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










Base de datos
Intervalo de año de publicación
1.
Alzheimers Dement ; 19(12): 5970-5987, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37768001

RESUMEN

INTRODUCTION: Experimental models are essential tools in neurodegenerative disease research. However, the translation of insights and drugs discovered in model systems has proven immensely challenging, marred by high failure rates in human clinical trials. METHODS: Here we review the application of artificial intelligence (AI) and machine learning (ML) in experimental medicine for dementia research. RESULTS: Considering the specific challenges of reproducibility and translation between other species or model systems and human biology in preclinical dementia research, we highlight best practices and resources that can be leveraged to quantify and evaluate translatability. We then evaluate how AI and ML approaches could be applied to enhance both cross-model reproducibility and translation to human biology, while sustaining biological interpretability. DISCUSSION: AI and ML approaches in experimental medicine remain in their infancy. However, they have great potential to strengthen preclinical research and translation if based upon adequate, robust, and reproducible experimental data. HIGHLIGHTS: There are increasing applications of AI in experimental medicine. We identified issues in reproducibility, cross-species translation, and data curation in the field. Our review highlights data resources and AI approaches as solutions. Multi-omics analysis with AI offers exciting future possibilities in drug discovery.


Asunto(s)
Demencia , Enfermedades Neurodegenerativas , Humanos , Inteligencia Artificial , Reproducibilidad de los Resultados , Aprendizaje Automático
2.
Brain Commun ; 5(2): fcad093, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37033334

RESUMEN

Poor maternal nutrition during pregnancy is known to impair fetal development. Moreover, the preimplantation period is vulnerable to adverse programming of disease. Here, we investigated the effect of a mouse maternal high-fat diet in healthy non-obese dams during preimplantation or throughout pregnancy and lactation on metabolism-related parameters and hippocampal neurogenesis in adult offspring. Female mice were fed from conception either a normal fat diet (normal fat diet group) or high-fat diet throughout gestation and lactation (high-fat diet group), or high-fat diet only during preimplantation (embryonic high-fat diet group, high-fat diet up to E3.5, normal fat diet thereafter). Maternal high-fat diet caused changes in the offspring, including increased systolic blood pressure, diurnal activity, respiratory quotient, and energy expenditure in high-fat diet females, and increased systolic blood pressure and respiratory quotient but decreased energy expenditure in high-fat diet males. High-fat diet males had a higher density of newborn neurons and a lower density of mature neurons in the dentate gyrus, indicating that exposure to a maternal high-fat diet may regulate adult neurogenesis. A maternal high-fat diet also increased the density of astrocytes and microglia in the hippocampus of high-fat diet males and females. Generally, a graded response (normal fat diet < embryonic high-fat < high-fat diet) was observed, with only 3 days of high-fat diet exposure altering offspring energy metabolism and hippocampal cell density. Thus, early maternal exposure to a fatty diet, well before neural differentiation begins and independently of maternal obesity, is sufficient to perturb offspring energy metabolism and brain physiology with lifetime consequences.

3.
Stem Cell Rev Rep ; 19(3): 767-783, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36517693

RESUMEN

Peri-conceptional environment can induce permanent changes in embryo phenotype which alter development and associate with later disease susceptibility. Thus, mouse maternal low protein diet (LPD) fed exclusively during preimplantation is sufficient to lead to cardiovascular, metabolic and neurological dysfunction in adult offspring. Embryonic stem cell (ESC) lines were generated from LPD and control NPD C57BL/6 blastocysts and characterised by transcriptomics, metabolomics, bioinformatics and molecular/cellular studies to assess early potential mechanisms in dietary environmental programming. Previously, we showed these lines retain cellular and epigenetic characteristics of LPD and NPD embryos after several passages. Here, three main changes were identified in LPD ESC lines. First, their derivation capacity was reduced but pluripotency marker expression was similar to controls. Second, LPD lines had impaired Mitogen-activated protein kinase (MAPK) pathway with altered gene expression of several regulators (e.g., Maff, Rassf1, JunD), reduced ERK1/2 signalling capacity and poorer cell survival characteristics which may contribute to reduced derivation. Third, LPD lines had impaired glucose metabolism comprising reduced upstream enzyme expression (e.g., Gpi, Mpi) and accumulation of metabolites (e.g., glucose-6-P, fructose-6-P) above the phosphofructokinase (PFK) gateway with PFK enzyme activity reduced. ESC lines may therefore permit investigation of peri-conceptional programming mechanisms with reduced need for animal experimentation.


