Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Oxf Open Neurosci ; 3: kvae007, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38638145

RESUMEN

Bipolar disorder (BD) is a severe mental illness that can result from neurodevelopmental aberrations, particularly in familial BD, which may include causative genetic variants. In the present study, we derived cortical organoids from BD patients and healthy (control) individuals from a clinically dense family in the Indian population. Our data reveal that the patient organoids show neurodevelopmental anomalies, including organisational, proliferation and migration defects. The BD organoids show a reduction in both the number of neuroepithelial buds/cortical rosettes and the ventricular zone size. Additionally, patient organoids show a lower number of SOX2-positive and EdU-positive cycling progenitors, suggesting a progenitor proliferation defect. Further, the patient neurons show abnormal positioning in the ventricular/intermediate zone of the neuroepithelial bud. Transcriptomic analysis of control and patient organoids supports our cellular topology data and reveals dysregulation of genes crucial for progenitor proliferation and neuronal migration. Lastly, time-lapse imaging of neural stem cells in 2D in vitro cultures reveals abnormal cellular migration in BD samples. Overall, our study pinpoints a cellular and molecular deficit in BD patient-derived organoids and neural stem cell cultures.

2.
Stem Cells ; 42(2): 128-145, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38152966

RESUMEN

Neurogenesis begins with neural stem cells undergoing symmetric proliferative divisions to expand and then switching to asymmetric differentiative divisions to generate neurons in the developing brain. Chromatin regulation plays a critical role in this switch. Histone lysine-specific demethylase LSD1 demethylates H3K4me1/2 and H3K9me1/2 but the mechanisms of its global regulatory functions in human neuronal development remain unclear. We performed genome-wide ChIP-seq of LSD1 occupancy, RNA-seq, and Histone ChIP-seq upon LSD1 inhibition to identify its repressive role in human neural stem cells. Novel downstream effectors of LSD1 were identified, including the Notch signaling pathway genes and human-neural progenitor-enriched extracellular matrix (ECM) pathway/cell adhesion genes, which were upregulated upon LSD1 inhibition. LSD1 inhibition led to decreased neurogenesis, and overexpression of downstream effectors mimicked this effect. Histone ChIP-seq analysis revealed that active and enhancer markers H3K4me2, H3K4me1, and H3K9me1 were upregulated upon LSD1 inhibition, while the repressive H3K9me2 mark remained mostly unchanged. Our work identifies the human-neural progenitor-enriched ECM pathway/cell adhesion genes and Notch signaling pathway genes as novel downstream effectors of LSD1, regulating neuronal differentiation in human neural stem cells.


Asunto(s)
Histonas , Células-Madre Neurales , Humanos , Adhesión Celular/genética , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histonas/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/genética
3.
PLoS Genet ; 19(8): e1010874, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37594984

RESUMEN

In the mammalian cerebral cortex, the hippocampal primordium (Hcp) occupies a discrete position in the dorsal telencephalic neuroepithelium adjacent to the neocortical primordium (Ncp). We examined transcriptomic and chromatin-level features that distinguish the Hcp from the Ncp in the mouse during the early neurogenic period, embryonic day (E)12.5. ATAC-seq revealed that the Hcp was more accessible than the Ncp at this stage. Motif analysis of the differentially accessible loci in these tissues revealed LHX2 as a candidate transcription factor for modulating gene regulatory networks (GRNs). We analyzed LHX2 occupancy profiles and compared these with transcriptomic data from control and Lhx2 mutant Hcp and Ncp at E12.5. Our results revealed that LHX2 directly regulates distinct genes in the Hcp and Ncp within a set of common pathways that control fundamental aspects of development namely pluripotency, axon pathfinding, Wnt, and Hippo signaling. Loss of Lhx2 caused a decrease in accessibility, specifically in hippocampal chromatin, suggesting that this factor may play a unique role in hippocampal development. We identified 14 genes that were preferentially enriched in the Hcp, for which LHX2 regulates both chromatin accessibility and mRNA expression, which have not thus far been examined in hippocampal development. Together, these results provide mechanistic insight into how LHX2 function in the Hcp may contribute to the process by which the hippocampus acquires features distinct from the neocortex.


Asunto(s)
Cromatina , Neocórtex , Animales , Ratones , Hipocampo , Proteínas con Homeodominio LIM , Mamíferos , Factores de Transcripción , Transcriptoma
4.
Neurochem Int ; 147: 105055, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33964373

RESUMEN

The diverse number of neurons in the cerebral cortex are generated during development by neural stem cells lining the ventricle, and they continue maturing postnatally. Dynamic chromatin regulation in these neural stem cells is a fundamental determinant of the emerging property of the functional neural network, and the chromatin remodellers are critical determinants of this process. Chromatin remodellers participate in several steps of this process from proliferation, differentiation, migration leading to complex network formation which forms the basis of higher-order functions of cognition and behaviour. Here we review the role of these ATP-dependent chromatin remodellers in cortical development in health and disease and highlight several key mouse mutants of the subunits of the complexes which have revealed how the remodelling mechanisms control the cortical stem cell chromatin landscape for expression of stage-specific transcripts. Consistent with their role in cortical development, several putative risk variants in the subunits of the remodelling complexes have been identified as the underlying causes of several neurodevelopmental disorders. A basic understanding of the detailed molecular mechanism of their action is key to understating how mutations in the same networks lead to disease pathologies and perhaps pave the way for therapeutic development for these complex multifactorial disorders.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Ensamble y Desensamble de Cromatina/genética , Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Trastornos del Neurodesarrollo/genética , Animales , Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Epigénesis Genética/genética , Humanos
5.
Development ; 147(20)2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-32994168

RESUMEN

The protein co-factor Ldb1 regulates cell fate specification by interacting with LIM-homeodomain (LIM-HD) proteins in a tetrameric complex consisting of an LDB:LDB dimer that bridges two LIM-HD molecules, a mechanism first demonstrated in the Drosophila wing disc. Here, we demonstrate conservation of this interaction in the regulation of mammalian hippocampal development, which is profoundly defective upon loss of either Lhx2 or Ldb1 Electroporation of a chimeric construct that encodes the Lhx2-HD and Ldb1-DD (dimerization domain) in a single transcript cell-autonomously rescues a comprehensive range of hippocampal deficits in the mouse Ldb1 mutant, including the acquisition of field-specific molecular identity and the regulation of the neuron-glia cell fate switch. This demonstrates that the LHX:LDB complex is an evolutionarily conserved molecular regulatory device that controls complex aspects of regional cell identity in the developing brain.


Asunto(s)
Linaje de la Célula , Secuencia Conservada , Proteínas de Unión al ADN/genética , Evolución Molecular , Hipocampo/citología , Proteínas con Dominio LIM/genética , Proteínas con Homeodominio LIM/genética , Factores de Transcripción/genética , Animales , Tipificación del Cuerpo , Proteínas de Unión al ADN/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Ratones , Mutación/genética , Neurogénesis , Neuroglía/citología , Neuroglía/metabolismo , Unión Proteica , Factores de Transcripción/metabolismo
6.
Int J Dev Biol ; 64(1-2-3): 123-132, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32659000

RESUMEN

The brain is the seat of all higher-order functions in the body. Brain development and the vast array of neurons and glia it produces is a baffling mystery to be studied. Neuroscientists using a vast number of model systems have been able to crack many of the nitty-gritty details using various model systems. One way has been to size down the problem by utilizing the power of genetics using simple model systems such as Drosophila to create a fundamental framework in order to unravel the basic principles of brain development. Scientists have used simpler organisms to uncover the fundamental principles of brain development and also to study the evo-devo angle to brain development. Complex circuitry has been unraveled in complex model systems, such as the mouse, to reveal the intricacies and regional specialization of brain function. This is an ever-growing field, and with newer genetic and molecular tools, together with several new centers of excellence, India's contribution to this fascinating field of study is continually rising. Here, I review the pioneering work done by Indian developmental neurobiologists in the past and their mounting contribution in the present.


Asunto(s)
Evolución Biológica , Encéfalo/embriología , Biología Evolutiva/historia , Biología Evolutiva/tendencias , Drosophila/embriología , Modelos Biológicos , Animales , Historia del Siglo XX , Historia del Siglo XXI
7.
Neuron ; 104(5): 827-829, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31805259

RESUMEN

The (GGGGCC)n repeat expansion in C9orf72, which is the most common cause of frontotemporal dementia and amyotrophic lateral sclerosis, is translated through repeat-associated non-AUG (RAN) translation. In this issue of Neuron, Cheng et al. (2019) report that the helicase DDX3X, which unwinds (or relaxes) RNA, suppresses RAN translation and toxicity.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Demencia Frontotemporal/genética , Proteína C9orf72 , Sistemas CRISPR-Cas , ARN Helicasas DEAD-box , Expansión de las Repeticiones de ADN , Humanos , ARN Helicasas
8.
Brain ; 141(12): 3428-3442, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30496365

RESUMEN

Mutations in the endosome-associated protein CHMP2B cause frontotemporal dementia and lead to lysosomal storage pathology in neurons. We here report that physiological levels of mutant CHMP2B causes reduced numbers and significantly impaired trafficking of endolysosomes within neuronal dendrites, accompanied by increased dendritic branching. Mechanistically, this is due to the stable incorporation of mutant CHMP2B onto neuronal endolysosomes, which we show renders them unable to traffic within dendrites. This defect is due to the inability of mutant CHMP2B to recruit the ATPase VPS4, which is required for release of CHMP2B from endosomal membranes. Strikingly, both impaired trafficking and the increased dendritic branching were rescued by treatment with antisense oligonucleotides targeting the well validated frontotemporal dementia risk factor TMEM106B, which encodes an endolysosomal protein. This indicates that reducing TMEM106B levels can restore endosomal health in frontotemporal dementia. As TMEM106B is a risk factor for frontotemporal dementia caused by both C9orf72 and progranulin mutations, and antisense oligonucleotides are showing promise as therapeutics for neurodegenerative diseases, our data suggests a potential new strategy for treating the wide range of frontotemporal dementias associated with endolysosomal dysfunction.


Asunto(s)
Dendritas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Endosomas/metabolismo , Demencia Frontotemporal/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Femenino , Técnicas de Silenciamiento del Gen , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal
9.
J Exp Neurosci ; 12: 1179069518767404, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29760561

RESUMEN

We established an efficient cell culture assay that permits combinatorial genetic perturbations in hippocampal progenitors to examine cell-autonomous mechanisms of fate specification. The procedure begins with ex vivo electroporation of isolated, intact embryonic brains, in a manner similar to in utero electroporation but with greatly improved access and targeting. The electroporated region is then dissected and transiently maintained in organotypic explant culture, followed by dissociation and plating of cells on coverslips for in vitro culture. This assay recapitulates data obtained in vivo with respect to the neuron-glia cell fate switch and can be effectively used to test intrinsic or extrinsic factors that regulate this process. The advantages of this ex vivo procedure over in utero electroporation include the fact that distinct combinations of perturbative reagents can be introduced in different embryos from a single litter, and issues related to embryonic lethality of transgenic animals can be circumvented.

10.
J Neurosci ; 37(46): 11245-11254, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29025924

RESUMEN

Regulation of the neuron-glia cell-fate switch is a critical step in the development of the CNS. Previously, we demonstrated that Lhx2 is a necessary and sufficient regulator of this process in the mouse hippocampal primordium, such that Lhx2 overexpression promotes neurogenesis and suppresses gliogenesis, whereas loss of Lhx2 has the opposite effect. We tested a series of transcription factors for their ability to mimic Lhx2 overexpression and suppress baseline gliogenesis, and also to compensate for loss of Lhx2 and suppress the resulting enhanced level of gliogenesis in the hippocampus. Here, we demonstrate a novel function of Dmrt5/Dmrta2 as a neurogenic factor in the developing hippocampus. We show that Dmrt5, as well as known neurogenic factors Neurog2 and Pax6, can each not only mimic Lhx2 overexpression, but also can compensate for loss of Lhx2 to different extents. We further uncover a reciprocal regulatory relationship between Dmrt5 and Lhx2, such that each can compensate for loss of the other. Dmrt5 and Lhx2 also have opposing regulatory control on Pax6 and Neurog2, indicating a complex bidirectionally regulated network that controls the neuron-glia cell-fate switch.SIGNIFICANCE STATEMENT We identify Dmrt5 as a novel regulator of the neuron-glia cell-fate switch in the developing hippocampus. We demonstrate Dmrt5 to be neurogenic, and reciprocally regulated by Lhx2: loss of either factor promotes gliogenesis; overexpression of either factor suppresses gliogenesis and promotes neurogenesis; each can substitute for loss of the other. Furthermore, each factor has opposing effects on established neurogenic genes Neurog2 and Pax6 Dmrt5 is known to suppress their expression, and we show that Lhx2 is required to maintain it. Our study reveals a complex regulatory network with bidirectional control of a fundamental feature of CNS development, the control of the production of neurons versus astroglia in the developing hippocampus.Finally, we confirm that Lhx2 binds a highly conserved putative enhancer of Dmrt5, suggesting an evolutionarily conserved regulatory relationship between these factors. Our findings uncover a complex network that involves Lhx2, Dmrt5, Neurog2, and Pax6, and that ensures the appropriate amount and timing of neurogenesis and gliogenesis in the developing hippocampus.


Asunto(s)
Hipocampo/fisiología , Proteínas con Homeodominio LIM/fisiología , Neurogénesis/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Hipocampo/citología , Hipocampo/embriología , Masculino , Ratones , Ratones Transgénicos , Embarazo
11.
J Neurosci ; 37(1): 194-203, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28053041

RESUMEN

In the developing cerebral cortex, sequential transcriptional programs take neuroepithelial cells from proliferating progenitors to differentiated neurons with unique molecular identities. The regulatory changes that occur in the chromatin of the progenitors are not well understood. During deep layer neurogenesis, we show that transcription factor LHX2 binds to distal regulatory elements of Fezf2 and Sox11, critical determinants of neuron subtype identity in the mouse neocortex. We demonstrate that LHX2 binds to the nucleosome remodeling and histone deacetylase histone remodeling complex subunits LSD1, HDAC2, and RBBP4, which are proximal regulators of the epigenetic state of chromatin. When LHX2 is absent, active histone marks at the Fezf2 and Sox11 loci are increased. Loss of LHX2 produces an increase, and overexpression of LHX2 causes a decrease, in layer 5 Fezf2 and CTIP2-expressing neurons. Our results provide mechanistic insight into how LHX2 acts as a necessary and sufficient regulator of genes that control cortical neuronal subtype identity. SIGNIFICANCE STATEMENT: The functional complexity of the cerebral cortex arises from an array of distinct neuronal subtypes with unique connectivity patterns that are produced from common progenitors. This study reveals that transcription factor LHX2 regulates the numbers of specific cortical output neuron subtypes by controlling the genes that are required to produce them. Loss or increase in LHX2 during neurogenesis is sufficient to increase or decrease, respectively, a particular subcerebrally projecting population. Mechanistically, LHX2 interacts with chromatin modifying protein complexes to edit the chromatin landscape of its targets Fezf2 and Sox11, which regulates their expression and consequently the identities of the neurons produced. Thus, LHX2 is a key component of the control network for producing neurons that will participate in cortical circuitry.


Asunto(s)
Corteza Cerebral/citología , Proteínas de Unión al ADN/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Factores de Transcripción SOXC/metabolismo , Factores de Transcripción/metabolismo , Animales , Corteza Cerebral/diagnóstico por imagen , Cromatina/genética , Epigénesis Genética , Femenino , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Masculino , Ratones , Nucleosomas/metabolismo , Embarazo
12.
Proc Natl Acad Sci U S A ; 110(50): E4913-21, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24262147

RESUMEN

LIM homeodomain transcription factors are critical regulators of early development in multiple systems but have yet to be examined for a role in circuit formation. The LIM homeobox gene Lhx2 is expressed in cortical progenitors during development and also in the superficial layers of the neocortex in maturity. However, analysis of Lhx2 function at later stages of cortical development has been hampered by severe phenotypes associated with early loss of function. We identified a particular Cre-recombinase line that acts in the cortical primordium after its specification is complete, permitting an analysis of Lhx2 function in neocortical lamination, regionalization, and circuit formation by selective elimination of Lhx2 in the dorsal telencephalon. We report a profound disruption of cortical neuroanatomical and molecular features upon loss of Lhx2 in the cortex from embryonic day 11.5. A unique feature of cortical circuitry, the somatosensory barrels, is undetectable, and molecular patterning of cortical regions appears disrupted. Surprisingly, thalamocortical afferents innervate the mutant cortex with apparently normal regional specificity. Electrophysiological recordings reveal a loss of responses evoked by stimulation of individual whiskers, but responses to simultaneous stimulation of multiple whiskers were present, suggesting that thalamic afferents are unable to organize the neurocircuitry for barrel formation because of a cortex-specific requirement of Lhx2. We report that Lhx2 is required for the expression of transcription factor paired box gene 6, axon guidance molecule Ephrin A5, and the receptor NMDA receptor 1. These genes may mediate Lhx2 function in the formation of specialized neurocircuitry necessary for neocortical function.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas con Homeodominio LIM/metabolismo , Corteza Somatosensorial/embriología , Factores de Transcripción/metabolismo , Animales , Inmunoprecipitación de Cromatina , Efrina-A5/metabolismo , Potenciales Evocados/fisiología , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Hibridación in Situ , Integrasas , Proteínas con Homeodominio LIM/deficiencia , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Vías Nerviosas/embriología , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Represoras/metabolismo , Corteza Somatosensorial/metabolismo , Factores de Transcripción/deficiencia
13.
Proc Natl Acad Sci U S A ; 108(27): E265-74, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21690374

RESUMEN

The sequential production of neurons and astrocytes from neuroepithelial precursors is a fundamental feature of central nervous system development. We report that LIM-homeodomain (LIM-HD) transcription factor Lhx2 regulates this transition in the developing hippocampus. Disrupting Lhx2 function in the embryonic hippocampus by in utero electroporation and in organotypic slice culture caused the premature production of astrocytes at stages when neurons are normally generated. Lhx2 function is therefore necessary to suppress astrogliogenesis during the neurogenic period. Furthermore, Lhx2 overexpression was sufficient to suppress astrogliogenesis and prolong the neurogenic period. We provide evidence that Lhx2 overexpression can counteract the instructive astrogliogenic effect of Notch activation. Lhx2 overexpression was also able to override and suppress the activation of the GFAP promoter by Nfia, a Notch-regulated transcription factor that is required for gliogenesis. Thus, Lhx2 appears to act as a "brake" on Notch/Nfia-mediated astrogliogenesis. This critical role for Lhx2 is spatially restricted to the hippocampus, because loss of Lhx2 function in the neocortex did not result in premature astrogliogenesis at the expense of neurogenesis. Our results therefore place Lhx2 as a central regulator of the neuron-glia cell fate decision in the hippocampus and reveal a striking regional specificity of this fundamental function within the dorsal telencephalon.


Asunto(s)
Hipocampo/embriología , Proteínas de Homeodominio/fisiología , Neurogénesis/fisiología , Factores de Transcripción/fisiología , Animales , Astrocitos/citología , Astrocitos/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Femenino , Hipocampo/citología , Hipocampo/fisiología , Proteínas de Homeodominio/genética , Proteínas con Homeodominio LIM , Ratones , Ratones Noqueados , Ratones Transgénicos , Factores de Transcripción NFI/fisiología , Neocórtex/citología , Neocórtex/embriología , Neocórtex/fisiología , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Neurogénesis/genética , Fenotipo , Embarazo , Receptores Notch/fisiología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
14.
J Biol Chem ; 286(16): 14146-56, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21357685

RESUMEN

Insulin is the key regulator of glucose homeostasis in mammals, and glucose-stimulated insulin biosynthesis is essential for maintaining glucose levels in a narrow range in mammals. Glucose specifically promotes the translation of insulin in pancreatic ß-islet, and the untranslated regions of insulin mRNA play a role in such regulation. Specific factors in the ß-islets bind to the insulin 5' UTR and regulate its translation. In the present study we identify protein-disulfide isomerase (PDI) as a key regulator of glucose-stimulated insulin biosynthesis. We show that both in vitro and in vivo PDI can specifically associate with the 5' UTR of insulin mRNA. Immunodepletion of PDI from the islet extract results in loss of glucose-stimulated translation indicating a critical role for PDI in insulin biosynthesis. Similarly, transient overexpression of PDI resulted in specific translation activation by glucose. We show that the RNA binding activity of PDI is mediated through PABP. PDI catalyzes the reduction of the PABP disulfide bond resulting in specific binding of PABP to the insulin 5' UTR. We also show that glucose stimulation of the islets results in activation of a specific kinase that can phosphorylate PDI. These findings identify PDI and PABP as important players in glucose homeostasis.


Asunto(s)
Regiones no Traducidas 5' , Regulación de la Expresión Génica , Glucosa/metabolismo , Células Secretoras de Insulina/citología , Proteína Disulfuro Isomerasas/química , Secuencia de Aminoácidos , Animales , Catálisis , Insulina/metabolismo , Ratones , Datos de Secuencia Molecular , Biosíntesis de Proteínas , Ratas , Ratas Wistar , Homología de Secuencia de Aminoácido
15.
FEBS Lett ; 584(6): 1169-73, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20153322

RESUMEN

Insulin is a secreted peptide that controls glucose homeostasis in mammals, and insulin biosynthesis is regulated by glucose at many levels. Rodent insulin is encoded by two non-allelic genes. We have identified a novel splice variant of the insulin2 gene in mice that constitutes about 75% of total insulin2 mRNA. The alternate splicing does not alter the ORF but reduces the 5'UTR by 12 bases. A reporter gene containing the novel short 5'UTR, is more efficiently expressed in cells, suggesting that alternative splicing of insulin mRNA in mice could result in an additional level of regulation in insulin biosynthesis.


Asunto(s)
Regulación de la Expresión Génica , Insulina/genética , Regiones no Traducidas 5'/genética , Animales , Secuencia de Bases , Células Cultivadas , Regulación de la Expresión Génica/genética , Humanos , Insulina/aislamiento & purificación , Insulina/metabolismo , Islotes Pancreáticos/química , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Polimorfismo Genético/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/aislamiento & purificación , Isoformas de Proteínas/metabolismo , Sitios de Empalme de ARN/genética , ARN Mensajero/aislamiento & purificación , ARN Mensajero/metabolismo
16.
FEBS Lett ; 581(21): 4103-8, 2007 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-17686473

RESUMEN

Glucose induced translation of insulin in pancreatic beta cells is mediated by the 5'UTR of insulin mRNA. We determined the minimal sequence/structure in the 5'UTR of rat insulin gene1 for this regulation. We show that specific factors in the pancreatic islets bind to the 5'UTR of the insulin mRNA upon glucose stimulation. We identified a minimal 29-nucleotide element in the 5'UTR that is sufficient to form the complex, and confer glucose mediated translation activation. Conserved residues in the predicted stem loop region of the un-translated region (UTR) seem to be important for the complex formation and the translation regulation.


Asunto(s)
Regiones no Traducidas 5'/metabolismo , Glucosa/metabolismo , Insulina/biosíntesis , Islotes Pancreáticos/metabolismo , Biosíntesis de Proteínas/fisiología , Regiones no Traducidas 5'/genética , Animales , Insulina/genética , Conformación de Ácido Nucleico , Especificidad de Órganos/fisiología , Proteínas de Unión al ARN/metabolismo , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA