Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Intervalo de año de publicación
1.
Nature ; 618(7966): 842-848, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37258671

RESUMEN

Nonsense mutations are the underlying cause of approximately 11% of all inherited genetic diseases1. Nonsense mutations convert a sense codon that is decoded by tRNA into a premature termination codon (PTC), resulting in an abrupt termination of translation. One strategy to suppress nonsense mutations is to use natural tRNAs with altered anticodons to base-pair to the newly emerged PTC and promote translation2-7. However, tRNA-based gene therapy has not yielded an optimal combination of clinical efficacy and safety and there is presently no treatment for individuals with nonsense mutations. Here we introduce a strategy based on altering native tRNAs into  efficient suppressor tRNAs (sup-tRNAs) by individually fine-tuning their sequence to the physico-chemical properties of the amino acid that they carry. Intravenous and intratracheal lipid nanoparticle (LNP) administration of sup-tRNA in mice restored the production of functional proteins with nonsense mutations. LNP-sup-tRNA formulations caused no discernible readthrough at endogenous native stop codons, as determined by ribosome profiling. At clinically important PTCs in the cystic fibrosis transmembrane conductance regulator gene (CFTR), the sup-tRNAs re-established expression and function in cell systems and patient-derived nasal epithelia and restored airway volume homeostasis. These results provide a framework for the development of tRNA-based therapies with a high molecular safety profile and high efficacy in targeted PTC suppression.


Asunto(s)
Codón sin Sentido , Regulador de Conductancia de Transmembrana de Fibrosis Quística , ARN de Transferencia , Animales , Ratones , Aminoácidos/genética , Codón sin Sentido/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , ARN de Transferencia/administración & dosificación , ARN de Transferencia/genética , ARN de Transferencia/uso terapéutico , Emparejamiento Base , Anticodón/genética , Biosíntesis de Proteínas , Mucosa Nasal/metabolismo , Perfilado de Ribosomas
2.
Int Rev Cell Mol Biol ; 372: 159-174, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36064263

RESUMEN

Phenylketonuria (PKU) is a metabolic rare disease characterized by a failure of the body to clear out the high levels of Phenylalanine (Phe), leading to devastating neurological defects and growth retardation. PKU was discovered in 1934 by AsbjrØrn FØlling, and even though there have been continuous efforts from the scientific community to find therapeutic approaches to modulate the high levels of phenylalanine found in the body of the PKU patients, an efficient therapy still needs to be developed. Current standard of care includes low phenylalanine diets, but the strict restrictions for patients and families makes it very difficult to adequately being implemented. FDA has approved two drugs to help reduce Phe levels in PKU patients: an enzyme substitution therapy, Palynziq® (PEGylated recombinant phenylalanine ammonia lyase), and Kuvan®, a supplemental tetrahydrobiopterin (BH4) cofactor that enhances residual enzyme activity. Both treatments are restricted to certain PKU patients' population, and, therefore, there are still high unmet needs for most of the patients. The present review will focus on current advancements in lipid nanoparticles (LNP)-mRNA technologies and their potential in treating the root cause of PKU, a therapeutic approach that will be analyzed in the context of other promising therapeutic approaches that are been developed for PKU.


Asunto(s)
Fenilcetonurias , Humanos , Liposomas , Nanopartículas , Fenilalanina/metabolismo , Fenilalanina/uso terapéutico , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/genética , ARN Mensajero/uso terapéutico , Tecnología
3.
Mol Ther Nucleic Acids ; 28: 87-98, 2022 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-35356682

RESUMEN

Phenylketonuria (PKU) is an inborn error caused by deficiencies in phenylalanine (Phe) metabolism. Mutations in the phenylalanine hydroxylase (PAH) gene are the main cause of the disease whose signature hallmarks of toxically elevated levels of Phe accumulation in plasma and organs such as the brain, result in irreversible intellectual disability. Here, we present a unique approach to treating PKU deficiency by using an mRNA replacement therapy. A full-length mRNA encoding human PAH (hPAH) is encapsulated in our proprietary lipid nanoparticle LUNAR and delivered to a Pah enu2 mouse model that carries a missense mutation in the mouse PAH gene. Animals carrying this missense mutation develop hyperphenylalanemia and hypotyrosinemia in plasma, two clinical features commonly observed in the clinical presentation of PKU. We show that intravenous infusion of LUNAR-hPAH mRNA can generate high levels of hPAH protein in hepatocytes and restore the Phe metabolism in the Pah enu2 mouse model. Together, these data establish a proof of principle of a novel mRNA replacement therapy to treat PKU.

4.
Chem Phys Lipids ; 243: 105178, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35122738

RESUMEN

Lipid nanoparticles (LNPs) mediated mRNA delivery has gained prominence due to the success of mRNA vaccines against Covid-19, without which it would not have been possible. However, there is little clinical validation of this technology for other mRNA-based therapeutic approaches. Systemic administration of LNPs predominantly targets the liver, but delivery to other organs remains a challenge. Local approaches remain a viable option for some disease indications, such as Cystic Fibrosis, where aerosolized delivery to airway epithelium is the preferred route of administration. With this in mind, novel cationic lipids (L1-L4) have been designed, synthesized and co-formulated with a proprietary ionizable lipid. These LNPs were further nebulized, along with baseline control DOTAP-based LNP (DOTAP+), and tested in vitro for mRNA integrity and encapsulation efficiency, as well as transfection efficiency and cytotoxicity in cell cultures. Improved biodegradability and potentially superior elimination profiles of L1-L4, in part due to physicochemical characteristics of putative metabolites, are thought to be advantageous for prospective therapeutic lung delivery applications using these lipids.


Asunto(s)
Liposomas/química , Pulmón , Nanopartículas/química , ARN Mensajero/administración & dosificación , Humanos
5.
Cell Rep ; 14(3): 560-571, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26776515

RESUMEN

The subventricular zone (SVZ) provides a specialized neurogenic microenvironment for proliferation and aggregation of basal progenitors (BPs). Our study reveals a mechanism for the aggregation of BPs within the SVZ required for their proliferation and generation of cortical layer neurons. The autism-related IgCAM, MDGA1, is locally expressed in the BP cell membrane where it co-localizes and complexes with the gap junction protein Connexin43. To address MDGA1 function, we created a floxed allele of MDGA1 and deleted it from BPs. MDGA1 deletion results in reduced BP proliferation and size of the SVZ, with an aberrant population of BPs ectopically positioned in the cortical plate. These defects are manifested in diminished production of cortical layer neurons and a significant reduction of the cortical layers. We conclude that MDGA1 functions to aggregate and maintain BPs within the SVZ providing the neurogenic niche required for their proliferation and generation of cortical layer neurons.


Asunto(s)
Ventrículos Laterales/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Conexina 43/genética , Conexina 43/metabolismo , Células HEK293 , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Moléculas de Adhesión de Célula Nerviosa/deficiencia , Moléculas de Adhesión de Célula Nerviosa/genética , Células Madre/citología , Células Madre/metabolismo
6.
Proc Natl Acad Sci U S A ; 112(21): 6736-41, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25971728

RESUMEN

Current knowledge suggests that cortical sensory area identity is controlled by transcription factors (TFs) that specify area features in progenitor cells and subsequently their progeny in a one-step process. However, how neurons acquire and maintain these features is unclear. We have used conditional inactivation restricted to postmitotic cortical neurons in mice to investigate the role of the TF LIM homeobox 2 (Lhx2) in this process and report that in conditional mutant cortices area patterning is normal in progenitors but strongly affected in cortical plate (CP) neurons. We show that Lhx2 controls neocortical area patterning by regulating downstream genetic and epigenetic regulators that drive the acquisition of molecular properties in CP neurons. Our results question a strict hierarchy in which progenitors dominate area identity, suggesting a novel and more comprehensive two-step model of area patterning: In progenitors, patterning TFs prespecify sensory area blueprints. Sequentially, sustained function of alignment TFs, including Lhx2, is essential to maintain and to translate the blueprints into functional sensory area properties in cortical neurons postmitotically. Our results reemphasize critical roles for Lhx2 that acts as one of the terminal selector genes in controlling principal properties of neurons.


Asunto(s)
Proteínas con Homeodominio LIM/fisiología , Modelos Neurológicos , Neocórtex/crecimiento & desarrollo , Neocórtex/fisiología , Factores de Transcripción/fisiología , Animales , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Epigénesis Genética , Proteínas con Homeodominio LIM/deficiencia , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Noqueados , Mitosis , Neocórtex/citología , Vías Nerviosas/citología , Vías Nerviosas/crecimiento & desarrollo , Vías Nerviosas/fisiología , Neuronas/citología , Neuronas/fisiología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
7.
Front Cell Neurosci ; 9: 472, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26733804

RESUMEN

The susceptibility genes for schizophrenia Neuregulin-1 (NRG1) and ErbB4 have critical functions during brain development and in the adult. Alterations in the ErbB4 signaling pathway cause a variety of neurodevelopmental defects including deficiencies in neuronal migration, synaptic plasticity, and myelination. I have used the ErbB4(-/-) HER4(heart) KO mice to study the neurodevelopmental insults associated to deficiencies in the NRG1-ErbB4 signaling pathway and their potential implication with brain disorders such as schizophrenia, a chronic psychiatric disease affecting 1% of the population worldwide. ErbB4 deletion results in an array of neurodevelopmental deficits that are consistent with a schizophrenic model. First, similar defects appear in multiple brain structures, from the cortex to the cerebellum. Second, these defects affect multiple aspects of brain development, from deficits in neuronal migration to impairments in excitatory/inhibitory systems, including reductions in brain volume, cortical and cerebellar heterotopias, alterations in number and distribution of specific subpopulations of interneurons, deficiencies in the astrocytic and oligodendrocytic lineages, and additional insults in major brain structures. This suggests that alterations in specific neurodevelopmental genes that play similar functions in multiple neuroanatomical structures might account for some of the symptomatology observed in schizophrenic patients, such as defects in cognition. ErbB4 mutation uncovers flaws in brain development that are compatible with a neurodevelopmental model of schizophrenia, and it establishes a comprehensive model to study the basis of the disorder before symptoms are detected in the adult.

8.
Proc Natl Acad Sci U S A ; 111(13): E1240-8, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24639535

RESUMEN

Breast cancer susceptibility gene 1 (BRCA1) is a breast and ovarian cancer tumor suppressor whose loss leads to DNA damage and defective centrosome functions. Despite its tumor suppression functions, BRCA1 is most highly expressed in the embryonic neuroepithelium when the neural progenitors are highly proliferative. To determine its functional significance, we deleted BRCA1 in the developing brain using a neural progenitor-specific driver. The phenotype is characterized by severe agenesis of multiple laminated cerebral structures affecting most notably the neocortex, hippocampus, cerebellum, and olfactory bulbs. Major phenotypes are caused by excess apoptosis, as these could be significantly suppressed by the concomitant deletion of p53. Certain phenotypes attributable to centrosomal and cell polarity functions could not be rescued by p53 deletion. A double KO with the DNA damage sensor kinase ATM was able to rescue BRCA1 loss to a greater extent than p53. Our results suggest distinct apoptotic and centrosomal functions of BRCA1 in neural progenitors, with important implications to understand the sensitivity of the embryonic brain to DNA damage, as well as the developmental regulation of brain size.


Asunto(s)
Proteína BRCA1/metabolismo , Encéfalo/embriología , Encéfalo/metabolismo , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Encéfalo/citología , Polaridad Celular , Proliferación Celular , Supervivencia Celular , Cognición/fisiología , Eliminación de Gen , Aprendizaje , Ratones , Ratones Noqueados , Actividad Motora/fisiología , Nestina/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Fenotipo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
9.
Neural Dev ; 7: 10, 2012 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-22376909

RESUMEN

BACKGROUND: Cortical GABAergic interneurons (INs) are generated in the medial ganglionic eminence (MGE) and migrate tangentially into cortex. Because most, if not all, migrating MGE-derived INs express the neuregulin (NRG) receptor, ErbB4, we investigated influences of Nrg1 isoforms and Nrg3 on IN migration through ventral telencephalon (vTel) and within cortex. RESULTS: During IN migration, NRG expression domains and distributions of ErbB4-expressing, MGE-derived INs are complementary with minimal overlap, both in vTel and cortex. In wild-type mice, within fields of NRG expression, these INs are focused at positions of low or absent NRG expression. However, in ErbB4-/- HER4(heart) mutant mice in which INs lack ErbB4, these complementary patterns are degraded with considerable overlap evident between IN distribution and NRG expression domains. These findings suggest that NRGs are repellents for migrating ErbB4-expressing INs, a function supported by in vitro and in vivo experiments. First, in collagen co-cultures, MGE-derived cells preferentially migrate away from a source of secreted NRGs. Second, cells migrating from wild-type MGE explants on living forebrain slices from wild-type embryonic mice tend to avoid endogenous NRG expression domains, whereas this avoidance behavior is not exhibited by ErbB4-deficient cells migrating from MGE explants and instead they have a radial pattern with a more uniform distribution. Third, ectopic NRG expression in the IN migration pathway produced by in utero electroporation blocks IN migration and results in cortex distal to the blockade being largely devoid of INs. Finally, fewer INs reach cortex in ErbB4 mutants, indicating that NRG-ErbB4 signaling is required for directing IN migration from the MGE to cortex. CONCLUSIONS: Our results show that NRGs act as repellents for migrating ErbB4-expressing, MGE-derived GABAergic INs and that the patterned expression of NRGs funnels INs as they migrate from the MGE to their cortical destinations.


Asunto(s)
Movimiento Celular/fisiología , Corteza Cerebral/fisiología , Receptores ErbB/metabolismo , Neuronas GABAérgicas/fisiología , Interneuronas/fisiología , Neurregulinas/metabolismo , Transducción de Señal/fisiología , Animales , Células Cultivadas , Corteza Cerebral/citología , Neuronas GABAérgicas/citología , Interneuronas/citología , Ratones , Ratones Noqueados , Receptor ErbB-4
10.
Proc Natl Acad Sci U S A ; 108(41): 17189-94, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21957071

RESUMEN

Lifeguard (LFG) is an inhibitor of Fas-mediated cell death and is highly expressed in the cerebellum. We investigated the biological role of LFG in the cerebellum in vivo, using mice with reduced LFG expression generated by shRNA lentiviral transgenesis (shLFG mice) as well as LFG null mice. We found that LFG plays a role in cerebellar development by affecting cerebellar size, internal granular layer (IGL) thickness, and Purkinje cell (PC) development. All these features are more severe in early developmental stages and show substantial recovery overtime, providing a remarkable example of cerebellar plasticity. In adult mice, LFG plays a role in PC maintenance shown by reduced cellular density and abnormal morphology with increased active caspase 8 and caspase 3 immunostaining in shLFG and knockout (KO) PCs. We studied the mechanism of action of LFG as an inhibitor of the Fas pathway and provided evidence of the neuroprotective role of LFG in cerebellar granule neurons (CGNs) and PCs in an organotypic cerebellar culture system. Biochemical analysis of the Fas pathway revealed that LFG inhibits Fas-mediated cell death by interfering with caspase 8 activation. This result is supported by the increased number of active caspase 8-positive PCs in adult mice lacking LFG. These data demonstrate that LFG is required for proper development and survival of granular and Purkinje cells and suggest LFG may play a role in cerebellar disorders.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Cerebelo/citología , Cerebelo/fisiología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Células de Purkinje/citología , Células de Purkinje/fisiología , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/genética , Secuencia de Bases , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Diferenciación Celular/fisiología , Supervivencia Celular/fisiología , Cerebelo/anomalías , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , ARN Interferente Pequeño/genética , Receptor fas/metabolismo
11.
Eur. j. anat ; 13(3): 111-115, dic. 2009. ilus
Artículo en Inglés | IBECS | ID: ibc-107642

RESUMEN

The subcommissural organ (SCO) releases glycoproteins into the ventricular cerebrospinal fluid (CSF), where they form Reissner's fibre (RF) and also secretes a CSF-soluble material different from RF-material. Pax6 is a transcription factor important for the regulation of cell proliferation, migration and differentiation in the developing brain. In the present work, we studied wild-type, heterozygous and homozygous Sey mice to compare the expression of RF-antibody and Pax6 in the SCO and adjacent structures. In wild-type mice between E15 to E18, we observed Pax6 expression in cells surrounding the secretory cells of the SCO, and RF-immunoreactive material only in the SCO ependymal cell layer and its basal process. In the heterozygous mice, the neuroanatomical structure of the SCO was present, but RF-antibody staining and Pax6 expression was scarce or almost undetectable; in the homozygous mice neither SCO nor other epithalamic structures were found. We suggest that Pax6 expression at the periphery of the SCO is essential for the development and activity of the organ (AU)


No disponible


Asunto(s)
Animales , Ratas , Inmunohistoquímica/métodos , Órgano Subcomisural/embriología , Factores de Transcripción Paired Box , Ratas/embriología , Desarrollo Fetal , Glicoproteínas , Encéfalo/embriología , Ventrículos Cerebrales/embriología , Tálamo/embriología
12.
Nat Neurosci ; 12(11): 1381-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19820705

RESUMEN

Cerebral cortex is comprised of regions, including six-layer neocortex and three-layer olfactory cortex, generated by telencephalic progenitors of an Emx1 lineage. The mechanism specifying region-specific subpopulations in this lineage is unknown. We found that the LIM homeodomain transcription factor Lhx2 in mice, expressed in graded levels by progenitors, determines their regional identity and fate decisions to generate neocortex or olfactory cortex. Deletion of Lhx2 with Emx1-cre at embryonic day 10.5 (E10.5) altered the fates of progenitors, causing them to generate three-layer cortex, phenocopying olfactory cortex rather than lateral neocortex. Progenitors did not generate ectopic olfactory cortex following Lhx2 deletion at E11.5. Thus, Lhx2 regulates a regional-fate decision by telencephalic progenitors during a critical period that ends as they differentiate from neuroepithelial cells to neuronogenic radial glia. These findings establish a genetic mechanism for determining regional-fate in the Emx1 lineage of telencephalic progenitors that generate cerebral cortex.


Asunto(s)
Corteza Cerebral/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/fisiología , Células Madre/fisiología , Telencéfalo/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Tipificación del Cuerpo/genética , Diferenciación Celular , Corteza Cerebral/embriología , Embrión de Mamíferos , Proteínas con Homeodominio LIM , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Telencéfalo/embriología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
13.
J Comp Neurol ; 503(6): 790-802, 2007 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-17570500

RESUMEN

The expression of estrogen receptors (ERs) in the developing and adult human brain has not been clearly established, although estrogens are crucial for neuronal differentiation, synapse formation, and cognitive functions. By using immunohistochemistry, we have studied the distribution of ER alpha and ER beta in human cerebral cortex and hippocampus from early prenatal stages to adult life. ER alpha was detected in the cortex at 9 gestational weeks (GW), with a high expression in proliferating zones and the cortical plate. The staining intensity decreased gradually during prenatal development but increased again from birth to adulthood. In contrast, ER beta was first detected at 15 GW in proliferating zones, and at 16/17 GW, numerous ER beta immunopositive cells were also observed in the cortical plate. ER beta expression persisted in the adult cortex, being widely distributed throughout cortical layers II-VI. In addition, from around 15 GW to adulthood, ER alpha and ER beta were expressed in human hippocampus mainly in pyramidal cells of Ammon's horn and in the dentate gyrus. Western blotting and immunohistochemistry in the adult cerebral cortex and hippocampus revealed lower protein expression of ER alpha compared with ER beta. Double immunostaining showed that during fetal life both ERs are expressed in neurons as well as in radial glia, although only ER alpha is expressed in the Cajal-Retzius neurons of the marginal zone. These observations demonstrate that the expression of ER alpha and ER beta displays different spatial-temporal patterns during human cortical and hippocampal development and suggest that both ERs may play distinct roles in several processes related to prenatal brain development.


Asunto(s)
Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Adolescente , Adulto , Western Blotting , Femenino , Feto , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neuronas/citología , Neuronas/metabolismo
14.
J Chem Neuroanat ; 27(1): 7-21, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15036359

RESUMEN

The expression of reelin, a large extracellular matrix glycoprotein, was studied in the brain of pre-spawning adult sea lampreys by immunohistochemistry using two monoclonal antibodies against this protein. Reelin immunoreactive (reln-ir) neurons were observed in the olfactory bulb, and pallial and subpallial regions in the telencephalon. In the diencephalon, reln-ir cells were observed in some hypothalamic nuclei, in the nucleus of Bellonci, and in the habenula. In the mesencephalon, this protein was detected in several nuclei related with the centrifugal visual system, although the optic tectum was devoid of immunoreactivity. The hindbrain showed several nuclei with immunopositive neurons, including the branchiomeric nerve motor nuclei and also some groups of non-giant cells of the reticular formation. The rostral spinal cord showed some immunopositive neurons mainly located in lateral and ventral positions. Overall, the pattern of distribution of reelin in the adult sea lamprey correlates with the previously reported in other adult vertebrates. Furthermore, the wide distribution of reelin in the adult lamprey brain is consistent with a possible existence of different roles for this protein not related with development in the central nervous system (CNS) of vertebrates (i.e. neuronal plasticity and/or maintenance).


Asunto(s)
Encéfalo/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Lampreas/metabolismo , Animales , Inmunohistoquímica , Proteínas del Tejido Nervioso , Neuronas/metabolismo , Proteína Reelina , Serina Endopeptidasas , Médula Espinal/metabolismo
15.
Cereb Cortex ; 12(12): 1225-36, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12427674

RESUMEN

The genes doublecortin (DCX) and LIS1 are required for proper cortical neuronal migration and differentiation in humans. Here, we study the expression pattern of the encoded proteins of these genes in developing human brain. LIS1 stained virtually all migrating neurons throughout periods of development. Initially, DCX extensively overlapped with Reelin in early preplate stage in radially oriented columns of cells in the ventricular zone, whereas at later stages, the majority of DCX-positive cells were horizontally oriented. During the cortical plate stage, two opposite patterns of DCX expression were found: in radially oriented apical processes, presumably of pyramidal cells in the cortical plate, and in non-radially oriented mono- or bipolar neurons with migratory morphologies in the deep compartments of the cerebral wall. The extensive co-localization of DCX and Calretinin in non-radially oriented neurons suggested a non-pyramidal phenotype. These cells assumed a more vertical orientation upon entering the subplate. In addition, DCX was expressed by cells in the subpial granular layer and by Cajal-Retzius cells. In a 19 week human fetal cortex with a LIS1 mutation, the number of Reelin-expressing Cajal-Retzius cells was reduced and their morphology was abnormal. DCX was expressed by cells in all regions, but in extremely low numbers, suggesting that LIS1 deficiency adversely affects the migration and differentiation of DCX- and Reelin-positive neurons.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Corteza Cerebral/crecimiento & desarrollo , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/fisiología , Neuropéptidos/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa , Calbindina 2 , Moléculas de Adhesión Celular Neuronal/metabolismo , Corteza Cerebral/anomalías , Corteza Cerebral/metabolismo , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Desarrollo Embrionario y Fetal , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Inmunohistoquímica , Interneuronas/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Mutación , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Células Piramidales/crecimiento & desarrollo , Células Piramidales/metabolismo , Proteína Reelina , Proteína G de Unión al Calcio S100/metabolismo , Serina Endopeptidasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...