Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 628(8007): 391-399, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38408487

RESUMEN

The human nervous system is a highly complex but organized organ. The foundation of its complexity and organization is laid down during regional patterning of the neural tube, the embryonic precursor to the human nervous system. Historically, studies of neural tube patterning have relied on animal models to uncover underlying principles. Recently, models of neurodevelopment based on human pluripotent stem cells, including neural organoids1-5 and bioengineered neural tube development models6-10, have emerged. However, such models fail to recapitulate neural patterning along both rostral-caudal and dorsal-ventral axes in a three-dimensional tubular geometry, a hallmark of neural tube development. Here we report a human pluripotent stem cell-based, microfluidic neural tube-like structure, the development of which recapitulates several crucial aspects of neural patterning in brain and spinal cord regions and along rostral-caudal and dorsal-ventral axes. This structure was utilized for studying neuronal lineage development, which revealed pre-patterning of axial identities of neural crest progenitors and functional roles of neuromesodermal progenitors and the caudal gene CDX2 in spinal cord and trunk neural crest development. We further developed dorsal-ventral patterned microfluidic forebrain-like structures with spatially segregated dorsal and ventral regions and layered apicobasal cellular organizations that mimic development of the human forebrain pallium and subpallium, respectively. Together, these microfluidics-based neurodevelopment models provide three-dimensional lumenal tissue architectures with in vivo-like spatiotemporal cell differentiation and organization, which will facilitate the study of human neurodevelopment and disease.


Asunto(s)
Tipificación del Cuerpo , Microfluídica , Tubo Neural , Humanos , Técnicas de Cultivo Tridimensional de Células , Diferenciación Celular , Cresta Neural/citología , Cresta Neural/embriología , Tubo Neural/citología , Tubo Neural/embriología , Células Madre Pluripotentes/citología , Prosencéfalo/citología , Prosencéfalo/embriología , Médula Espinal/citología , Médula Espinal/embriología
2.
Nature ; 609(7929): 907-910, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36171373

RESUMEN

Self-organizing three-dimensional cellular models derived from human pluripotent stem cells or primary tissue have great potential to provide insights into how the human nervous system develops, what makes it unique and how disorders of the nervous system arise, progress and could be treated. Here, to facilitate progress and improve communication with the scientific community and the public, we clarify and provide a basic framework for the nomenclature of human multicellular models of nervous system development and disease, including organoids, assembloids and transplants.


Asunto(s)
Consenso , Sistema Nervioso , Organoides , Terminología como Asunto , Humanos , Modelos Biológicos , Sistema Nervioso/citología , Sistema Nervioso/patología , Organoides/citología , Organoides/patología , Células Madre Pluripotentes/citología
3.
Bioessays ; 45(9): e2300039, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37439444

RESUMEN

Heterogeneous nuclear ribonucleoprotein U (HNRNPU) is a nuclear protein that plays a crucial role in various biological functions, such as RNA splicing and chromatin organization. HNRNPU/scaffold attachment factor A (SAF-A) activities are essential for regulating gene expression, DNA replication, genome integrity, and mitotic fidelity. These functions are critical to ensure the robustness of developmental processes, particularly those involved in shaping the human brain. As a result, HNRNPU is associated with various neurodevelopmental disorders (HNRNPU-related neurodevelopmental disorder, HNRNPU-NDD) characterized by developmental delay and intellectual disability. Our research demonstrates that the loss of HNRNPU function results in the death of both neural progenitor cells and post-mitotic neurons, with a higher sensitivity observed in the former. We reported that HNRNPU truncation leads to the dysregulation of gene expression and alternative splicing of genes that converge on several signaling pathways, some of which are likely to be involved in the pathology of HNRNPU-related NDD.


Asunto(s)
Trastornos del Neurodesarrollo , Humanos , Trastornos del Neurodesarrollo/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo U/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo U/metabolismo , Encéfalo/metabolismo , Genoma , Empalme del ARN
4.
Mol Psychiatry ; 26(5): 1535-1550, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-31740755

RESUMEN

The broad impairments in cognitive and neurologic functioning found in Autism Spectrum Disorder (ASD) patients are thought to originate during early prenatal developmental stages. Indeed, postmortem and imaging studies in ASD patients detected white-matter abnormalities, as well as prefrontal and temporal cortex deficits, evident from early childhood. Here, we used Maternal Immune Activation (MIA), a mouse model for ASD, in which the offsprings exhibit Autistic-like behaviors as well as cortical abnormalities. However, the dynamics that influence the number and the identity of newly born cortical neurons following maternal inflammation remains unknown. Our study shows early changes in the duration of the S-phase of PAX6+ progenitors, leading to an increased proportion of neurogenic divisions and a reciprocal decrease in the proliferative divisions. In two different time points of maternal inflammation, MIA resulted in an overproduction of CTIP2+ cortical neurons, which remained overrepresented at the end of gestation and in postnatal mice. Interestingly, MIA-resistant IL6-KO mice did not exhibit these changes. Lastly, we propose that elevated levels of the transcription factor PAX6 following MIA supports the overproduction of CTIP2+ neurons. Taken together, our data reveals a possible link between maternal immune activation and the excess of cortical neurons found in the cortex of ASD patients.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Efectos Tardíos de la Exposición Prenatal , Animales , Preescolar , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación , Ratones , Neuronas , Embarazo
5.
Mol Psychiatry ; 26(3): 725-735, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33154567

RESUMEN

In recent years, it has been revealed that Parkinson's disease pathology may begin to manifest in the gastrointestinal track at a much earlier time point than in the brain. This paradigm shift has been suggested following evidence in humans that has been reproduced in animal models. Since rodent models cannot recapitulate many of the human disease features, human induced pluripotent stem cells derived from Parkinson's patients have been used to generate brain organoids, greatly contributing to our understanding of the disease pathophysiology. To understand the multifaced aspects of Parkinson's disease, it may be desirable to expand the complexity of these models, to include different brain regions, vasculature, immune cells as well as additional diverse organ-specific organoids such as gut and intestine. Furthermore, the contribution of gut microbiota to disease progression cannot be underestimated. Recent biotechnological advances propose that such combinations may be feasible. Here we discuss how this need can be met and propose that additional brain diseases can benefit from this approach.


Asunto(s)
Microbioma Gastrointestinal , Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Animales , Humanos , Técnicas de Cultivo de Órganos , Organoides
6.
Hum Mol Genet ; 26(9): 1678, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334871

RESUMEN

Mutations in the depalmitoylation enzyme, palmitoyl protein thioesterase (PPT1), result in the early onset neurodegenerative disease known as Infantile Neuronal Ceroid Lipofuscinosis. Here, we provide proteomic evidence suggesting that PPT1 deficiency could be considered as a ciliopathy. Analysis of membrane proteins from brain enriched for acylated proteins from neonate Ppt1 knock out and control mice revealed a list of 88 proteins with differential expression levels. Amongst them, we identified Rab3IP, which regulates ciliogenesis in concert with Rab8 and Rab11. Immunostaining analysis revealed that PPT1 is localized in the cilia. Indeed, an unbiased proteomics analysis on isolated cilia revealed 660 proteins, which differed in their abundance levels between wild type and Ppt1 knock out. We demonstrate here that Rab3IP, Rab8 and Rab11 are palmitoylated, and that palmitoylation of Rab11 is required for correct intracellular localization. Cells and brain preparations from Ppt1-/- mice exhibited fewer cells with cilia and abnormally longer cilia, with both acetylated tubulin and Rab3IP wrongly distributed along the length of cilia. Most importantly, the analysis revealed a difference in the distribution and levels of the modified proteins in cilia in the retina of mutant mice versus the wildtype, which may be important in the early neurodegenerative phenotype. Overall, our results suggest a novel link between palmitoylated proteins, cilial organization and the pathophysiology of Neuronal Ceroid Lipofuscinosis.


Asunto(s)
Proteínas de la Membrana/fisiología , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/metabolismo , Tioléster Hidrolasas/genética , Tioléster Hidrolasas/metabolismo , Animales , Encéfalo/metabolismo , Cilios/metabolismo , Cilios/patología , Células HEK293 , Humanos , Lipoilación , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Mutación , Células 3T3 NIH , Neuronas/metabolismo , Proteómica/métodos , Retina/metabolismo , Tioléster Hidrolasas/deficiencia
7.
Cereb Cortex ; 28(9): 3115-3128, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28981589

RESUMEN

The evolutionarily conserved Notch pathway plays an important role in regulation of stem cell renewal and cell fate determination in numerous organs, and as such is a key pathway in normal health and disease processes. Canonical Notch signaling is usually activated by cell contact where transmembrane ligands such as Delta-like and Jagged bind to Notch receptors. Notch activation results in the translocation of the cleaved Notch intracellular domain (NICD) into the nucleus and subsequent activation of transcription. Poly-ubiquitination leading to proteosome degradation of pathway components is one mean of regulating the Notch pathway. Here, we identified that Shootin1 exhibits the surprising propensity of activating the pathway either by interacting with LNX1/2 and promoting poly-ubiquitination of Numb or by complexing with Itch and impairing poly-ubiquitination of NICD. Within the developing brain Shootin1 modulates neuroblasts cell fate by executing 2 opposing activities on ubiquitin ligases, which control Notch signaling on 2 different levels.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Diferenciación Celular/fisiología , Activación Enzimática/fisiología , Ratones , Ratones Noqueados , Células-Madre Neurales/metabolismo
8.
J Neurochem ; 146(5): 500-525, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29570795

RESUMEN

The cerebral cortex is a highly organized structure whose development depends on diverse progenitor cell types, namely apical radial glia, intermediate progenitors, and basal radial glia cells, which are responsible for the production of the correct neuronal output. In recent years, these progenitor cell types have been deeply studied, particularly basal radial glia and their role in cortical expansion and gyrification. We review here a broad series of factors that regulate progenitor behavior and daughter cell fate. We first describe the different neuronal progenitor types, emphasizing the differences between lissencephalic and gyrencephalic species. We then review key factors shown to influence progenitor proliferation versus differentiation, discussing their roles in progenitor dynamics, neuronal production, and potentially brain size and complexity. Although spindle orientation has been considered a critical factor for mode of division and daughter cell output, we discuss other features that are emerging as crucial for these processes such as organelle and cell cycle dynamics. Additionally, we highlight the importance of adhesion molecules and the polarity complex for correct cortical development. Finally, we briefly discuss studies assessing progenitor multipotency and its possible contribution to the production of specific neuronal populations. This review hence summarizes recent aspects of cortical progenitor cell biology, and pinpoints emerging features critical for their behavior.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Corteza Cerebral/citología , Células Madre/fisiología , Animales , Humanos
9.
J Neurosci ; 35(3): 936-42, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25609612

RESUMEN

Spinal muscular atrophy with pontocerebellar hypoplasia (SMA-PCH) is an infantile SMA variant with additional manifestations, particularly severe microcephaly. We previously identified a nonsense mutation in Vaccinia-related kinase 1 (VRK1), R358X, as a cause of SMA-PCH. VRK1-R358X is a rare founder mutation in Ashkenazi Jews, and additional mutations in patients of different origins have recently been identified. VRK1 is a nuclear serine/threonine protein kinase known to play multiple roles in cellular proliferation, cell cycle regulation, and carcinogenesis. However, VRK1 was not known to have neuronal functions before its identification as a gene mutated in SMA-PCH. Here we show that VRK1-R358X homozygosity results in lack of VRK1 protein, and demonstrate a role for VRK1 in neuronal migration and neuronal stem cell proliferation. Using shRNA in utero electroporation in mice, we show that Vrk1 knockdown significantly impairs cortical neuronal migration, and affects the cell cycle of neuronal progenitors. Expression of wild-type human VRK1 rescues both proliferation and migration phenotypes. However, kinase-dead human VRK1 rescues only the migration impairment, suggesting the role of VRK1 in neuronal migration is partly noncatalytic. Furthermore, we found that VRK1 deficiency in human and mouse leads to downregulation of amyloid-ß precursor protein (APP), a known neuronal migration gene. APP overexpression rescues the phenotype caused by Vrk1 knockdown, suggesting that VRK1 affects neuronal migration through an APP-dependent mechanism.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Movimiento Celular/genética , Cerebelo/anomalías , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Neuronas/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Atrofias Musculares Espinales de la Infancia/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Ciclo Celular/genética , Línea Celular Tumoral , Enfermedades Cerebelosas/genética , Enfermedades Cerebelosas/metabolismo , Enfermedades Cerebelosas/patología , Cerebelo/metabolismo , Cerebelo/patología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Imagen por Resonancia Magnética , Ratones , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/patología , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Atrofias Musculares Espinales de la Infancia/genética , Atrofias Musculares Espinales de la Infancia/patología
10.
J Neurochem ; 136(3): 440-56, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26485324

RESUMEN

Autism spectrum disorders (ASD) encompass a group of neurodevelopmental diseases that demonstrate strong heritability, however, the inheritance is not simple and many genes have been associated with these disorders. ASD is regarded as a neurodevelopmental disorder, and abnormalities at different developmental stages are part of the disease etiology. This review provides a general background on neuronal migration during brain development and discusses recent advancements in the field connecting ASD and aberrant neuronal migration. We propose that neuronal migration impairment may be an important common pathophysiology in autism spectrum disorders (ASD). This review provides a general background on neuronal migration during brain development and discusses recent advancements in the field connecting ASD and aberrant neuronal migration.


Asunto(s)
Trastorno del Espectro Autista/patología , Encéfalo/patología , Movimiento Celular/fisiología , Neuronas/fisiología , Trastorno del Espectro Autista/genética , Humanos , Neurogénesis/genética
11.
Dev Growth Differ ; 58(5): 481-91, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27111774

RESUMEN

The opportunity to model autism spectrum disorders (ASD) through generation of patient-derived induced pluripotent stem cells (iPSCs) is currently an emerging topic. Wide-scale research of altered brain circuits in syndromic ASD, including Rett Syndrome, Fragile X Syndrome, Angelman's Syndrome and sporadic Schizophrenia, was made possible through animal models. However, possibly due to species differences, and to the possible contribution of epigenetics in the pathophysiology of these diseases, animal models fail to recapitulate many aspects of ASD. With the advent of iPSCs technology, 3D cultures of patient-derived cells are being used to study complex neuronal phenotypes, including both syndromic and nonsyndromic ASD. Here, we review recent advances in using iPSCs to study various aspects of the ASD neuropathology, with emphasis on the efforts to create in vitro model systems for syndromic and nonsyndromic ASD. We summarize the main cellular activity phenotypes and aberrant genetic interaction networks that were found in iPSC-derived neurons of syndromic and nonsyndromic autistic patients.


Asunto(s)
Trastorno del Espectro Autista/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Animales , Trastorno del Espectro Autista/patología , Humanos , Células Madre Pluripotentes Inducidas/patología
12.
Stem Cells ; 32(10): 2657-67, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24965231

RESUMEN

Emergence of genomic instability is a practical issue in preparing neural stem cells (NSCs) and induced pluripotent stem cells (iPSCs). However, it is still not fully understood what the origins and mechanisms for formation are for the genomic alternations observed. Here, we studied the extent of genomic variation on the scale of individual cells originating from the same animal. We used mouse NSCs grown from embryonic cells and iPSCs generated from embryonic brain cells, B cells or fibroblasts, and performed comparative analysis with cultures of fibroblasts from the same mouse. In the first passage of these cell lines, aneuploidies were only observed for chromosomes 6, 11, 12, 19, and Y, which is overall at a rate lower than previously reported; de novo copy number variations (CNVs) were observed in 4.3% of neural iPSCs, 29% of B cell iPSCs, 10% of fibroblast iPSCs, and 1.3% of neurospheres. In contrast, propagation of these first passage cells to a later passage induced additional aneuploidies and CNVs. Breakpoint sequencing analysis suggested that the majority of the detected CNVs arose by replicative mechanisms. Interestingly, we detected identical de novo CNVs in different single cell colonies that appeared to have arisen independently from each other, which suggests a novel CNV formation mechanism in these cells. Our findings provide insights into mechanisms of CNV formation during reprogramming and suggest that replicative mechanisms for CNV formation accompany mitotic divisions.


Asunto(s)
Variación Estructural del Genoma , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Aneuploidia , Animales , Secuencia de Bases , Proliferación Celular , Rotura Cromosómica , Células Clonales , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN/genética , Sitios Genéticos , Inestabilidad Genómica , Ratones , Datos de Secuencia Molecular , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/metabolismo
13.
J Neurosci ; 33(29): 11932-48, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23864681

RESUMEN

Shootin1 has been ascribed a role in regulating polarization of primary hippocampal neurons. To better understand the possible role of Shootin1 in the developing brain, we identified a member of the kinesin superfamily, KIF20B, as a novel Shootin1 interacting protein and a potential mediator of Shootin1 interaction with microtubules. KIF20B/Shootin1 binding was mapped to a 57 aa KIF20B sequence, which was used as a dominant-negative fragment. Direct interaction between that peptide (MBD) and Shootin1 was confirmed by surface plasmon resonance-based technology and the affinity was determined in the 10⁻7 m range. The proteins are expressed in the developing brain and formed a complex in vivo based on coimmunoprecipitation experiments and coimmunostaining in primary neurons. In primary hippocampal neurons Kif20b knockdown reduced Shootin1 mobilization to the developing axon, as evidenced by immunostaining and fluorescence recovery after photobleaching analysis, suggesting that Shootin1 is a novel KIF20B cargo. shRNA targeting of Shootin1 reduced PIP3 accumulation in the growth cone, as did Kif20b shRNA. In the developing mouse brain, Kif20b knockdown or expression of the KIF20B minimal binding domain inhibited neuronal migration, and in vivo migration assays suggested that Shootin1/Kif20b acts in the same genetic pathway. Time-lapse imaging of multipolar cells in the subventricular zone revealed that downregulating levels of either Shootin1 or Kif20b hindered the transition from multipolar to bipolar cells. Collectively, our data demonstrate the importance of the Shootin1/KIF20B interaction to the dynamic process of pyramidal neuronal polarization and migration.


Asunto(s)
Movimiento Celular/fisiología , Polaridad Celular/fisiología , Hipocampo/metabolismo , Cinesinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Animales , Axones/metabolismo , Regulación del Desarrollo de la Expresión Génica , Conos de Crecimiento/metabolismo , Cinesinas/genética , Ratones , Proteínas del Tejido Nervioso/genética
14.
Hum Mol Genet ; 21(8): 1681-92, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22194194

RESUMEN

Microdeletions encompassing the MAPT (Tau) locus resulting in intellectual disability raised the hypothesis that Tau may regulate early functions in the developing brain. Our results indicate that neuronal migration was inhibited in mouse brains following Tau reduction. In addition, the leading edge of radially migrating neurons was aberrant in spite of normal morphology of radial glia. Furthermore, intracellular mitochondrial transport and morphology were affected. In early postnatal brains, a portion of Tau knocked down neurons reached the cortical plate. Nevertheless, they exhibited far less developed dendrites and a striking reduction in connectivity evident by the size of boutons. Our novel results strongly implicate MAPT as a dosage-sensitive gene in this locus involved in intellectual disability. Furthermore, our results are likely to impact our understanding of other diseases involving Tau.


Asunto(s)
Encéfalo/embriología , Discapacidad Intelectual/genética , Neuronas/fisiología , Proteínas tau/genética , Proteínas tau/metabolismo , Animales , Axones/ultraestructura , Encéfalo/citología , Encéfalo/metabolismo , Movimiento Celular , Forma de la Célula , Células Cultivadas , Dendritas/ultraestructura , Electroporación , Embrión de Mamíferos , Desarrollo Embrionario , Técnicas de Silenciamiento del Gen , Discapacidad Intelectual/metabolismo , Ratones , Mitocondrias/ultraestructura , Neuroglía/ultraestructura , Neuronas/citología , Neuronas/ultraestructura , Terminales Presinápticos/ultraestructura , ARN Interferente Pequeño
15.
EMBO J ; 29(1): 107-19, 2010 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19927128

RESUMEN

Regulated activity of the retrograde molecular motor, cytoplasmic dynein, is crucial for multiple biological activities, and failure to regulate this activity can result in neuronal migration retardation or neuronal degeneration. The activity of dynein is controlled by the LIS1-Ndel1-Nde1 protein complex that participates in intracellular transport, mitosis, and neuronal migration. These biological processes are subject to tight multilevel modes of regulation. Palmitoylation is a reversible posttranslational lipid modification, which can dynamically regulate protein trafficking. We found that both Ndel1 and Nde1 undergo palmitoylation in vivo and in transfected cells by specific palmitoylation enzymes. Unpalmitoylated Ndel1 interacts better with dynein, whereas the interaction between Nde1 and cytoplasmic dynein is unaffected by palmitoylation. Furthermore, palmitoylated Ndel1 reduced cytoplasmic dynein activity as judged by Golgi distribution, VSVG and short microtubule trafficking, transport of endogenous Ndel1 and LIS1 from neurite tips to the cell body, retrograde trafficking of dynein puncta, and neuronal migration. Our findings indicate, to the best of our knowledge, for the first time that Ndel1 palmitoylation is a new mean for fine-tuning the activity of the retrograde motor cytoplasmic dynein.


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Dineínas/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa/química , 1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Células COS , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Chlorocebus aethiops , Citoplasma/metabolismo , Femenino , Aparato de Golgi/metabolismo , Humanos , Técnicas In Vitro , Lipoilación , Ratones , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Complejos Multiproteicos , Neuronas/metabolismo , Embarazo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Transfección
16.
Cell Mol Life Sci ; 70(7): 1255-68, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23117601

RESUMEN

Microtubules are known to drive chromosome movements and to induce nuclear envelope breakdown during mitosis and meiosis. Here we show that microtubules can enforce nuclear envelope folding and alter the levels of nuclear envelope-associated heterochromatin during interphase, when the nuclear envelope is intact. Microtubule reassembly, after chemically induced depolymerization led to folding of the nuclear envelope and to a transient accumulation of condensed chromatin at the site nearest the microtubule organizing center (MTOC). This microtubule-dependent chromatin accumulation next to the MTOC is dependent on the composition of the nuclear lamina and the activity of the dynein motor protein. We suggest that forces originating from simultaneous polymerization of microtubule fibers deform the nuclear membrane and the underlying lamina. Whereas dynein motor complexes localized to the nuclear envelope that slide along the microtubules transfer forces and/or signals into the nucleus to induce chromatin reorganization and accumulation at the nuclear membrane folds. Thus, our study identified a molecular mechanism by which mechanical forces generated in the cytoplasm reshape the nuclear envelope, alter the intranuclear organization of chromatin, and affect the architecture of the interphase nucleus.


Asunto(s)
Núcleo Celular/metabolismo , Interfase/fisiología , Microtúbulos/metabolismo , Multimerización de Proteína , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Cromatina/efectos de los fármacos , Cromatina/metabolismo , Interfase/efectos de los fármacos , Cinética , Ratones , Microscopía por Video , Centro Organizador de los Microtúbulos/efectos de los fármacos , Centro Organizador de los Microtúbulos/metabolismo , Centro Organizador de los Microtúbulos/ultraestructura , Microtúbulos/efectos de los fármacos , Microtúbulos/ultraestructura , Nocodazol/farmacología , Membrana Nuclear/efectos de los fármacos , Membrana Nuclear/metabolismo , Membrana Nuclear/ultraestructura , Multimerización de Proteína/efectos de los fármacos , Multimerización de Proteína/fisiología , Moduladores de Tubulina/farmacología , Células Tumorales Cultivadas
17.
Adv Exp Med Biol ; 800: 97-111, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24243102

RESUMEN

Proper brain development requires the orchestrated migration of neurons from their place of birth to their final positioning, where they will form appropriate connections with their target cells. These events require coordinated activity of multiple elements of the cytoskeleton, in which the MARK/Par-1 polarity kinase plays an important role. Here, the various roles and modes of regulation of MARK/Par-1 are reviewed. MARK/Par-1 participates in axon formation in primary hippocampal neurons. Balanced levels of MARK/Par-1 are required for proper radial migration, as well as for migration in the rostral migratory stream. Normal neuronal migration requires at least two of MARK/Par-1 substrates, DCX and tau. Overall, the positioning of MARK/Par-1 at the crosstalk of regulating cytoskeletal dynamics allows its participation in neuronal polarity decisions.


Asunto(s)
Axones/enzimología , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Citoesqueleto/enzimología , Hipocampo/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas tau/genética , Proteínas tau/metabolismo
18.
Oxf Open Neurosci ; 3: kvae001, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38595939

RESUMEN

PRDM16 is a dynamic transcriptional regulator of various stem cell niches, including adipocytic, hematopoietic, cardiac progenitors, and neural stem cells. PRDM16 has been suggested to contribute to 1p36 deletion syndrome, one of the most prevalent subtelomeric microdeletion syndromes. We report a patient with a de novo nonsense mutation in the PRDM16 coding sequence, accompanied by lissencephaly and microcephaly features. Human stem cells were genetically modified to mimic this mutation, generating cortical organoids that exhibited altered cell cycle dynamics. RNA sequencing of cortical organoids at day 32 unveiled changes in cell adhesion and WNT-signaling pathways. ChIP-seq of PRDM16 identified binding sites in postmortem human fetal cortex, indicating the conservation of PRDM16 binding to developmental genes in mice and humans, potentially at enhancer sites. A shared motif between PRDM16 and LHX2 was identified and further examined through comparison with LHX2 ChIP-seq data from mice. These results suggested a collaborative partnership between PRDM16 and LHX2 in regulating a common set of genes and pathways in cortical radial glia cells, possibly via their synergistic involvement in cortical development.

19.
J Neurosci ; 32(50): 18204-14, 2012 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-23238734

RESUMEN

Amyloid-ß peptide (Aß) is believed to play a central role in the pathogenesis of Alzheimer's disease. In view of the side effects associated with inhibiting the secretases that produce Aß, new molecular targets are required to provide alternative therapeutic options. We used RNA interference (RNAi) to systematically screen the Drosophila genome to identify genes that modulate Aß production upon knockdown. RNAi of 41 genes in Drosophila cells significantly lowered Aß without affecting general secretion or viability. After the γ-secretase complex components, the most potent effect was observed for platelet activating factor acetylhydrolase α (Paf-AHα), and, in mammalian cells, the effect was replicated for its ortholog PAFAH1B2. Knockdown of PAFAH1B2 strongly reduced Aß secretion from human cells, and this effect was confirmed in primary cells derived from PAFAH1B2 knock-out mice. Reduced Aß production was not attributable to altered ß-amyloid precursor protein (APP) ectodomain shedding but was a result of an enhanced degradation of APP C-terminal fragments (CTFs) in the absence of PAFAH1B2 but not its close homolog PAFAH1B3. Enhanced degradation of APP CTFs was selective because no such effects were obtained for Notch or E-/N-cadherin. Thus, we have identified an important protein that can selectively modify Aß generation via a novel mechanism, namely enhanced degradation of its immediate precursor. In view of the absence of a neurological phenotype in PAFAH1B2 knock-out mice, targeted downregulation of PAFAH1B2 may be a promising new strategy for lowering Aß.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa/genética , Animales , Drosophila , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Fragmentos de Péptidos/metabolismo , Interferencia de ARN , Transfección
20.
J Cell Sci ; 124(Pt 23): 3989-4000, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22159412

RESUMEN

Completion of mitosis requires microtubule-dependent transport of membranes to the midbody. Here, we identified a role in cytokinesis for doublecortin domain-containing protein 5 (DCDC5), a member of the doublecortin protein superfamily. DCDC5 is a microtubule-associated protein expressed in both specific and dynamic fashions during mitosis. We show that DCDC5 interacts with cytoplasmic dynein and Rab8 (also known as Ras-related protein Rab-8A), as well as with the Rab8 nucleotide exchange factor Rabin8 (also known as Rab-3A-interacting protein). Following DCDC5 knockdown, the durations of the metaphase to anaphase transition and cytokinesis, and the proportion of multinucleated cells increases, whereas cell viability decreases. Furthermore, knockdown of DCDC5 or addition of a dynein inhibitor impairs the entry of Golgi-complex-derived Rab8-positive vesicles to the midbody. These findings suggest that DCDC5 plays an important role in mediating dynein-dependent transport of Rab8-positive vesicles and in coordinating late cytokinesis.


Asunto(s)
Citocinesis , Dineínas Citoplasmáticas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Apoptosis , Supervivencia Celular , Clonación Molecular , Dineínas Citoplasmáticas/genética , Proteínas de Dominio Doblecortina , Técnicas de Silenciamiento del Gen , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Células HeLa , Humanos , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Mitosis , Neuropéptidos/genética , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , Transfección , Vesículas Transportadoras/metabolismo , Proteínas de Unión al GTP rab/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA