Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Intervalo de año de publicación
3.
Mol Neurobiol ; 56(12): 8277-8295, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31214863

RESUMEN

MeCP2 is an X-linked gene; its mutation causes Rett Syndrome (RTT), a severe neurodevelopmental disability that affects mainly girls. Acting as a transcription factor, the MeCP2 protein is able to regulate several hormone-related genes, such as the thyroid hormones (TH), which are known to play an important role in the development of the central nervous system (CNS). Although only a few studies have associated RTT and TH, TH deficit can lead to neurological deregulation by triggering functional deficiencies during adulthood. Here, we used human-induced pluripotent stem cell (iPSC) to generate MeCP2-knockout neuronal progenitor cells and adult neurons. Using this cellular model, we then investigated the expression of genes associated with TH homeostasis, such as the TH transporters (LAT1, LAT2, MCT8, MCT10, and OATP4A1) and deiodinases (DIO1, 2, and 3). Then, we treated the neural cells with THs and analyzed the expression of several genes related to neurodevelopment and functional maintenance. Our results showed that several TH-related genes, such as deiodinases, are altered in RTT samples when compared to WT cells. Moreover, the treatment of the neural cells with THs increased the amount of MAP2 and synapsin-1 expression in RTT cells. Our work provided evidences that TH homeostasis is compromised in RTT-derived neural cells, which could be an important factor to contribute to the imbalance in the neurodevelopmental phenotype presented in this syndrome and can lead us to better understand other neurodevelopmental diseases.


Asunto(s)
Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Yoduro Peroxidasa/genética , Proteínas de Transporte de Membrana/genética , Proteína 2 de Unión a Metil-CpG/deficiencia , Neuronas/metabolismo , Hormonas Tiroideas/metabolismo , Humanos , Yoduro Peroxidasa/metabolismo , Cariotipificación , Masculino , Proteínas de Transporte de Membrana/metabolismo , Redes y Vías Metabólicas , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Síndrome de Rett/enzimología , Síndrome de Rett/genética
5.
Cell Tissue Res ; 371(1): 153-160, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28918504

RESUMEN

Autism spectrum disorders (ASD) represent a variety of disorders characterized as complex lifelong neurodevelopment disabilities, which may affect the ability of communication and socialization, including typical comportments like repetitive and stereotyped behavior. Other comorbidities are usually present, such as echolalia, hypotonia, intellectual disability and difficulties in processing figured speech. Furthermore, some ASD individuals may present certain abilities, such as eidetic memory, outstanding musical or painting talents and special mathematical skills, among others. Considering the variability of the clinical symptoms, one autistic individual can be severely affected in communication while others can speak perfectly, sometimes having a vocabulary above average in early childhood. The same variability can be seen in other clinical symptoms, thus the "spectrum" can vary from severe to mild. Induced pluripotent stem cell technology has been used to model several neurological diseases, including syndromic and non-syndromic autism. We discuss how modeling the central nervous system cells in a dish may help to reach a better understanding of ASD pathology and variability, as well as personalize their treatment.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Células Madre Pluripotentes Inducidas/fisiología , Modelos Neurológicos , Neuronas/fisiología , Animales , Técnicas de Cultivo , Humanos , Ratones
6.
Biol Psychiatry ; 83(7): 569-578, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29129319

RESUMEN

BACKGROUND: Autism spectrum disorder (ASD) is a neurodevelopmental disorder with unclear etiology and imprecise genetic causes. The main goal of this work was to investigate neuronal connectivity and the interplay between neurons and astrocytes from individuals with nonsyndromic ASD using induced pluripotent stem cells. METHODS: Induced pluripotent stem cells were derived from a clinically well-characterized cohort of three individuals with nonsyndromic ASD sharing common behaviors and three control subjects, two clones each. We generated mixed neural cultures analyzing synaptogenesis and neuronal activity using a multielectrode array platform. Furthermore, using an enriched astrocyte population, we investigated their role in neuronal maintenance. RESULTS: ASD-derived neurons had a significant decrease in synaptic gene expression and protein levels, glutamate neurotransmitter release, and, consequently, reduced spontaneous firing rate. Based on co-culture experiments, we observed that ASD-derived astrocytes interfered with proper neuronal development. In contrast, control-derived astrocytes rescued the morphological neuronal phenotype and synaptogenesis defects from ASD neuronal co-cultures. Furthermore, after identifying interleukin-6 secretion from astrocytes in individuals with ASD as a possible culprit for neural defects, we were able to increase synaptogenesis by blocking interleukin-6 levels. CONCLUSIONS: Our findings reveal the contribution of astrocytes to neuronal phenotype and confirm previous studies linking interleukin-6 and autism, suggesting potential novel therapeutic pathways for a subtype of individuals with ASD. This is the first report demonstrating that glial dysfunctions could contribute to nonsyndromic autism pathophysiology using induced pluripotent stem cells modeling disease technology.


Asunto(s)
Astrocitos/fisiología , Trastorno del Espectro Autista , Expresión Génica , Células Madre Pluripotentes Inducidas/fisiología , Interleucina-6/metabolismo , Neuronas/fisiología , Sinapsis/fisiología , Astrocitos/metabolismo , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/fisiopatología , Técnicas de Cultivo de Célula , Niño , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Modelos Neurológicos , Neuronas/metabolismo , Sinapsis/metabolismo
7.
Adv Exp Med Biol ; 976: 137-148, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28508319

RESUMEN

Transient receptor potential canonical (TRPC) channels mediate the influx of different types of cations through the cell membrane and are involved in many functions of the organism. Evidences of involvement of TRPC channels in neuronal development suggest that this family of proteins might play a role in certain neurological disorders. As reported, knockout mice for different TRPC channels show alterations in neuronal morphological and functional parameters, with behavioral abnormalities, such as in exploratory and social behaviors. Although mutations in TRPC channels could be related to mental/neurological disorders, there are only a few cases reported in literature, indicating that this correlation should be further explored. Nonetheless, other functional evidences support the implication of these channels in neurological diseases. In this chapter, we summarize the main findings relating TRPC channels to neurological disorders, such as autism spectrum disorders, bipolar disorder, and intellectual disability among others.


Asunto(s)
Encéfalo/metabolismo , Calcio/metabolismo , Trastornos Mentales/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Trastornos Mentales/genética , Ratones , Ratones Noqueados , Canales Catiónicos TRPC/genética
8.
Mutat Res ; 784-785: 25-33, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26811994

RESUMEN

Oxidative DNA damage is considered to be a major cause of neurodegeneration and internal tumors observed in syndromes that result from nucleotide excision repair (NER) deficiencies, such as Xeroderma Pigmentosum (XP) and Cockayne Syndrome (CS). Recent evidence has shown that NER aids in removing oxidized DNA damage and may interact with base excision repair (BER) enzymes. Here, we investigated APE1 and OGG1 expression, localization and activity after oxidative stress in XPC-deficient cells. The endogenous APE1 and OGG1 mRNA levels were lower in XPC-deficient fibroblasts. However, XPC-deficient cells did not show hypersensitivity to oxidative stress compared with NER-proficient cells. To confirm the impact of an XPC deficiency in regulating APE1 and OGG1 expression and activity, we established an XPC-complemented cell line. Although the XPC complementation was only partial and transient, the transfected cells exhibited greater OGG1 expression and activity compared with XPC-deficient cells. However, the APE1 expression and activity did not significantly change. Furthermore, we observed a physical interaction between the XPC and APE1 proteins. Together, the results indicate that the responses of XPC-deficient cells under oxidative stress may not only be associated with NER deficiency per se but may also include new XPC functions in regulating BER proteins.


Asunto(s)
ADN Glicosilasas/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Cultivadas , ADN Glicosilasas/genética , Reparación del ADN/fisiología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Peróxido de Hidrógeno/farmacología , Inmunoprecipitación , Oxidantes/farmacología , Estrés Oxidativo , ARN Mensajero/metabolismo , Xerodermia Pigmentosa/genética , Xerodermia Pigmentosa/patología
9.
Biol Psychiatry ; 79(8): 642-9, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25861701

RESUMEN

The lack of live human brain cells for research has slowed progress toward understanding the mechanisms underlying autism spectrum disorders. A human model using reprogrammed patient somatic cells offers an attractive alternative, as it captures a patient's genome in relevant cell types. Despite the current limitations, the disease-in-a-dish approach allows for progressive time course analyses of target cells, offering a unique opportunity to investigate the cellular and molecular alterations before symptomatic onset. Understanding the current drawbacks of this model is essential for the correct data interpretation and extrapolation of conclusions applicable to the human brain. Innovative strategies for collecting biological material and clinical information from large patient cohorts are important for increasing the statistical power that will allow for the extraction of information from the noise resulting from the variability introduced by reprogramming and differentiation methods. Working with large patient cohorts is also important for understanding how brain cells derived from diverse human genetic backgrounds respond to specific drugs, creating the possibility of personalized medicine for autism spectrum disorders.


Asunto(s)
Trastorno Autístico/fisiopatología , Encéfalo/fisiopatología , Animales , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Modelos Biológicos
10.
Neurotherapeutics ; 12(3): 534-45, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25851569

RESUMEN

Autism spectrum disorders (ASDs) are a heterogeneous group of neurodevelopmental disorders sharing a core set of symptoms, including impaired social interaction, language deficits, and repetitive behaviors. While ASDs are highly heritable and demonstrate a clear genetic component, the cellular and molecular mechanisms driving ASD etiology remain undefined. The unavailability of live patient-specific neurons has contributed to the difficulty in studying ASD pathophysiology. The recent advent of induced pluripotent stem cells (iPSCs) has provided the ability to generate patient-specific human neurons from somatic cells. The iPSC field has quickly grown, as researchers have demonstrated the utility of this technology to model several diseases, especially neurologic disorders. Here, we review the current literature around using iPSCs to model ASDs, and discuss the notable findings, and the promise and limitations of this technology. The recent report of a nonsyndromic ASD iPSC model and several previous ASD models demonstrating similar results points to the ability of iPSC to reveal potential novel biomarkers and therapeutics.


Asunto(s)
Trastorno del Espectro Autista/tratamiento farmacológico , Trastorno del Espectro Autista/fisiopatología , Células Madre Pluripotentes Inducidas/fisiología , Células-Madre Neurales/fisiología , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...