Asunto(s)
Desnutrición , Células Madre Embrionarias de Ratones , Animales , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Dieta con Restricción de Proteínas
4.
Biochim Biophys Acta Gene Regul Mech ; 1865(5): 194848, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35905858

RESUMEN

Histone modifying enzymes are involved in the posttranslational modification of histones and the epigenetic control of gene expression. They play a critical role in normal development, and there is increasing evidence of their role in developmental disorders (DDs). DDs are a group of chronic, severe conditions that impact the physical, intellectual, language and/or behavioral development of an individual. There are very few treatment options available for DDs such that these are conditions with significant unmet clinical need. Recessive variants in the gene encoding histone modifying enzyme KDM5B are associated with a DD characterized by developmental delay, facial dysmorphism and camptodactyly. KDM5B is responsible for the demethylation of lysine 4 on the amino tail of histone 3 and plays a vital role in normal development and regulating cell differentiation. This review explores the literature on KDM5B and what is currently known about its roles in development and developmental disorders.


Asunto(s)
Histonas , Histona Demetilasas con Dominio de Jumonji , Niño , Discapacidades del Desarrollo/genética , Histonas/genética , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo
5.
Genes (Basel) ; 12(10)2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34680959

RESUMEN

Non-communicable diseases (NCDs) sauch as diabetes, obesity and cardiovascular diseases are rising rapidly in all countries world-wide. Environmental maternal factors (e.g., diet, oxidative stress, drugs and many others), maternal illnesses and other stressors can predispose the newborn to develop diseases during different stages of life. The connection between environmental factors and NCDs was formulated by David Barker and colleagues as the Developmental Origins of Health and Disease (DOHaD) hypothesis. In this review, we describe the DOHaD concept and the effects of several environmental stressors on the health of the progeny, providing both animal and human evidence. We focus on cardiovascular diseases which represent the leading cause of death worldwide. The purpose of this review is to discuss how in vitro studies with pluripotent stem cells (PSCs), such as embryonic and induced pluripotent stem cells (ESC, iPSC), can underpin the research on non-genetic heart conditions. The PSCs could provide a tool to recapitulate aspects of embryonic development "in a dish", studying the effects of environmental exposure during cardiomyocyte (CM) differentiation and maturation, establishing a link to molecular mechanism and epigenetics.


Asunto(s)
Enfermedades Cardiovasculares/genética , Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Estrés Fisiológico , Animales , Diferenciación Celular , Células Madre Embrionarias/citología , Epigénesis Genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología
6.
J Neurosci Res ; 99(5): 1276-1307, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33583054

RESUMEN

Neurological disorders are the leading cause of disability and the second largest cause of death worldwide. Despite significant research efforts, neurology remains one of the most failure-prone areas of drug development. The complexity of the human brain, boundaries to examining the brain directly in vivo, and the significant evolutionary gap between animal models and humans, all serve to hamper translational success. Recent advances in microfluidic in vitro models have provided new opportunities to study human cells with enhanced physiological relevance. The ability to precisely micro-engineer cell-scale architecture, tailoring form and function, has allowed for detailed dissection of cell biology using microphysiological systems (MPS) of varying complexities from single cell systems to "Organ-on-chip" models. Simplified neuronal networks have allowed for unique insights into neuronal transport and neurogenesis, while more complex 3D heterotypic cellular models such as neurovascular unit mimetics and "Organ-on-chip" systems have enabled new understanding of metabolic coupling and blood-brain barrier transport. These systems are now being developed beyond MPS toward disease specific micro-pathophysiological systems, moving from "Organ-on-chip" to "Disease-on-chip." This review gives an outline of current state of the art in microfluidic technologies for neurological disease research, discussing the challenges and limitations while highlighting the benefits and potential of integrating technologies. We provide examples of where such toolsets have enabled novel insights and how these technologies may empower future investigation into neurological diseases.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Microfluídica/tendencias , Enfermedades del Sistema Nervioso/metabolismo , Animales , Transporte Biológico/fisiología , Epigénesis Genética/fisiología , Humanos , Técnicas In Vitro/métodos , Técnicas In Vitro/tendencias , Microfluídica/métodos , Enfermedades del Sistema Nervioso/genética , Organoides/metabolismo
7.
Genes (Basel) ; 13(1)2021 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-35052371

RESUMEN

The societal burden of non-communicable disease is closely linked with environmental exposures and lifestyle behaviours, including the adherence to a poor maternal diet from the earliest preimplantation period of the life course onwards. Epigenetic variations caused by a compromised maternal nutritional status can affect embryonic development. This review summarises the main epigenetic modifications in mammals, especially DNA methylation, histone modifications, and ncRNA. These epigenetic changes can compromise the health of the offspring later in life. We discuss different types of nutritional stressors in human and animal models, such as maternal undernutrition, seasonal diets, low-protein diet, high-fat diet, and synthetic folic acid supplement use, and how these nutritional exposures epigenetically affect target genes and their outcomes. In addition, we review the concept of thrifty genes during the preimplantation period, and some examples that relate to epigenetic change and diet. Finally, we discuss different examples of maternal diets, their effect on outcomes, and their relationship with assisted reproductive technology (ART), including their implications on epigenetic modifications.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Desarrollo Embrionario/genética , Epigénesis Genética/genética , Epigenoma/genética , Técnicas Reproductivas Asistidas/efectos adversos , Útero/fisiología , Animales , Metilación de ADN/genética , Femenino , Humanos
8.
Proc Natl Acad Sci U S A ; 115(31): E7398-E7407, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-29941596

RESUMEN

Maternal protein malnutrition throughout pregnancy and lactation compromises brain development in late gestation and after birth, affecting structural, biochemical, and pathway dynamics with lasting consequences for motor and cognitive function. However, the importance of nutrition during the preimplantation period for brain development is unknown. We have previously shown that maternal low-protein diet (LPD) confined to the preimplantation period (Emb-LPD) in mice, with normal nutrition thereafter, is sufficient to induce cardiometabolic and locomotory behavioral abnormalities in adult offspring. Here, using a range of in vivo and in vitro techniques, we report that Emb-LPD and sustained LPD reduce neural stem cell (NSC) and progenitor cell numbers at E12.5, E14.5, and E17.5 through suppressed proliferation rates in both ganglionic eminences and cortex of the fetal brain. Moreover, Emb-LPD causes remaining NSCs to up-regulate the neuronal differentiation rate beyond control levels, whereas in LPD, apoptosis increases to possibly temper neuron formation. Furthermore, Emb-LPD adult offspring maintain the increase in neuron proportion in the cortex, display increased cortex thickness, and exhibit short-term memory deficit analyzed by the novel-object recognition assay. Last, we identify altered expression of fragile X family genes as a potential molecular mechanism for adverse programming of brain development. Collectively, these data demonstrate that poor maternal nutrition from conception is sufficient to cause abnormal brain development and adult memory loss.


Asunto(s)
Encéfalo/embriología , Dieta con Restricción de Proteínas , Fenómenos Fisiologicos Nutricionales Maternos , Memoria a Corto Plazo , Células-Madre Neurales/patología , Neurogénesis , Animales , Apoptosis , Encéfalo/patología , Diferenciación Celular , Proliferación Celular , Femenino , Ratones
9.
Stem Cell Reports ; 8(3): 743-757, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28216148

RESUMEN

The success of stem cell-mediated gene therapy in cancer treatment largely depends on the specific homing ability of stem cells. We have previously demonstrated that after in vitro induction of neuronal differentiation and dedifferentiation, bone marrow stromal cells (BMSCs) revert to a primitive stem cell population (De-neu-BMSCs) distinct from naive BMSCs. We report here that De-neu-BMSCs express significantly higher levels of chemokines, and display enhanced homing abilities to glioma, the effect of which is mediated by the activated CCL5/CCR1/ERK axis. Intriguingly, we find that the activated chemokine axis in De-neu-BMSCs is epigenetically regulated by histone modifications. On the therapeutic front, we show that De-neu-BMSCs elicit stronger homing and glioma-killing effects together with cytosine deaminase/5-fluorocytosine compared with unmanipulated BMSCs in vivo. Altogether, the current study provides an insight into chemokine regulation in BMSCs, which may have more profound effects on BMSC function and their application in regenerative medicine and cancer targeting.


Asunto(s)
Quimiocina CCL5/metabolismo , Epigénesis Genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glioma/genética , Glioma/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Receptores CCR1/metabolismo , Animales , Desdiferenciación Celular , Movimiento Celular/genética , Reprogramación Celular , Quimiocinas/metabolismo , Histonas/metabolismo , Humanos , Ratones , Transducción de Señal
10.
Mol Neurobiol ; 54(5): 3893-3905, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27541285

RESUMEN

Glioblastoma is the most common form of primary malignant brain tumour. These tumours are highly proliferative and infiltrative resulting in a median patient survival of only 14 months from diagnosis. The current treatment regimens are ineffective against the small population of cancer stem cells residing in the tumourigenic niche; however, a new therapeutic approach could involve the removal of these cells from the microenvironment that maintains the cancer stem cell phenotype. We have isolated multipotent sphere-forming cells from human high grade glioma (glioma sphere-forming cells (GSCs)) to investigate the adhesive and migratory properties of these cells in vitro. We have focused on the role of two closely related metalloproteinases ADAM10 and ADAM17 due to their high expression in glioblastoma and GSCs and their ability to activate cytokines and growth factors. Here, we report that ADAM10 and ADAM17 inhibition selectively increases GSC, but not neural stem cell, migration and that the migrated GSCs exhibit a differentiated phenotype. We also observed a correlation between nestin, a stem/progenitor marker, and fibronectin, an extracellular matrix protein, expression in high grade glioma tissues. GSCs adherence on fibronectin is mediated by α5ß1 integrin, where fibronectin further promotes GSC migration and is an effective candidate for in vivo cancer stem cell migration out of the tumourigenic niche. Our results suggest that therapies against ADAM10 and ADAM17 may promote cancer stem cell migration away from the tumourigenic niche resulting in a differentiated phenotype that is more susceptible to treatment.


Asunto(s)
Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Movimiento Celular , Esferoides Celulares/metabolismo , Adulto , Anciano , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Matriz Extracelular/ultraestructura , Femenino , Fibronectinas/farmacología , Glioblastoma/patología , Humanos , Integrina alfa5beta1/metabolismo , Masculino , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo
11.
Expert Opin Drug Discov ; 11(4): 355-67, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26878555

RESUMEN

INTRODUCTION: Although many disease models exist for neurodegenerative disease, the translation of basic research findings to clinic is very limited. Studies using freshly resected human brain tissue, commonly discarded from neurosurgical procedures, should complement on-going work using stem cell-derived human neurons and glia thus increasing the likelihood of success in clinical trials. AREAS COVERED: Herein, the authors discuss key issues in the lack of translation from basic research to clinic. They also review the evidence that human neurons, both freshly resected brain tissue and stem cell-derived neurons, such as induced pluripotent stem cells (iPSCs), can be used for analysis of physiological and molecular mechanisms in health and disease. Furthermore, the authors compare and contrast studies using live human brain tissue and studies using induced human stem cell-derived neuron models. Using an example from the area of neurodegeneration, the authors suggest that replicating elements of research findings from animals and stem cell models in resected human brain tissue would strengthen our understanding of disease mechanisms and the therapeutic strategies and aid translation. EXPERT OPINION: The use of human brain tissue alongside iPSC-derived neural models can validate molecular mechanisms identified in rodent disease models and strengthen their relevance to humans. If drug target engagement and mechanism of cellular action can be validated in human brain tissue, this will increase the success rate in clinical research. The combined use of resected human brain tissue, alongside iPSC-derived neural models, could be considered a standard step in pre-clinical research and help to bridge the gap to clinical trials.


Asunto(s)
Demencia/tratamiento farmacológico , Descubrimiento de Drogas/métodos , Neuronas/metabolismo , Animales , Encéfalo/fisiopatología , Demencia/fisiopatología , Modelos Animales de Enfermedad , Humanos , Células Madre Pluripotentes Inducidas , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/fisiopatología , Investigación Biomédica Traslacional/métodos
12.
J Pathol Clin Res ; 1(2): 67-75, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27499894

RESUMEN

Brain tumour stem cells and microglia both promote the growth of astrocytomas, the commonest form of primary brain tumour, with recent emerging evidence that these cell types may interact in glioma models. It is unclear whether microglia and stem cells are associated in human gliomas. To investigate this question, we used the technique of tissue microarrays to perform a correlative study of a large number of tumour samples. We quantified immunostaining of human astrocytic tumour tissue microarrays (86 patients; World Health Organisation grade II-IV) for microglia Ionized calcium binding adaptor molecule 1 (Iba1) and CD68, and stem cell nestin, SOX2 and CD133. Ki67 was used to assess proliferation and GFAP for astrocytic differentiation. Immunoreactivity for both microglial markers and stem cell markers nestin and SOX2 significantly increased with increasing tumour grade. GFAP was higher in low grade astrocytomas. There was a positive correlation between: (i) both microglial markers and nestin and CD133, (ii) nestin and tumour cell proliferation Ki67 and (iii) both microglial markers and Ki67. SOX2 was not associated with microglia or tumour proliferation. To test the clinical relevance, we investigated the putative association of these markers with clinical outcomes. High expression for nestin and Iba1 correlated with significantly shorter survival times, and high expression for nestin, Iba1, CD68 and Ki67 was associated with faster tumour progression on univariate analysis. On multivariate analysis, nestin, CD133 and Ki67 remained significant predictors of poorer survival, after adjustment for other markers. These results confirm previous in vitro findings, demonstrating their functional relevance as a therapeutic target in humans. This is the first report of a novel correlation between microglia and stem cells that may drive human astrocytic tumour development.

13.
Cancer Lett ; 326(1): 79-87, 2012 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-22841667

RESUMEN

It has been proposed that gliomas contain a subpopulation of 'Brain Tumor Stem Cells' (BTSCs), which demonstrate resistance to conventional therapies. A potential component of the environment governing the behavior of these BTSCs is a class of transmembrane proteins with structural and signaling functions, the A-Disintegrin And Metalloproteases (ADAMs). In this study we confirm overexpression of ADAM10 and 17 in human glioma tissue compared to human controls, and especially in tumor sphere cultures thought to enrich for BTSCs. Inhibition of ADAM10/17 function impairs the growth of tumor spheres with evidence of depletion of the sphere forming cell population. This results from a combination of reduced proliferation, cell death and a switch of sphere-forming cells away from symmetric self-renewal division towards neuronal differentiation. A developing appreciation of the role of ADAMs in BTSC promises insights into pathophysiology and potential therapeutic avenues in this intractable group of tumors.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Células Madre Neoplásicas/metabolismo , Esferoides Celulares , Células Tumorales Cultivadas , Proteínas ADAM/antagonistas & inhibidores , Proteína ADAM10 , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Fertilinas , Humanos
14.
J Neurosci ; 29(12): 3885-96, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-19321785

RESUMEN

E-Cadherin, a cell adhesion protein, has been shown to take part in the compartmentalization, proliferation, survival, and differentiation of cells. E-Cadherin is expressed in the adult and embryonic forebrain germinal zones in vivo, and in clonal colonies of cells derived from these regions and grown in vitro. Mice carrying E-Cadherin floxed genes crossed to mice expressing Cre under the Nestin promoter demonstrate defects in the self-renewal of neural stem cells both in vivo and in vitro. The functional role of E-Cadherin is further demonstrated using adhesion-blocking antibodies in vitro, which specifically target cadherin extracellular adhesive domains. Adult neural stem cell colonies decrease in the presence of E-Cadherin antibodies in a dosage-dependent manner, in contrast to P-Cadherin antibody. On overexpression of normal E-Cadherin and a mutated E-Cadherin, containing no intracellular binding domain, an increased number of clonal adult neural stem cell colonies are observed. These data suggest it is specifically E-Cadherin adhesion that is responsible for these self-renewal effects. These data show the importance of E-Cadherin in the neural stem cell niche and suggest E-Cadherin regulates the number of these cells.


Asunto(s)
Cadherinas/fisiología , Neuronas/citología , Prosencéfalo/metabolismo , Células Madre/citología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Cadherinas/genética , Proliferación Celular , Células Cultivadas , Cruzamientos Genéticos , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Ratones , Ratones Transgénicos , Mutación , Neuronas/metabolismo , Prosencéfalo/citología , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Células Madre/metabolismo
15.
J Neuropathol Exp Neurol ; 67(6): 565-77, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18520775

RESUMEN

Schimke immuno-osseous dysplasia (OMIM 242900) is an uncommon autosomal-recessive multisystem disease caused by mutations in SMARCAL1 (swi/snf-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a-like 1), a gene encoding a putative chromatin remodeling protein. Neurologic manifestations identified to date relate to enhanced atherosclerosis and cerebrovascular disease. Based on a clinical survey, we determined that half of Schimke immuno-osseous dysplasia patients have a small head circumference, and 15% have social, language, motor, or cognitive abnormalities. Postmortem examination of 2 Schimke immuno-osseous dysplasia patients showed low brain weights and subtle brain histologic abnormalities suggestive of perturbed neuron-glial migration such as heterotopia, irregular cortical thickness, incomplete gyral formation, and poor definition of cortical layers. We found that SMARCAL1 is highly expressed in the developing and adult mouse and human brain, including neural precursors and neuronal lineage cells. These observations suggest that SMARCAL1 deficiency may influence brain development and function in addition to its previously recognized effect on cerebral circulation.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/patología , ADN Helicasas/biosíntesis , Síndromes de Inmunodeficiencia/metabolismo , Osteocondrodisplasias/metabolismo , Animales , Northern Blotting , Western Blotting , Encéfalo/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunohistoquímica , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/patología , Hibridación in Situ , Ratones , Microcefalia/etiología , Osteocondrodisplasias/complicaciones , Osteocondrodisplasias/patología , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Eur J Neurosci ; 27(9): 2354-62, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18445225

RESUMEN

Neural stem cells can be isolated from the mouse embryonic cortex but do not persist in the adult cortex. In contrast, neural stem cells from the striatal embryonic germinal zone persist in the adult subependyma. Emx1-lineage analysis revealed that cortex-derived neural stem cells survive and migrate ventrally into the subependyma where they intermix with the host striatal neural stem cells [S. Willaime-Morawek et al. (2006)J. Cell Biol. 175, 159-168]. Cortex-derived cells proliferate faster in the subependyma and reach the olfactory bulb earlier than striatum-derived cells. In the olfactory bulb, cortex-derived cells produce more cells and more dopaminergic neurons in the glomerular layer than striatum-derived cells. Cortex-derived cells also give rise to more astrocytes and less neurons in the striatum than striatum-derived cells. Thus, history matters; cortex-derived neural stem cells in the subependyma give rise to progeny in the olfactory bulb and striatum but in different proportions than striatum-derived neural stem cells.


Asunto(s)
Células Madre Adultas/citología , Corteza Cerebral/citología , Cuerpo Estriado/citología , Neuronas/citología , Bulbo Olfatorio/citología , Animales , Bromodesoxiuridina , Linaje de la Célula , Movimiento Celular/fisiología , Inmunohistoquímica , Ratones
17.
Genome Biol ; 8(6): R108, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17565696

RESUMEN

BACKGROUND: Alternative splicing (AS) functions to expand proteomic complexity and plays numerous important roles in gene regulation. However, the extent to which AS coordinates functions in a cell and tissue type specific manner is not known. Moreover, the sequence code that underlies cell and tissue type specific regulation of AS is poorly understood. RESULTS: Using quantitative AS microarray profiling, we have identified a large number of widely expressed mouse genes that contain single or coordinated pairs of alternative exons that are spliced in a tissue regulated fashion. The majority of these AS events display differential regulation in central nervous system (CNS) tissues. Approximately half of the corresponding genes have neural specific functions and operate in common processes and interconnected pathways. Differential regulation of AS in the CNS tissues correlates strongly with a set of mostly new motifs that are predominantly located in the intron and constitutive exon sequences neighboring CNS-regulated alternative exons. Different subsets of these motifs are correlated with either increased inclusion or increased exclusion of alternative exons in CNS tissues, relative to the other profiled tissues. CONCLUSION: Our findings provide new evidence that specific cellular processes in the mammalian CNS are coordinated at the level of AS, and that a complex splicing code underlies CNS specific AS regulation. This code appears to comprise many new motifs, some of which are located in the constitutive exons neighboring regulated alternative exons. These data provide a basis for understanding the molecular mechanisms by which the tissue specific functions of widely expressed genes are coordinated at the level of AS.


Asunto(s)
Empalme Alternativo , Sistema Nervioso Central/metabolismo , Regulación de la Expresión Génica , Animales , Exones , Perfilación de la Expresión Génica , Intrones , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , Secuencias Reguladoras de Ácidos Nucleicos
18.
J Cell Biol ; 175(1): 159-68, 2006 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-17030986

RESUMEN

Embryonic cortical neural stem cells apparently have a transient existence, as they do not persist in the adult cortex. We sought to determine the fate of embryonic cortical stem cells by following Emx1(IREScre); LacZ/EGFP double-transgenic murine cells from midgestation into adulthood. Lineage tracing in combination with direct cell labeling and time-lapse video microscopy demonstrated that Emx1-lineage embryonic cortical stem cells migrate ventrally into the striatal germinal zone (GZ) perinatally and intermingle with striatal stem cells. Upon integration into the striatal GZ, cortical stem cells down-regulate Emx1 and up-regulate Dlx2, which is a homeobox gene characteristic of the developing striatum and striatal neural stem cells. This demonstrates the existence of a novel dorsal-to-ventral migration of neural stem cells in the perinatal forebrain.


Asunto(s)
Movimiento Celular , Corteza Cerebral/citología , Embrión de Mamíferos/citología , Neuronas/citología , Células Madre/fisiología , Animales , Ganglios Basales/citología , Linaje de la Célula , Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Proteínas de Homeodominio/análisis , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Microscopía por Video , Fenotipo , Células Madre/citología , Factores de Transcripción/análisis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
J Neurosci ; 26(41): 10452-60, 2006 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-17035529

RESUMEN

Neural stem and progenitor cells are located in the subependyma of the adult forebrain. An increase in adult subependymal cell proliferation is reported after various kinds of brain injury. We demonstrate an expansion of neural precursor cells in the postnatal subependyma in a murine genetic disease model of Huntington's disease (HD), the R6/2 mouse. We used the in vitro neurosphere assay as an index of the number of neural stem cells in vivo and to assess proliferation kinetics in vitro and in vivo bromodeoxyuridine labeling to assess the progenitor cell population and their fates. Disease progression in this model leads to an increase in the numbers of neural stem cells in the adult striatal subependyma. This increase is produced cell non-autonomously by events in the R6/2 brains as the mice become increasingly symptomatic. Once the neural stem cell increase is induced in vivo, it is maintained during in vitro passaging of neural stem cells, but the neural stem cell increase is not reproduced during in vitro passaging of neural stem cells from presymptomatic R6/2 mice. In addition, we show that some of the R6/2 neural progenitor cells show a change from their normal migration destiny toward the olfactory bulb. Instead, some of these cells migrate into the striatum, one of the main affected areas in HD. Our findings demonstrate that HD damage recruits precursor cells in two ways: expansion of neural stem cells and altered migration of progenitor cells.


Asunto(s)
Proliferación Celular , Cuerpo Estriado/citología , Enfermedad de Huntington/patología , Neuronas/citología , Células Madre/citología , Animales , Movimiento Celular/fisiología , Cuerpo Estriado/fisiología , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Ratones , Ratones Mutantes Neurológicos , Neuronas/fisiología , Células Madre/fisiología
20.
Brain Res Mol Brain Res ; 142(2): 97-106, 2005 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-16290312

RESUMEN

Current understanding of IGF-I-mediated neuroprotection implies the activation of phosphatidylinositol-3-kinase (PI-3K), which leads to the activation of Akt/Protein Kinase B. In non-neuronal cells, Akt phosphorylates and activates the transcription factor CREB, implicated in the transcription of the anti-apoptotic bcl-2 gene. This paper further analyses the anti-apoptotic IGF-I action in neurons. We show that IGF-I protects cortical neurons against ceramide-induced apoptosis. Ceramide decreases Akt phosphorylation during apoptotic process whereas a simultaneous treatment with IGF-I increases Akt phosphorylation. Analysis of the signal transduction pathways revealed that IGF-I induces CREB phosphorylation via PI-3K and ERK, whereas simultaneous ceramide and IGF-I treatment decreases CREB phosphorylation. Although an overexpression of Bcl-2 protects cortical neurons against ceramide-induced apoptosis, our data indicate that the Bcl-2 protein level is not modulated during IGF-I, ceramide and/or LY294002 treatment. In consequence, we demonstrated that IGF protects neurons against ceramide-induced apoptosis and that IGF-I protection involves the PI-3K/Akt and ERK pathways; this protection may be independent of CREB and Bcl-2.


Asunto(s)
Apoptosis/efectos de los fármacos , Corteza Cerebral/citología , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Western Blotting/métodos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ceramidas/toxicidad , Cromonas/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Embrión de Mamíferos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Genes bcl-2/fisiología , Proteínas Fluorescentes Verdes/biosíntesis , Inmunohistoquímica/métodos , Ratones , Ratones Transgénicos , Morfolinas/farmacología , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...