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
11.
Neurobiol Learn Mem ; 101: 94-102, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23357282

RESUMEN

It has been described that exercise can modulate both inflammatory response and epigenetic modifications, although the effect of exercise on these parameters during the normal brain aging process yet remains poorly understood. Here, we investigated the effect of aging and treadmill exercise on inflammatory and epigenetic parameters specifically pro and anti-inflammatory cytokines levels, activation of NF-kB and histone H4 acetylation levels in hippocampus from Wistar rats. Additionally, we evaluated aversive memory through inhibitory avoidance task. Rats of 3 and 20 months of age were assigned to non-exercised (sedentary) and exercised (running daily for 20 min for 2 weeks) groups. The effect of daily forced exercise in the treadmill was assessed. The levels of inflammatory and epigenetic parameters were determined 1h, 18 h, 3 days or 7 days after the last training session of exercise. It was observed an age-related decline on aversive memory, as well as aged rats showed increased hippocampal levels of inflammatory markers, such as TNFα, IL1-ß and NF-kB and decreased IL-4 levels, an anti-inflammatory cytokine. Moreover, lower levels of global histone H4 acetylation were also observed in hippocampi from aged rats. Interestingly, there was a significant correlation between the biochemical markers and the inhibitory avoidance test performance. The forced exercise protocol ameliorated aging-related memory decline, decreased pro-inflammatory markers and increased histone H4 acetylation levels in hippocampi 20-months-old rats, while increased acutely IL-4 levels in hippocampi from young adult rats. Together, these results suggest that an imbalance of inflammatory markers might be involved to the aging-related aversive memory impairment. Additionally, our exercise protocol may reverse aging-related memory decline through improving cytokine profile.


Asunto(s)
Envejecimiento/metabolismo , Hipocampo/metabolismo , Mediadores de Inflamación/metabolismo , Memoria/fisiología , Condicionamiento Físico Animal/fisiología , Acetilación , Envejecimiento/fisiología , Animales , Reacción de Prevención/fisiología , Epigénesis Genética , Histonas/metabolismo , Interleucina-1beta/metabolismo , Interleucina-4/metabolismo , Masculino , FN-kappa B/metabolismo , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo
12.
Exp Gerontol ; 48(2): 136-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23201423

RESUMEN

Regular exercise improves learning and memory, including during aging process. Interestingly, the imbalance of epigenetic mechanisms has been linked to age-related cognitive deficits. However, studies about epigenetic alterations after exercise during the aging process are rare. In this preliminary study we investigated the effect of aging and exercise on DNA methyltransferases (DNMT1 and DNMT3b) and H3-K9 methylation levels in hippocampus from 3 and 20-months aged Wistar rats. The animals were submitted to two exercise protocols: single session or chronic treadmill protocol. DNMT1 and H3-K9 methylation levels were decreased in hippocampus from aged rats. The single exercise session decreased both DNMT3b and DNMT1 levels in young adult rats, without any effect in the aged group. Both exercise protocols reduced H3-K9 methylation levels in young adult rats, while the single session reversed the changes on H3-K9 methylation levels induced by aging. Together, these results suggest that an imbalance on DNMTs and H3-K9 methylation levels might be linked to the brain aging process and that the outcome to exercise seems to vary through lifespan.


Asunto(s)
Envejecimiento/genética , Metilación de ADN , Epigénesis Genética , Hipocampo/metabolismo , Esfuerzo Físico/genética , Factores de Edad , Animales , Biomarcadores/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Histonas/metabolismo , Lisina , Masculino , Ratas , Ratas Wistar , ADN Metiltransferasa 3B
13.
Proteomics ; 12(17): 2607-17, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22778083

RESUMEN

Human mesenchymal stem cells (hMSCs) are adult multipotent cells that have high therapeutic potential due to their immunological properties. They can be isolated from several different tissues with bone marrow (BM) being the most common source. Because the isolation procedure is invasive, other tissues such as human umbilical cord vein (UCV) have been considered. However, their interchangeability remains unclear. In the present study, total protein extracts of BM-hMSCs and UCV-hMSCs were quantitatively compared using gel-LC-MS/MS. Previous SAGE analysis of the same cells was re-annotated to enable comparison and combination of these two data sets. We observed a more than 63% correlation between proteomic and transcriptomic data. In silico analysis of highly expressed genes in cells of both origins suggests that they can be modulated by microRNA, which can change protein abundance. Our results showed that MSCs from both tissues shared high similarity in metabolic and functional processes relevant to their therapeutic potential, especially in the immune system process, response to stimuli, and processes related to the delivery of the hMSCs to a given tissue, such as migration and adhesion. Hence, our results support the idea that the more accessible UCV could be a potentially less invasive source of MSCs.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteoma/análisis , Transcriptoma , Venas Umbilicales/citología , Adulto , Células Cultivadas , Cromatografía Liquida/métodos , Humanos , Proteoma/metabolismo , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos
14.
Hum Mol Genet ; 21(17): 3825-34, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22661500

RESUMEN

Cockayne syndrome (CS) is a human premature aging disorder associated with neurological and developmental abnormalities, caused by mutations mainly in the CS group B gene (ERCC6). At the molecular level, CS is characterized by a deficiency in the transcription-couple DNA repair pathway. To understand the role of this molecular pathway in a pluripotent cell and the impact of CSB mutation during human cellular development, we generated induced pluripotent stem cells (iPSCs) from CSB skin fibroblasts (CSB-iPSC). Here, we showed that the lack of functional CSB does not represent a barrier to genetic reprogramming. However, iPSCs derived from CSB patient's fibroblasts exhibited elevated cell death rate and higher reactive oxygen species (ROS) production. Moreover, these cellular phenotypes were accompanied by an up-regulation of TXNIP and TP53 transcriptional expression. Our findings suggest that CSB modulates cell viability in pluripotent stem cells, regulating the expression of TP53 and TXNIP and ROS production.


Asunto(s)
Envejecimiento Prematuro/patología , Síndrome de Cockayne/patología , Células Madre Pluripotentes Inducidas/patología , Estrés Oxidativo , Muerte Celular/genética , Hipoxia de la Célula/genética , Supervivencia Celular/genética , Células Clonales , Síndrome de Cockayne/genética , Daño del ADN/genética , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo
15.
Curr Opin Neurobiol ; 22(5): 785-90, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22717528

RESUMEN

The cellular and molecular mechanisms of neurodevelopmental conditions such as autism spectrum disorders have been studied intensively for decades. The unavailability of live patient neurons for research, however, has represented a major obstacle in the elucidation of the disease etiologies. Recently, the development of induced pluripotent stem cell (iPSC) technology allows for the generation of human neurons from somatic cells of patients. We review ongoing studies using iPSCs as an approach to model neurodevelopmental disorders, the promise and caveats of this technique and its potential for drug screening. The reproducible findings of relevant phenotypes in Rett syndrome iPSC-derived neurons suggest that iPSC technology offers a novel and unique opportunity for the understanding of and the development of therapeutics for other autism spectrum disorders.


Asunto(s)
Diferenciación Celular/fisiología , Enfermedades del Sistema Nervioso Central , Discapacidades del Desarrollo , Células Madre Pluripotentes Inducidas/fisiología , Neuronas/fisiología , Enfermedades del Sistema Nervioso Central/complicaciones , Enfermedades del Sistema Nervioso Central/patología , Enfermedades del Sistema Nervioso Central/terapia , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/patología , Discapacidades del Desarrollo/terapia , Humanos
16.
Stem Cells Dev ; 21(4): 513-20, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22066548

RESUMEN

Autophagy is a lysosome-dependent degradation pathway that allows cells to recycle damaged or superfluous cytoplasmic content, such as proteins, organelles, and lipids. As a consequence of autophagy, the cells generate metabolic precursors for macromolecular biosynthesis or ATP generation. Deficiencies in this pathway were associated to several pathological conditions, such as neurodegenerative and cardiac diseases, cancer, and aging. The aim of this review is to summarize recent discoveries showing that autophagy also plays a critical role in stem cell maintenance and in a variety of cell differentiation processes. We also discuss a possible role for autophagy during cellular reprogramming and induced pluripotent stem (iPS) cell generation by taking advantage of ATP generation for chromatin remodeling enzyme activity and mitophagy. Finally, the significance of autophagy modulation is discussed in terms of augmenting efficiency of iPS cell generation and differentiation processes.


Asunto(s)
Autofagia/fisiología , Diferenciación Celular/fisiología , Células Madre Pluripotentes/metabolismo , Adenosina Trifosfato/metabolismo , Envejecimiento/metabolismo , Animales , Ensamble y Desensamble de Cromatina/fisiología , Cardiopatías/metabolismo , Humanos , Lisosomas/metabolismo , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo
17.
Estud. av ; 24(70): 71-79, 2010. ilus
Artículo en Portugués | LILACS | ID: lil-566044

RESUMEN

Grande parte do conhecimento atual dos fenótipos celulares relacionados a doenças neurológicas foi obtida a partir de estudos de tecidos cerebrais coletados após a morte do indivíduo. Essas amostras geralmente representam os estágios finais da doença e, portanto, não servem como fiel representação de como os sintomas aparecem. Além disso, nessas circunstâncias, a patologia observada pode muito bem ser um efeito secundário do processo patológico ou mesmo da deterioração do tecido em vez de um fenótipo celular autêntico. Da mesma forma, modelos animais nem sempre recapitulam exatamente a patologia das doenças em humanos. Neste artigo, pretendo apresentar uma visão crítica dos recentes avanços obtidos a partir da modelagem de doenças neurológicas humanas, utilizando células-tronco pluripotentes. O foco na reprogramação celular de células somáticas, gerando células-tronco pluripotentes induzidas, justifica-se em razão do grande potencial experimental não só para a modelagem de doenças humanas, mas também como ferramenta biotecnológica para triagem de novas drogas, contribuindo para uma futura medicina personalizada.


Most of our current knowledge about cellular phenotypes related to neurological diseases was gathered from studies performed in brain tissue collected postmortem. These samples often represent the end-stage of the disease process and may not represent a fair picture of how the disease developed over time. Futhermore, under these conditions, the pathology may as well be a secundary effect of the disease process or even due to the poor tissue condition and may not represent an authentic cellular phenotype. Likewise, animal models not always recapitulate the pathology from human disorders. In this article, I will present a critical view on the recent advances obtained from disease modeling using human pluripotent stem cells. The focus on cellular reprogramming as tool to generate patient-specific induced pluripotent stem cells is justified by the great experimental potential, not only for disease modeling, but also as a biotecnological tool for future drug-screening platforms and personalized medicine.


Asunto(s)
Humanos , Investigación Genética , Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Células Madre , Manifestaciones Neurológicas , Neuronas
18.
Epilepsy Behav ; 14 Suppl 1: 81-5, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18845273

RESUMEN

Pluripotency is generally defined by the ability to differentiate into cell types representing all three germ layers: ectoderm, mesoderm, and endoderm. Human pluripotent stem cells hold great promise in regenerative medicine and in cell replacement therapies because of their ability to self-renew and their developmental potential to become all cell types in the body. Moreover, pluripotent cells represent a unique system in which to study the normal development of the human nervous system and the several instances where the process may fail. Here, I propose several strategies for how pluripotent stem cells, both human embryonic stem cells and induced pluripotent stem cells, can potentially be used to gain insights into the biology of temporal lobe epilepsy.


Asunto(s)
Epilepsia/patología , Modelos Neurológicos , Células Madre Pluripotentes/fisiología , Animales , Quimera/fisiología , Epilepsia del Lóbulo Temporal/patología , Humanos , Neuroglía/fisiología
19.
Int J Cardiol ; 113(3): 348-54, 2006 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-16675047

RESUMEN

BACKGROUND: We tested a preemptive combined cell/gene therapy strategy of skeletal myoblasts transfected with Ad(5)RSVVEGF-165 in an ischemia/reperfusion rat model to increase collateral blood flow to nonischemic heart tissue. METHODS: Lewis rats were injected with placebo (Control), 10(6) skeletal myoblasts (SkM), or 10(6) skeletal myoblasts transfected with Ad(5)RSVVEGF-165 (SkM(+)) into the left ventricle 1week before ischemia. Left ventricle end-diastolic pressure, scar area, and capillary density were assessed 4weeks later. RESULTS: Local expression of human vascular endothelial growth factor was accompanied by an increase in capillary density in the SkM(+) group compared with that in the SkM and Control groups (700+/-40 vs. 289+/-18 and 318+/-59capillaries/mm(2), respectively; p<0.05). After 3weeks, the myocardial scar area was reduced in SkM(+) vs. Control (5.3+/-0.4% and 14.8+/-1.6%, p<0.05), while injected cells alone (SkM) did not cause improvement compared with Control (11.8+/-2.1% vs. 14.8+/-1.6%, p>0.05). The decrease in the scar area in SkM(+) was accompanied by an increase in the capillary density compared with that in SkM and Control 30days after cell injection (1005+/-108 vs. 524+/-16 and 528+/-26capillaries/mm(2), respectively; p<0.05). The scar areas were discrete (5.3-14.8%) and left ventricle end-diastolic pressure in all groups were comparable (p>0.05). CONCLUSIONS: The combined cell/gene therapy strategy of genetically modified myoblast cells expressing angiogenic factors injected into the myocardium induced capillary formation and prevented the extension and development of cardiac damage associated with ischemia/reperfusion in rats.


Asunto(s)
Trasplante de Células , Terapia Genética , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Neovascularización Fisiológica , Daño por Reperfusión/terapia , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Capilares , Humanos , Ratas , Ratas Endogámicas Lew
20.
Cancer Gene Ther ; 12(4): 389-96, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15650764

RESUMEN

The nucleotide excision repair (NER) is one of the major human DNA repair pathways. Defects in one of the proteins that act in this system result in three distinct autosomal recessive syndromes: xeroderma pigmentosum (XP), Cockayne syndrome (CS) and trichothiodystrophy (TTD). TFIIH is a nine-protein complex essential for NER activity, initiation of RNA polymerase II transcription and with a possible role in cell cycle regulation. XPD is part of the TFIIH complex and has a helicase function, unwinding the DNA in the 5' --> 3' direction. Mutations in the XPD gene are found in XP, TTD and XP/CS patients, the latter exhibiting both XP and CS symptoms. Correction of DNA repair defects of these cells by transducing the complementing wild-type gene is one potential strategy for helping these patients. Over the last years, adenovirus vectors have been largely used in gene delivering because of their efficient transduction, high titer, and stability. In this work, we present the construction of a recombinant adenovirus carrying the XPD gene, which is coexpressed with the EGFP reporter gene by an IRES sequence, making it easier to follow cell infection. Infection by this recombinant adenovirus grants full correction of SV40-transformed and primary skin fibroblasts obtained from XP-D, TTD and XP/CS patients.


Asunto(s)
Adenoviridae/genética , ADN Helicasas/genética , Reparación del ADN , Proteínas de Unión al ADN/genética , Recombinación Genética , Factores de Transcripción/genética , Western Blotting , Línea Celular , Línea Celular Transformada , Línea Celular Tumoral , Separación Celular , Supervivencia Celular , Células Cultivadas , Síndrome de Cockayne/genética , Síndrome de Cockayne/terapia , ADN/metabolismo , Relación Dosis-Respuesta en la Radiación , Displasia Ectodérmica/genética , Displasia Ectodérmica/terapia , Fibroblastos/metabolismo , Citometría de Flujo , Prueba de Complementación Genética , Humanos , Cinética , Masculino , Microscopía Fluorescente , Modelos Genéticos , Mutación , Fenotipo , ARN Polimerasa II/metabolismo , Sensibilidad y Especificidad , Piel/metabolismo , Factor de Transcripción TFIIH , Factores de Transcripción TFII/genética , Rayos Ultravioleta , Xerodermia Pigmentosa/genética , Xerodermia Pigmentosa/terapia , Proteína de la Xerodermia Pigmentosa del Grupo D
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...