Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Biosci (Landmark Ed) ; 27(4): 115, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35468674

RESUMEN

BACKGROUND: Mitochondrial biogenesis occurs in response to chronic stresses as an adaptation to the increased energy demands and often renders cells more refractive to subsequent injuries which is referred to as preconditioning. This phenomenon is observed in several non-neuronal cell types, but it is not yet fully established in neurons, although it is fundamentally important for neuroprotection and could be exploited for therapeutic purposes. METHODS: This study was designed to examine whether the preconditioning treatment with hypoxia or nitric oxide could trigger biogenesis in undifferentiated and differentiated neuronal cells (rat PC12 and human NT2 cells) as well as in primary mouse cortical neurons. RESULTS: The results showed that both preconditioning paradigms induced mitochondrial biogenesis in undifferentiated cell lines, as indicated by an increase of mitochondrial mass (measured by flow cytometry of NAO fluorescence) and increased expression of genes required for mitochondrial biogenesis (Nrf1, Nrf2, Tfam, Nfκb1) and function (Cox3, Hk1). All these changes translated into an increase in the organelle copy number from an average of 20-40 to 40-60 mitochondria per cell. The preconditioning treatments also rendered the cells significantly less sensitive to the subsequent oxidative stress challenge brought about by oxygen/glucose deprivation, consistent with their improved cellular energy status. Mitochondrial biogenesis was abolished when preconditioning treatments were performed in the presence of antioxidants (vitamin E or CoQ10), indicating clearly that ROS-signaling pathway(s) played a critical role in the induction of this phenomenon in undifferentiated cells. However, mitochondrial biogenesis could not be re-initiated by preconditioning treatments in any of the post-mitotic neuronal cells tested, i.e., neither rat PC12 cells differentiated with NGF, human NT2 cells differentiated with retinoic acid nor mouse primary cortical neurons. Instead, differentiated neurons had a much higher organelle copy number per cell than their undifferentiated counterparts (100-130 mitochondria per neuron vs. 20-40 in proliferating cells), and this feature was not altered by preconditioning. CONCLUSIONS: Our study demonstrates that mitochondrial biogenesis occurred during the differentiation process resulting in more beneficial energy status and improved tolerance to oxidative stress in neurons, putting in doubt whether additional enhancement of this phenomenon could be achieved and successfully exploited as a way for better neuroprotection.


Asunto(s)
Neuronas , Biogénesis de Organelos , Animales , Diferenciación Celular , Ratones , Mitocondrias/metabolismo , Neuronas/metabolismo , Ratas , Transducción de Señal
2.
Methods Mol Biol ; 2454: 397-410, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33881753

RESUMEN

The development of translational and predictive models in vitro for assessing blood-brain barrier (BBB) delivery has become an important requirement in preclinical testing of CNS-targeting therapeutics. Here we describe a directed monolayer differentiation strategy to generate a population of brain endothelial-like cells (BECs) from human induced pluripotent stem cell (iPSC) with robust BBB properties. To generate BBB permeability assays, the BECs are seeded as a monolayer on a semipermeable Transwell insert placed inside a companion plate to generate a two-compartment Transwell model. The BECs provide a BBB-like separation between the luminal (blood) and abluminal (brain) compartments to assess BBB permeability of CNS-targeting therapeutics.


Asunto(s)
Barrera Hematoencefálica , Células Madre Pluripotentes Inducidas , Encéfalo , Células Cultivadas , Células Endoteliales , Humanos , Permeabilidad
3.
Stem Cell Rev Rep ; 18(1): 259-277, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34687385

RESUMEN

Human induced pluripotent stem cell (iPSC)-derived neurons are of interest for studying neurological disease mechanisms, developing potential therapies and deepening our understanding of the human nervous system. However, compared to an extensive history of practice with primary rodent neuron cultures, human iPSC-neurons still require more robust characterization of expression of neuronal receptors and ion channels and functional and predictive pharmacological responses. In this study, we differentiated human amniotic fluid-derived iPSCs into a mixed population of neurons (AF-iNs). Functional assessments were performed by evaluating electrophysiological (patch-clamp) properties and the effect of a panel of neuropharmacological agents on spontaneous activity (multi-electrode arrays; MEAs). These electrophysiological data were benchmarked relative to commercially sourced human iPSC-derived neurons (CNS.4U from Ncardia), primary human neurons (ScienCell™) and primary rodent cortical/hippocampal neurons. Patch-clamp whole-cell recordings showed that mature AF-iNs generated repetitive firing of action potentials in response to depolarizations, similar to that of primary rodent cortical/hippocampal neurons, with nearly half of the neurons displaying spontaneous post-synaptic currents. Immunochemical and MEA-based analyses indicated that AF-iNs were composed of functional glutamatergic excitatory and inhibitory GABAergic neurons. Principal component analysis of MEA data indicated that human AF-iN and rat neurons exhibited distinct pharmacological and electrophysiological properties. Collectively, this study establishes a necessary prerequisite for AF-iNs as a human neuron culture model suitable for pharmacological studies.


Asunto(s)
Células Madre Pluripotentes Inducidas , Animales , Benchmarking , Fenómenos Electrofisiológicos , Humanos , Neuronas , Ratas , Roedores
4.
Fluids Barriers CNS ; 15(1): 15, 2018 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-29759080

RESUMEN

Zika virus (ZIKV) is a flavivirus that is highly neurotropic causing congenital abnormalities and neurological damage to the central nervous systems (CNS). In this study, we used a human induced pluripotent stem cell (iPSC)-derived blood brain barrier (BBB) model to demonstrate that ZIKV can infect brain endothelial cells (i-BECs) without compromising the BBB barrier integrity or permeability. Although no disruption to the BBB was observed post-infection, ZIKV particles were released on the abluminal side of the BBB model and infected underlying iPSC-derived neural progenitor cells (i-NPs). AXL, a putative ZIKV cellular entry receptor, was also highly expressed in ZIKV-susceptible i-BEC and i-NPs. This iPSC-derived BBB model can help elucidate the mechanism by which ZIKV can infect BECs, cross the BBB and gain access to the CNS.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/virología , Virus Zika/metabolismo , Permeabilidad Capilar/fisiología , Técnicas de Cultivo de Célula , Células Endoteliales/metabolismo , Células Endoteliales/virología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/virología , Microvasos/metabolismo , Microvasos/virología , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología
5.
Sci Rep ; 8(1): 1873, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382846

RESUMEN

We have developed a renewable, scalable and transgene free human blood-brain barrier model, composed of brain endothelial cells (BECs), generated from human amniotic fluid derived induced pluripotent stem cells (AF-iPSC), which can also give rise to syngeneic neural cells of the neurovascular unit. These AF-iPSC-derived BECs (i-BEC) exhibited high transendothelial electrical resistance (up to 1500 Ω cm2) inducible by astrocyte-derived molecular cues and retinoic acid treatment, polarized expression of functional efflux transporters and receptor mediated transcytosis triggered by antibodies against specific receptors. In vitro human BBB models enable pre-clinical screening of central nervous system (CNS)-targeting drugs and are of particular importance for assessing species-specific/selective transport mechanisms. This i-BEC human BBB model discriminates species-selective antibody- mediated transcytosis mechanisms, is predictive of in vivo CNS exposure of rodent cross-reactive antibodies and can be implemented into pre-clinical CNS drug discovery and development processes.


Asunto(s)
Anticuerpos/farmacología , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Receptores de Superficie Celular/metabolismo , Transcitosis/fisiología , Animales , Astrocitos/citología , Astrocitos/fisiología , Transporte Biológico , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/fisiología , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Masculino , Neuronas/citología , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/antagonistas & inhibidores
6.
Biochem Cell Biol ; 96(4): 391-406, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29370536

RESUMEN

Traumatic brain injury (TBI) is a leading cause of morbidity and mortality worldwide. Due to its high incidence rate and often long-term sequelae, TBI contributes significantly to increasing costs of health care expenditures annually. Unfortunately, advances in the field have been stifled by patient and injury heterogeneity that pose a major challenge in TBI prevention, diagnosis, and treatment. In this review, we briefly discuss the causes of TBI, followed by its prevalence, classification, and pathophysiology. The current imaging detection methods and animal models used to study brain injury are examined. We discuss the potential use of molecular markers in detecting and monitoring the progression of TBI, with particular emphasis on microRNAs as a novel class of molecular modulators of injury and its repair in the neural tissue.


Asunto(s)
Biomarcadores/análisis , Lesiones Traumáticas del Encéfalo , Neuroimagen Funcional , MicroARNs/uso terapéutico , Animales , Encéfalo/diagnóstico por imagen , Lesiones Traumáticas del Encéfalo/diagnóstico , Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Lesiones Traumáticas del Encéfalo/terapia , Modelos Animales de Enfermedad , Humanos
7.
Can J Physiol Pharmacol ; 92(12): 1001-11, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25388371

RESUMEN

S-nitrosoglutathione (GSNO) is an endogenously produced S-nitrosylating compound that controls the function of various proteins. While a number of rodent cell lines have been used to study GSNO-induced apoptosis, the mechanisms of action remain to be evaluated in human cells and in parallel with other common apoptosis-inducing agents. In this study, we compared the pro-apoptotic effects of GSNO and staurosporine (STS) on human neural progenitors (NT2, hNP1) and neuroblasts (SH-SY5Y). We show that these cells exhibit comparable levels of susceptibility to GSNO- and STS-induced apoptotic cell death, as demonstrated by condensed nuclei and CASP3 activation. Mechanistic differences in apoptotic responses were observed as differential patterns of DNA fragmentation and levels of BAX, BCL-XL, CASP8, and p-ERK in response to GSNO and STS treatment. Mitochondrial membrane potential analysis revealed that NT2 and hNP1 cells, but not SH-SY5Y cells, undergo mitochondrial hyperpolarization in response to short-term exposure to STS prior to undergoing subsequent depolarization. This is the first study to report differences in apoptotic responses to GSNO and STS in 3 complementary human neural cell lines. Furthermore, these cells represent useful tools in cell pharmacological paradigms in which susceptibility to apoptosis-inducing agents needs to be assessed at different stages of neural cell fate commitment and differentiation.


Asunto(s)
Apoptosis/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , S-Nitrosoglutatión/farmacología , Estaurosporina/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Diferenciación Celular , Línea Celular , Núcleo Celular/patología , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , S-Nitrosoglutatión/metabolismo
8.
Stem Cell Rev Rep ; 10(2): 251-68, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24415130

RESUMEN

Brain injury continues to be one of the leading causes of disability worldwide. Despite decades of research, there is currently no pharmacologically effective treatment for preventing neuronal loss and repairing the brain. As a result, novel therapeutic approaches, such as cell-based therapies, are being actively pursued to repair tissue damage and restore neurological function after injury. In this study, we examined the neuroprotective potential of amniotic fluid (AF) single cell clones, engineered to secrete glial cell derived neurotrophic factor (AF-GDNF), both in vitro and in a surgically induced model of brain injury. Our results show that pre-treatment with GDNF significantly increases cell survival in cultures of AF cells or cortical neurons exposed to hydrogen peroxide. Since improving the efficacy of cell transplantation depends on enhanced graft cell survival, we investigated whether AF-GDNF cells seeded on polyglycolic acid (PGA) scaffolds could enhance graft survival following implantation into the lesion cavity. Encouragingly, the AF-GDNF cells survived longer than control AF cells in serum-free conditions and continued to secrete GDNF both in vitro and following implantation into the injured motor cortex. AF-GDNF implantation in the acute period following injury was sufficient to activate the MAPK/ERK signaling pathway in host neural cells in the peri-lesion area, potentially boosting endogenous neuroprotective pathways. These results were complemented with promising trends in beam walk tasks in AF-GDNF/PGA animals during the 7 day timeframe. Further investigation is required to determine whether significant behavioural improvement can be achieved at a longer timeframe.


Asunto(s)
Líquido Amniótico/citología , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Trasplante de Células Madre , Células Madre/fisiología , Animales , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Lesiones Encefálicas/terapia , Supervivencia Celular , Células Cultivadas , Femenino , Expresión Génica , Humanos , Peróxido de Hidrógeno/farmacología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Corteza Motora/patología , Células-Madre Neurales/fisiología , Oxidantes/farmacología , Estrés Oxidativo , Prótesis e Implantes , Desempeño Psicomotor , Andamios del Tejido
9.
Biochem Cell Biol ; 91(5): 271-86, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24032676

RESUMEN

There is a need for improved therapy for acquired brain injury, which has proven resistant to treatment by numerous drugs in clinical trials and continues to represent one of the leading causes of disability worldwide. Research into cell-based therapies for the treatment of brain injury is growing rapidly, but the ideal cell source has yet to be determined. Subpopulations of cells found in amniotic fluid, which is readily obtained during routine amniocentesis, can be easily expanded in culture, have multipotent differentiation capacity, are non-tumourigenic, and avoid the ethical complications associated with embryonic stem cells, making them a promising cell source for therapeutic purposes. Beneficial effects of amniotic fluid cell transplantation have been reported in various models of nervous system injury. However, evidence that amniotic fluid cells can differentiate into mature, functional neurons in vivo and incorporate into the existing circuitry to replace lost or damaged neurons is lacking. The mechanisms by which amniotic fluid cells improve outcomes after experimental nervous system injury remain unclear. However, studies reporting the expression and release of neurotrophic, angiogenic, and immunomodulatory factors by amniotic fluid cells suggest they may provide neuroprotection and (or) stimulate endogenous repair and remodelling processes in the injured nervous system. In this paper, we address recent research related to the neuronal differentiation of amniotic fluid-derived cells, the therapeutic efficacy of these cells in animal models of nervous system injury, and the possible mechanisms mediating the positive outcomes achieved by amniotic fluid cell transplantation.


Asunto(s)
Líquido Amniótico/citología , Lesiones Encefálicas/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Multipotentes/trasplante , Amniocentesis , Animales , Diferenciación Celular , Humanos , Ratones , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Neuronas/citología , Accidente Cerebrovascular/terapia , Ingeniería de Tejidos/métodos
10.
J Neurosci Methods ; 205(1): 17-27, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22209770

RESUMEN

Bone morphogenetic protein 7 (BMP7), a member of the transforming growth factor ß (TGF-ß) superfamily, plays important roles in the development of various tissues and organs in mouse and human. In particular, BMP7 is critical for the formation of the nervous system and it is considered to have therapeutic potential in brain injury and stroke. One approach to make BMP7 more suitable for therapeutic purposes is the development of efficient vectors that allow the consistent, reliable and cost-effective production of the BMP7 protein. In this study, we developed an efficient BMP7 delivery system, using a third generation lentiviral vector to produce functional BMP7 protein. The lentiviral transduction of several human cell types, including human embryonic kidney 293 (HEK293) cells, amniotic fluid cells, NTera2 neurons (NT2-N) and primary neuronal cultures resulted in BMP7 expression. The production of BMP7 protein was achieved for at least 4 weeks post-transduction, as determined by enzyme-linked immunosorbent assay (ELISA). SMAD phosphorylation and neuronal differentiation assays verified the bioactivity and functionality of the lentiviral-based BMP7 protein, respectively. In addition, the intracerebroventricular injection of the lentivirus resulted in exogenous BMP7 expression in both neurons and astrocytes in the mouse brain. Taken together, this gene delivery system provides a reliable source of functional BMP7 protein for future in vitro and in vivo studies.


Asunto(s)
Proteína Morfogenética Ósea 7/biosíntesis , Técnicas de Transferencia de Gen , Lentivirus/genética , Transfección/métodos , Líquido Amniótico/citología , Animales , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Células Cultivadas , ADN Complementario/administración & dosificación , ADN Complementario/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes , Humanos , Inmunohistoquímica , Inyecciones Intraventriculares , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Embarazo , Proteínas Smad/biosíntesis , Proteínas Smad/genética , Transducción Genética
11.
J Neurosci Methods ; 186(1): 60-7, 2010 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19903493

RESUMEN

Neuro 2A (N2a) is a mouse neural crest-derived cell line that has been extensively used to study neuronal differentiation, axonal growth and signaling pathways. A convenient characteristic of these cells is their ability to differentiate into neurons within a few days. However, most differentiation methods reported for N2a cells do not provide information about the neuronal types obtained after each treatment. In this study, we evaluated the generation of N2a dopamine neurons following treatment with a number of factors known to induce neuronal differentiation. Our results showed that N2a cells express Nurr-related factor 1 (Nurr1) and produce low levels of tyrosine hydroxylase (TH) and dopamine. Both TH and dopamine levels were significantly enhanced in the presence of dibutyryl cyclic adenosine monophosphate (dbcAMP), as evidenced by Western blot, immunocytochemistry and high performance liquid chromatography (HPLC). In contrast to dbcAMP, other factors such as transforming growth factor beta1 (TGF beta 1), bone morphogenetic protein 4 (BMP4), glial cell-derived neurotrophic factor (GDNF) and retinoic acid (RA) did not increase TH expression. Further investigation confirmed that the effect of dbcAMP on production of TH-positive neurons was mediated through cyclic AMP (cAMP) responsive element binding protein (CREB) and it was antagonized by RA. Thus, although various treatments can be used to generate N2a neurons, only dbcAMP significantly enhanced the formation of dopamine neurons. Taken together, this study provided a simple and reliable method to generate dopamine neurons for rapid and efficient physiological and pharmacological assays.


Asunto(s)
Diferenciación Celular/fisiología , Dopamina/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Células Madre/metabolismo , Animales , Bucladesina/farmacología , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ratones , Factores de Crecimiento Nervioso/farmacología , Neurogénesis/efectos de los fármacos , Neuronas/citología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos , Tretinoina/metabolismo , Tretinoina/farmacología , Tirosina 3-Monooxigenasa/metabolismo
12.
Exp Cell Res ; 316(1): 68-77, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19744480

RESUMEN

Myosin phosphatase target subunit 1 (MYPT1), together with catalytic subunit of type1 delta isoform (PP1cdelta) and a small 20-kDa regulatory unit (M20), form a heterotrimeric holoenzyme, myosin phosphatase (MP), which is responsible for regulating the extent of myosin light chain phosphorylation. Here we report the identification and characterization of a molecular interaction between Seven in absentia homolog 2 (SIAH2) and MYPT1 that resulted in the proteasomal degradation of the latter in mammalian cells, including neurons and glia. The interaction involved the substrate binding domain of SIAH2 (aa 116-324) and a central region of MYPT1 (aa 445-632) containing a degenerate consensus Siah-binding motif RLAYVAP (aa 493-499) evolutionally conserved from fish to humans. These findings suggest a novel mechanism whereby the ability of MP to modulate myosin light chain might be regulated by the degradation of its targeting subunit MYPT1 through the SIAH2-ubiquitin-proteasomal pathway. In this manner, the turnover of MYPT1 would serve to limit the duration and/or magnitude of MP activity required to achieve a desired physiological effect.


Asunto(s)
Fosfatasa de Miosina de Cadena Ligera/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Animales , Astrocitos/metabolismo , Sitios de Unión/fisiología , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Secuencia de Consenso/fisiología , Inhibidores de Cisteína Proteinasa/farmacología , Citoplasma/metabolismo , Expresión Génica/genética , Humanos , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Mutación/fisiología , Fosfatasa de Miosina de Cadena Ligera/genética , Neuronas/metabolismo , Proteínas Nucleares/genética , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Unión Proteica/fisiología , Dominios y Motivos de Interacción de Proteínas/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Transfección , Técnicas del Sistema de Dos Híbridos , Ubiquitina-Proteína Ligasas/genética
13.
Muscle Nerve ; 40(3): 381-94, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19626676

RESUMEN

The potential pathogenicity of two homoplasmic mtDNA point mutations, 9035T>C and 4452T>C, found in a family afflicted with maternally transmitted cognitive developmental delay, learning disability, and progressive ataxia was evaluated using transmitochondrial cybrids. We confirmed that the 4452T>C transition in tRNA(Met) represented a polymorphism; however, 9035T>C conversion in the ATP6 gene was responsible for a defective F(0)-ATPase. Accordingly, mutant cybrids had a reduced oligomycin-sensitive ATP hydrolyzing activity. They had less than half of the steady-state content of ATP and nearly an 8-fold higher basal level of reactive oxygen species (ROS). Mutant cybrids were unable to cope with additional insults, i.e., glucose deprivation or tertiary-butyl hydroperoxide, and they succumbed to either apoptotic or necrotic cell death. Both of these outcomes were prevented by the antioxidants CoQ(10) and vitamin E, suggesting that the abnormally high levels of ROS were the triggers of cell death. In conclusion, the principal metabolic defects, i.e., energy deficiency and ROS burden, resulted from the 9035T>C mutation and could be responsible for the development of clinical symptoms in this family. Furthermore, antioxidant therapy might prove helpful in the management of this disease.


Asunto(s)
Ataxia/genética , ADN Mitocondrial/genética , Enfermedades Mitocondriales/genética , Polimorfismo Genético/genética , Adenosina Trifosfato/metabolismo , Adulto , Análisis de Varianza , Antioxidantes/farmacología , Ataxia/complicaciones , Caspasa 3/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Preescolar , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/genética , Análisis Mutacional de ADN/métodos , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/genética , Salud de la Familia , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Glioblastoma/patología , Humanos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Persona de Mediana Edad , Enfermedades Mitocondriales/complicaciones , Especies Reactivas de Oxígeno/metabolismo , Vitaminas/farmacología
14.
Neurobiol Dis ; 33(3): 405-14, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19118631

RESUMEN

In recent years, GDNF has emerged as a protective and restorative agent in several models of neurodegeneration; however, the exact molecular mechanisms responsible for these effects are not yet fully understood. Here we examined the effects of astrocytes secreting GDNF on neurons subjected to 6OHDA toxicity using in vitro neuron-astroglia co-cultures. Astrocytes were transduced with lentiviral vectors carrying the GDNF gene under the control of either human glial fibrillary acidic protein or cytomegalovirus promoters. The overexpression of GDNF, regardless of the promoter employed, had no obvious adverse effects on astroglia and the engineered cells stably produced and secreted GDNF for extended periods of time (> or =3 weeks). These astrocytes very effectively protected neurons against 6OHDA, in both mouse and human co-culture systems. The neuroprotective effects were mediated not only by GDNF, but also by the antioxidant GSH since its depletion reduced the level of GDNF protection. Furthermore, neurons and astrocytes expressed different components of GDNF signaling complex, suggesting that they might utilize separate pathways to mediate autocrine and paracrine effects of GDNF.


Asunto(s)
Astrocitos/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Glutatión/metabolismo , Neuronas/fisiología , Oxidopamina/toxicidad , Análisis de Varianza , Animales , Antioxidantes/metabolismo , Western Blotting , Butionina Sulfoximina/administración & dosificación , Supervivencia Celular/fisiología , Técnicas de Cocultivo , Expresión Génica , Glutatión/antagonistas & inhibidores , Humanos , Inmunohistoquímica , Ratones , Neuronas/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Transducción de Señal/fisiología , Transfección
15.
J Neurosci Res ; 86(8): 1680-93, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18293417

RESUMEN

SOX2 is a key neurodevelopmental gene involved in maintaining the pluripotency of stem cells and proliferation of neural progenitors and astroglia. Two evolutionally conserved enhancers, SRR1 and SRR2, are involved in controlling SOX2 expression during neurodevelopment; however, the molecular mechanisms regulating their activity are not known. We have examined DNA methylation and histone H3 acetylation at both enhancers in NT2-D1 progenitors, neurons and astrocytes, to establish the role of epigenetic mechanisms in cell-type-specific SOX2 expression. This study showed that 1) unmethylated DNA and acetylated histones at both enhancers correlated with a high level of SOX2 expression in proliferating neural progenitors and 2) reversible modifications of the SRR1 element were observed during gene reexpression in astrocytes, whereas permanent epigenetic marks on the SRR2 enhancer were seen in neurons where the gene was silenced. Taken together, these results are clear illustrations of cell-type-specific epigenomes and suggest mechanisms by which they may be created and maintained.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Diferenciación Celular/fisiología , Proteínas de Unión al ADN/biosíntesis , Elementos de Facilitación Genéticos/fisiología , Epigénesis Genética/fisiología , Proteínas HMGB/biosíntesis , Glicoproteínas de Membrana/fisiología , Neuronas/citología , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores de Péptidos/fisiología , Factores de Transcripción/biosíntesis , Acetilación , Astrocitos/citología , Astrocitos/metabolismo , Secuencia de Bases , Proteínas de Unión al Calcio/genética , Células Cultivadas , Metilación de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Datos de Secuencia Molecular , Neuronas/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Péptidos/genética , Factores de Transcripción SOXB1 , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
BMC Genomics ; 7: 286, 2006 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-17090317

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a complex disorder that involves multiple biological processes. Many genes implicated in these processes may be present in low abundance in the human brain. DNA microarray analysis identifies changed genes that are expressed at high or moderate levels. Complementary to this approach, we described here a novel technology designed specifically to isolate rare and novel genes previously undetectable by other methods. We have used this method to identify differentially expressed genes in brains affected by AD. Our method, termed Subtractive Transcription-based Amplification of mRNA (STAR), is a combination of subtractive RNA/DNA hybridization and RNA amplification, which allows the removal of non-differentially expressed transcripts and the linear amplification of the differentially expressed genes. RESULTS: Using the STAR technology we have identified over 800 differentially expressed sequences in AD brains, both up- and down- regulated, compared to age-matched controls. Over 55% of the sequences represent genes of unknown function and roughly half of them were novel and rare discoveries in the human brain. The expression changes of nearly 80 unique genes were further confirmed by qRT-PCR and the association of additional genes with AD and/or neurodegeneration was established using an in-house literature mining tool (LitMiner). CONCLUSION: The STAR process significantly amplifies unique and rare sequences relative to abundant housekeeping genes and, as a consequence, identifies genes not previously linked to AD. This method also offers new opportunities to study the subtle changes in gene expression that potentially contribute to the development and/or progression of AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Encéfalo/metabolismo , Perfilación de la Expresión Génica/métodos , Genes , Proteínas del Tejido Nervioso/genética , Técnicas de Amplificación de Ácido Nucleico/métodos , ARN Mensajero/genética , Transcripción Genética , Regiones no Traducidas 3'/genética , Enfermedad de Alzheimer/metabolismo , ADN Complementario/genética , Regulación de la Expresión Génica , Biblioteca de Genes , Humanos , Proteínas del Tejido Nervioso/biosíntesis , Hibridación de Ácido Nucleico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Técnica de Sustracción
17.
Dev Biol ; 295(1): 52-66, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16631155

RESUMEN

The mammalian neocortex is established from neural stem and progenitor cells that utilize specific transcriptional and environmental factors to create functional neurons and astrocytes. Here, we examined the mechanism of Sox2 action during neocortical neurogenesis and gliogenesis. We established a robust Sox2 expression in neural stem and progenitor cells within the ventricular zone, which persisted until the cells exited the cell cycle. Overexpression of constitutively active Sox2 in neural progenitors resulted in upregulation of Notch1, recombination signal-sequence binding protein-J (RBP-J) and hairy enhancer of split 5 (Hes5) transcripts and the Sox2 high mobility group (HMG) domain seemed sufficient to confer these effects. While Sox2 overexpression permitted the differentiation of progenitors into astroglia, it inhibited neurogenesis, unless the Notch pathway was blocked. Moreover, neuronal precursors engaged a serine protease(s) to eliminate the overexpressed Sox2 protein and relieve the repression of neurogenesis. Glial precursors and differentiated astrocytes, on the other hand, maintained Sox2 expression until they reached a quiescent state. Sox2 expression was re-activated by signals that triggered astrocytic proliferation (i.e., injury, mitogenic and gliogenic factors). Taken together, Sox2 appears to act upstream of the Notch signaling pathway to maintain the cell proliferative potential and to ensure the generation of sufficient cell numbers and phenotypes in the developing neocortex.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Neocórtex/embriología , Neocórtex/crecimiento & desarrollo , Transactivadores/metabolismo , Animales , Astrocitos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Regulación hacia Abajo , Femenino , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Ratones , Neocórtex/citología , Neocórtex/metabolismo , Neuroglía/metabolismo , Receptor Notch1/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción SOXB1 , Serina Endopeptidasas/metabolismo , Transducción de Señal , Células Madre/metabolismo , Transactivadores/genética , Transactivadores/inmunología
18.
J Neurosci Res ; 72(6): 691-703, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12774309

RESUMEN

Although glutamate excitotoxicity has long been implicated in neuronal cell death associated with a variety of neurological disorders, the molecular mechanisms underlying this process are not yet fully understood. In part, this is due to the lack of relevant experimental cell systems recapitulating the in vivo neuronal environment, mainly neuronal-glial interactions. To explore these mechanisms, we have analyzed the cytotoxic effects of glutamate on mixed cultures of NT2/N neurons and NT2/A astrocytes derived from human NT2/D1 cells. In these cultures, the neurons were resistant to glutamate alone (up to 2 mM for 24-48 hr), but they responded to a simultaneous exposure to 0.5 mM glutamate and 6 hr of hypoxia. Neuronal cell death occurred during subsequent periods of reoxygenation (>30% within 24 hr). This was associated with a marked decrease of intracellular ATP, a significant increase in reactive oxygen species (ROS) and downregulation of glutamate uptake by astrocytes. Thus, under energy failure and high levels of ROS production, only the neurons from these mixed cultures succumbed to glutamate neurotoxicity; the astrocytic cells remained unaffected by the treatment. Taken together, our data suggested that glutamate excitotoxicity might be due to the energy failure and oxidative stress affecting the properties of the NMDA glutamate receptors and causing impairment of glutamate transporters. Cells pretreated for 72 hr with 10 microg/ml of coenzyme Q(10) (functions both as a ROS scavenger and co-factor of mitochondrial electron transport), were protected, suggesting a useful role for coenzyme Q(10) in treatments of neurological diseases associated with glutamate excitotoxicity. A model of the complex interactions between neurons and astrocytes in regulating glutamate metabolism is presented.


Asunto(s)
Astrocitos/efectos de los fármacos , Citoprotección/efectos de los fármacos , Ácido Glutámico/toxicidad , Neuronas/efectos de los fármacos , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Astrocitos/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Coenzimas , Citoprotección/fisiología , Humanos , Neuronas/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
19.
J Biol Chem ; 278(29): 26401-9, 2003 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-12738784

RESUMEN

We have identified a functional cAMP-response element (CRE) in the human brain-derived neurotrophic factor (BDNF) gene promoter III and established that it participated in the modulation of BDNF expression in NT2/N neurons via downstream signaling from the D1 class of dopamine (DA) receptors. The up-regulation of BDNF expression, in turn, produced neuroprotective signals through receptor tyrosine kinase B (TrkB) and promoted cell survival under the conditions of oxygen and glucose deprivation. To our knowledge this is the first evidence showing the presence of a functional CRE in the human BDNF gene and the role of DA signaling in establishing transcriptional competence of CRE in post-mitotic NT2/N neurons. This ability of DA to regulate the expression of the BDNF survival factor has a profound significance for the nigrostriatal pathway, because it indicates the existence of a feedback loop between the neutrophin, which promotes both the maturation and survival of dopaminergic neurons, and the neurotransmitter, which the mature neurons ultimately produce and release.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Dopamina/metabolismo , Regiones Promotoras Genéticas , Animales , Secuencia de Bases , Bucladesina/farmacología , Diferenciación Celular , Línea Celular , Supervivencia Celular , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , ADN/genética , ADN/metabolismo , Humanos , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Receptor trkB/metabolismo , Receptores de Dopamina D1/metabolismo , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Transducción de Señal , Activación Transcripcional/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...