation of the membrane pathway(s) participating in remodeling of the growth-cone surface remain elusive. Here, we identify a constitutive, high-capacity plasma-membrane-recycling activity in the axonal growth cones, which is mediated by a novel bulk endocytic pathway that is mechanistically related to macropinocytosis. This pathway generates large compartments at sites of intense actin-based membrane ruffling through the actions of phosphatidylinositol 3-kinase, the small GTPase Rac1 and the pinocytic chaperone Pincher. At early developmental stages, bulk endocytosis is the primary endocytic pathway for rapid retrieval of the growth-cone plasma membrane. At later stages, during the onset of synaptogenesis, an intrinsic program of maturation leads to downregulation of basal bulk endocytosis and the emergence of depolarization-induced synaptic-vesicle exo-endocytosis. We propose that the control of bulk membrane retrieval contributes to the homeostatic regulation of the axonal plasma membrane and to growth-cone remodeling during axonal outgrowth. In addition, we suggest that the downregulation of bulk endocytosis during synaptogenesis might contribute to the preservation of synaptic-vesicle specificity.


Asunto(s)
Membrana Celular/metabolismo , Conos de Crecimiento/metabolismo , Hipocampo/crecimiento & desarrollo , Neuronas/citología , Neuronas/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Endocitosis , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , Neurogénesis , Ratas , Ratas Sprague-Dawley , Vesículas Sinápticas/metabolismo
19.
J Neurosci Methods ; 175(1): 70-8, 2008 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-18761373

RESUMEN

Optical stimulation is a promising approach to investigate the local dynamic responses of cultured neurons. In particular, flash photolysis of caged compounds offers the advantage of allowing the rapid change of concentration of either extracellular or intracellular molecules, such as neurotransmitters or second messengers, for the stimulation or modulation of neuronal activity. We describe here the use of an ultra-violet (UV) laser diode coupled to an optical fibre for the local activation of caged compounds combined with a Micro-Electrode Array (MEA) device. Local uncaging was achieved by UV irradiation through the optical fibre previously positioned by using a red laser diode. The size of the stimulation was determined using caged fluorescein, whereas its efficacy was tested by studying the effect of uncaging the neurotransmitter glutamate. Uncaged glutamate evoked neuronal responses that were recorded using either fluorescence measurements or electrophysiological recordings with MEAs, thus showing the ability of our system to induce local neuronal excitation. This method allows overcoming the limitations of the MEA system related to unfocused electrical stimulation and induction of electrical artefacts. In addition, the coupling of a UV laser diode to an optical fibre allows a precise local stimulation and a quick change of the stimulation point.


Asunto(s)
Glutamatos/farmacología , Indoles/farmacología , Rayos Láser , Microelectrodos , Neuronas/efectos de los fármacos , Óptica y Fotónica/instrumentación , Potenciales de Acción/efectos de los fármacos , Anestésicos Locales/farmacología , Animales , Calcio/metabolismo , Células Cultivadas , Embrión de Mamíferos , Fluorescencia , Hipocampo/citología , Red Nerviosa/efectos de los fármacos , Fibras Ópticas , Óptica y Fotónica/métodos , Técnicas de Placa-Clamp , Fotólisis , Ratas , Tetrodotoxina/farmacología , Factores de Tiempo
20.
J Cell Sci ; 120(Pt 18): 3228-37, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17726061

RESUMEN

Post-tetanic potentiation (PTP) is a form of homosynaptic plasticity important for information processing and short-term memory in the nervous system. The synapsins, a family of synaptic vesicle (SV)-associated phosphoproteins, have been implicated in PTP. Although several synapsin functions are known to be regulated by phosphorylation by multiple protein kinases, the role of individual phosphorylation sites in synaptic plasticity is poorly understood. All the synapsins share a phosphorylation site in the N-terminal domain A (site 1) that regulates neurite elongation and SV mobilization. Here, we have examined the role of phosphorylation of synapsin domain A in PTP and other forms of short-term synaptic enhancement (STE) at synapses between cultured Helix pomatia neurons. To this aim, we cloned H. pomatia synapsin (helSyn) and overexpressed GFP-tagged wild-type helSyn or site-1-mutant helSyn mutated in the presynaptic compartment of C1-B2 synapses. We found that PTP at these synapses depends both on Ca2+/calmodulin-dependent and cAMP-dependent protein kinases, and that overexpression of the non-phosphorylatable helSyn mutant, but not wild-type helSyn, specifically impairs PTP, while not altering facilitation and augmentation. Our findings show that phosphorylation of site 1 has a prominent role in the expression of PTP, thus defining a novel role for phosphorylation of synapsin domain A in short-term homosynaptic plasticity.


Asunto(s)
Caracoles Helix/fisiología , Memoria a Corto Plazo/fisiología , Plasticidad Neuronal/fisiología , Terminales Presinápticos/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Sinapsinas/metabolismo , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Expresión Génica , Mutación , Fosforilación , Estructura Terciaria de Proteína/genética , Sinapsinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA