Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Dev Dyn ; 249(5): 646-655, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31872525

RESUMEN

BACKGROUND: Dysfunction of GABAergic and glutamatergic neurons in the brain, which establish inhibitory and excitatory networks, respectively, may cause diverse neurological disorders. The mechanism underlying the determination of GABAergic vs. glutamatergic neurotransmitter phenotype in the caudal diencephalon remains largely unknown. RESULTS: In this study, we investigated the consequence of Tcf7l2 (transcription factor 7-like 2) ablation on the neurotransmitter identity of GABAergic and glutamatergic neurons in the caudal diencephalon. We identified positive and negative activity in the control of glutamatergic and GABAergic neuronal gene expression by Tcf7l2. Loss of Tcf7l2 did not alter the initial acquisition of the neurotransmitter identity in thalamic neurons. However, glutamatergic thalamic neurons failed to maintain their excitatory neurotransmitter phenotype in the absence of Tcf7l2. Moreover, a subset of Tcf7l2-deficient thalamic neurons underwent a glutamatergic to GABAergic neurotransmitter identity switch. Our data indicate that Tcf7l2 may promote glutamatergic neuronal differentiation and repress GABAergic neurotransmitter identity in the caudal thalamus. CONCLUSIONS: This study provides evidence for a novel and crucial role of Tcf7l2 in the molecular mechanism by which the neurotransmitter identity of glutamatergic thalamic neurons is established. Our findings exemplify a clear case of neurotransmitter identity regulation that is partitioned into initiation and maintenance phases.


Asunto(s)
Tálamo , Proteína 2 Similar al Factor de Transcripción 7 , Diencéfalo , Neuronas/metabolismo , Neurotransmisores/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/genética , Proteína 2 Similar al Factor de Transcripción 7/metabolismo
2.
Protein Expr Purif ; 176: 105744, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32890706

RESUMEN

The rhLIF is widely used as an essential factor in stem cell cultures for cell therapies. However, all the recombinant LIFs commercially available are expensive, and no commercially available rhLIF meet the standards recommended by USP for use in cell therapies. The current study reports the efficient production of N-glycosylated and bioactive rhLIF in CHO cells. The production rate of established rhLIF-expressing rCHO cells was approximately 0.85 g/l in 12-day fed-batch cultures using a 7.5 l bioreactor. The rhLIF protein was purified via a four-step purification procedure with approximately 57% recovery rate and greater than 99% purity. The purified rhLIF was N-glycosylated and biologically active with an EC50 of 0.167 ng/ml and a specific activity of 0.86 × 103 IU/mg. The purification procedure controlled the levels of process-related impurities below critical levels recommended by USP for cytokines used in cell therapies. The current work is the first production process of N-glycosylated and bioactive rhLIF, which can be applied to large-scale manufacture of GMP-grade rhLIF for use as an ancillary material in cell therapy.


Asunto(s)
Factor Inhibidor de Leucemia , Animales , Células CHO , Cricetulus , Glicosilación , Humanos , Factor Inhibidor de Leucemia/biosíntesis , Factor Inhibidor de Leucemia/química , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación
3.
Cell Mol Biol Lett ; 24: 28, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31061665

RESUMEN

BACKGROUND: Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor with a pivotal role in physiological and pathological responses to hypoxia. While HIF-1α is known to be involved in hypoxia-induced upregulation of microRNA (miRNA) expression, HIF-1α is also targeted by miRNAs. In this study, miRNAs targeting HIF-1α were identified and their effects on its expression and downstream target genes under hypoxic conditions were investigated. Cell migration under the same conditions was also assessed. METHODS: microRNAs that target HIF-1α were screened using 3'-untranslated region luciferase (3'-UTR-luciferase) reporter assays. The expression levels of HIF-1α and its downstream target genes after transfection with miRNA were assessed using quantitative RT-PCR and western blot analyses. The effect of the miRNAs on the transcriptional activity of HIF-1α was determined using hypoxia-responsive element luciferase (HRE-luciferase) assays. Cell migration under hypoxia was examined using the wound-healing assay. RESULTS: Several of the 19 screened miRNAs considerably decreased the luciferase activity. Transfection with miR-200c had substantial impact on the expression level and transcription activity of HIF-1α. The mRNA level of HIF-1α downstream genes decreased in response to miR-200c overexpression. MiR-200c inhibited cell migration in normoxia and, to a greater extent, in hypoxia. These effects were partly reversed by HIF-1α expression under hypoxic conditions. CONCLUSION: miR-200c negatively affects hypoxia-induced responses by downregulating HIF-1α, a key regulator of hypoxia. Therefore, overexpression of miR-200c might have therapeutic potential as an anticancer agent that inhibits tumor hypoxia.


Asunto(s)
Movimiento Celular/genética , Regulación hacia Abajo/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , MicroARNs/metabolismo , Regiones no Traducidas 3'/genética , Secuencia de Bases , Hipoxia de la Célula/genética , Línea Celular Tumoral , Metilación de ADN/genética , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Luciferasas/metabolismo , MicroARNs/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética , Regulación hacia Arriba/genética , Cicatrización de Heridas
4.
Dev Biol ; 424(1): 62-76, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28219675

RESUMEN

The thalamus acts as a central integrator for processing and relaying sensory and motor information to and from the cerebral cortex, and the habenula plays pivotal roles in emotive decision making by modulating dopaminergic and serotonergic circuits. These neural compartments are derived from a common developmental progenitor domain, called prosomere 2, in the caudal forebrain. Thalamic and habenular neurons exhibit distinct molecular profile, neurochemical identity, and axonal circuitry. However, the mechanisms of how their progenitors in prosomere 2 give rise to these two populations of neurons and contribute to the forebrain circuitry remains unclear. In this study, we discovered a previously unrecognized role for Tcf7l2, a transcription factor known as the canonical Wnt nuclear effector and diabetes risk-conferring gene, in establishing neuronal identity and circuits of the caudal forebrain. Using genetic and chemical axon tracers, we showed that efferent axons of the thalamus, known as the thalamocortical axons (TCAs), failed to elongate normally and strayed from their normal course to inappropriate locations in the absence of Tcf7l2. Further experiments with thalamic explants revealed that the pathfinding defects of Tcf7l2-deficient TCAs were associated at least in part with downregulation of guidance receptors Robo1 and Robo2 expression. Moreover, the fasciculus retroflexus, the main habenular output tract, was missing in embryos lacking Tcf7l2. These axonal defects may result from dysregulation of Nrp2 guidance receptor. Strikingly, loss of Tcf7l2 caused a post-mitotic identity switch between thalamic and habenular neurons. Despite normal acquisition of progenitor identity in prosomere 2, Tcf7l2-deficient thalamic neurons adopted a molecular profile of a neighboring forebrain derivative, the habenula. Conversely, habenular neurons failed to maintain their normal post-mitotic neuronal identity and acquired a subset of thalamic neuronal features in the absence of Tcf7l2. Our findings suggest a unique role for Tcf7l2 in generating distinct neuronal phenotypes from homogeneous progenitor population, and provide a better understanding of the mechanism underlying neuronal specification, differentiation, and connectivity of the developing caudal forebrain.


Asunto(s)
Habénula/citología , Habénula/embriología , Red Nerviosa/metabolismo , Neuronas/metabolismo , Tálamo/citología , Tálamo/embriología , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Animales , Orientación del Axón , Axones/metabolismo , Biomarcadores/metabolismo , Tipificación del Cuerpo , Diencéfalo/embriología , Diencéfalo/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Mitosis , Mutación/genética , Unión Proteica , Células Madre/metabolismo , Transcripción Genética
5.
PLoS Genet ; 11(10): e1005560, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26447474

RESUMEN

Functional diversification of motor neurons has occurred in order to selectively control the movements of different body parts including head, trunk and limbs. Here we report that transcription of Isl1, a major gene necessary for motor neuron identity, is controlled by two enhancers, CREST1 (E1) and CREST2 (E2) that allow selective gene expression of Isl1 in motor neurons. Introduction of GFP reporters into the chick neural tube revealed that E1 is active in hindbrain motor neurons and spinal cord motor neurons, whereas E2 is active in the lateral motor column (LMC) of the spinal cord, which controls the limb muscles. Genome-wide ChIP-Seq analysis combined with reporter assays showed that Phox2 and the Isl1-Lhx3 complex bind to E1 and drive hindbrain and spinal cord-specific expression of Isl1, respectively. Interestingly, Lhx3 alone was sufficient to activate E1, and this may contribute to the initiation of Isl1 expression when progenitors have just developed into motor neurons. E2 was induced by onecut 1 (OC-1) factor that permits Isl1 expression in LMCm neurons. Interestingly, the core region of E1 has been conserved in evolution, even in the lamprey, a jawless vertebrate with primitive motor neurons. All E1 sequences from lamprey to mouse responded equally well to Phox2a and the Isl1-Lhx3 complex. Conversely, E2, the enhancer for limb-innervating motor neurons, was only found in tetrapod animals. This suggests that evolutionarily-conserved enhancers permit the diversification of motor neurons.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Proteínas de Homeodominio/biosíntesis , Proteínas con Homeodominio LIM/biosíntesis , Factores de Transcripción/biosíntesis , Animales , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas con Homeodominio LIM/genética , Lampreas/genética , Ratones , Neuronas Motoras/metabolismo , Neuronas Motoras/fisiología , Rombencéfalo/metabolismo , Rombencéfalo/fisiología , Médula Espinal/metabolismo , Médula Espinal/fisiología , Factores de Transcripción/genética
6.
Dev Dyn ; 246(10): 749-758, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28685891

RESUMEN

BACKGROUND: The neurons contributing to thalamic nuclei are derived from at least two distinct progenitor domains: the caudal (cTH) and rostral (rTH) populations of thalamic progenitors. These neural compartments exhibit unique neurogenic patterns, and the molecular mechanisms underlying the acquisition of neurotransmitter identity remain largely unclear. RESULTS: T-cell acute lymphocytic leukemia protein 1 (Tal1) was expressed in the early postmitotic cells in the rTH domain, and its expression was maintained in mature thalamic neurons in the ventrolateral geniculate nucleus (vLG) and the intergeniculate leaflet (IGL). To investigate a role of Tal1 in thalamic development, we used a newly generated mouse line driving Cre-mediated recombination in the rTH domain. Conditional deletion of Tal1 did not alter regional patterning in the developing diencephalon. However, in the absence of Tal1, rTH-derived thalamic neurons failed to maintain their postmitotic neuronal features, including neurotransmitter profile. Tal1-deficient thalamic neurons lost their GABAergic markers such as Gad1, Npy, and Penk in IGL/vLG. These defects may be associated at least in part with down-regulation of Nkx2.2, which is known as a critical regulator of rTH-derived GABAergic neurons. CONCLUSIONS: Our results demonstrate that Tal1 plays an essential role in regulating neurotransmitter phenotype in the developing thalamic nuclei. Developmental Dynamics 246:749-758, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Neurotransmisores , Proteína 1 de la Leucemia Linfocítica T Aguda/fisiología , Núcleos Talámicos/citología , Animales , Proteína Homeobox Nkx-2.2 , Ratones , Células Madre , Núcleos Talámicos/embriología , Tálamo/citología , Tálamo/embriología
7.
Genesis ; 55(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28093895

RESUMEN

The sine oculis homeobox protein Six3 plays pivotal roles in the development of the brain and craniofacial structures. In humans, SIX3 haploinsufficiency results in holoprosencephaly, a defect in anterior midline formation. Although much is known about the evolutionarily conserved functions of Six3, the regulatory mechanism responsible for the expression pattern of Six3 remains relatively unexplored. To understand how the transcription of Six3 is controlled during embryogenesis, we screened ∼300 kb of genomic DNA encompassing the Six3 locus for cis-acting regulatory elements capable of directing reporter gene expression to sites of Six3 transcription in transgenic mouse embryos. We identified a novel enhancer element, whose activity recapitulates endogenous Six3 expression in the ventral midbrain, pretectum, and thalamus. Cross-species comparisons revealed that this Six3 brain enhancer is functionally conserved in other vertebrates. We also showed that normal Six3 transcription in the ventral midbrain and pretectum is dependent on Ascl1, a basic helix-loop-helix proneural factor. Moreover, loss of Ascl1 resulted in downregulation of the Six3 brain enhancer activity, emphasizing its unique role in regulating Six3 expression in the developing brain.


Asunto(s)
Encéfalo/metabolismo , Secuencia Conservada , Elementos de Facilitación Genéticos , Proteínas del Ojo/genética , Proteínas de Homeodominio/genética , Proteínas del Tejido Nervioso/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Encéfalo/embriología , Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteína Homeobox SIX3
8.
Dev Biol ; 398(2): 280-91, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25512300

RESUMEN

The mammalian thalamus is an essential diencephalic derivative that plays unique roles in processing and relaying sensory and motor information to and from the cerebral cortex. The profile of transcription factors and lineage tracing experiments revealed a spatiotemporal relationship between diencephalic progenitor domains and discrete differentiated neurons contributing to thalamic nuclei. However, the precise molecular mechanisms by which heterogeneous thalamic neurons become specified and assemble into distinct thalamic nuclei are still poorly understood. Here, we show that a combinatorial interaction between the bHLH transcription factors Ascl1 and Helt is required for acquiring thalamic progenitor identity. Surprisingly, in the combined absence of Ascl1 and Helt, rostral thalamic progenitors (TH-R) adopt a molecular profile of a more rostral diencephalic derivative, the prethalamus. Furthermore, we show that the prethalamic factors Dlxs upregulated by Ascl1/Helt deficiency play unique roles in regulating thalamic progenitor specification, and that derepression of Dlx2 and Dlx5 suppress generation of TH-R neurons. Taken together, our results suggest a model whereby the combined activity of two distinct bHLH factors plays a key role in the development of discrete classes of thalamic interneurons.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Homeodominio/metabolismo , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Tálamo/citología , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Tipificación del Cuerpo/genética , Linaje de la Célula , Factor de Transcripción GATA2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Transactivadores/metabolismo , Factores de Transcripción/genética
9.
Protein Expr Purif ; 120: 87-91, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26711959

RESUMEN

Bone morphogenetic protein-7 is a multifunctional growth factor involved in various cellular processes such as osteogenesis, kidney and eye development, brown adipogenesis, and bone metastasis, and thus has been considered to have therapeutic potential for treating various diseases. In this study, we established a Chinese hamster ovary (CHO) cell line stably overexpressing recombinant human BMP-7 (rhBMP-7). Over the course of a 14-day fed-batch culture process in a 7.5-l bioreactor (5-l working volume) using chemically defined medium, the established cells could produce over 188 mg/l of rhBMP-7 protein. The rhBMP-7 was purified to homogeneity from the culture supernatant using a two-step chromatographic procedure that resulted in a recovery rate of approximately 55%, with protein purity greater than 95%. The purified rhBMP-7 was further demonstrated to be functionally active by measuring the proliferation of MC3T3-E1 cells, revealing a half-maximal effective concentration of 28.31 ng/ml.


Asunto(s)
Proteína Morfogenética Ósea 7 , Animales , Reactores Biológicos , Células CHO , Cromatografía , Clonación Molecular , Cricetulus/genética , Humanos , Proteínas Recombinantes
10.
Exp Cell Res ; 339(2): 320-32, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26485640

RESUMEN

Micro(mi)RNAs play important and varied roles in tumorigenesis; however, the full repertoire of miRNAs that affect cancer cell growth is not known. In this study, an miRNA library was screened to identify those that affect the growth of A549 tumor cells. Among 300 miRNAs, miR-28-5p, -323-5p, -510-5p, -552-3p, and -608 were the most effective in inhibiting cell growth. More specifically, overexpressing miR-28-5p, -323-5p, and -510-5p induced G1 arrest, as determined by flow cytometry, whereas that of miR-608 induced cell death in a caspase-dependent manner. Moreover, several genes involved in apoptosis and cell cycle progression were downregulated upon overexpression of each of the five miRNAs, with the functional targets of miR-552-3p and miR-608 confirmed by microarray, quantitative real-time PCR, and luciferase reporter assay. In miR-608-transfected cells, B cell lymphoma 2-like 1 (BCL2L1), D-type cyclin 1 (CCND1), CCND3, cytochrome b5 reductase 3 (CYB5R3), phosphoinositide 3-kinase regulatory subunit 2 (PIK3R2), specificity protein 1 (SP1), and phosphorylated Akt were all downregulated, while Bcl-2-interacting killer (BIK) was upregulated. Moreover, miR-608 was determined to have a suppressive function on tumor growth in an NCI-H460 xenograft model. These findings provide insights into the roles of five miRNAs in growth inhibition and their potential function as cancer therapeutics.


Asunto(s)
Apoptosis/genética , Ciclo Celular/genética , Biblioteca de Genes , MicroARNs/análisis , MicroARNs/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/metabolismo , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Genesis ; 53(6): 356-65, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25950659

RESUMEN

The mammalian diencephalon is the caudal derivative of the embryonic forebrain. Early events in diencephalic regionalization include its subdivision along the dorsoventral and anteroposterior axes. The prosomeric model by Puelles and Rubenstein (1993) suggests that the alar plate of the posterior diencephalon is partitioned into three different prosomeres (designated p1-p3), which develop into the pretectum, thalamus, and prethalamus, respectively. Here, we report the developmental consequences of genetic ablation of cell populations from the diencephalic basal plate. The strategy for conditionally regulated cell ablation is based on the targeted expression of the diphtheria toxin gene (DTA) to the diencephalic basal plate via tamoxifen- induced, Cre-mediated recombination of the ROSA(DTA) allele. We show that activation of DTA leads to specific cell loss in the basal plate of the posterior diencephalon, and disrupted early regionalization of distinct alar territories. In the basal plate-deficient embryos, the p1 alar plate exhibited reduced expression of subtype-specific markers in the pretectum, whereas p2 alar plate failed to further subdivide into two discrete thalamic subpopulations. We also show that these defects lead to abnormal nuclear organization at later developmental stages. Our data have implications for increased understanding of the interactive roles between discrete diencephalic compartments.


Asunto(s)
Diencéfalo/embriología , Diencéfalo/metabolismo , Toxina Diftérica/genética , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diencéfalo/anatomía & histología , Toxina Diftérica/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Inmunohistoquímica , Hibridación in Situ , Ratones Transgénicos , Organogénesis/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Tiempo
12.
Protein Expr Purif ; 110: 102-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25727048

RESUMEN

Transforming growth factor beta 3 (TGFß3) is an important cytokine, functioning in cell proliferation and differentiation, and has been considered to have therapeutic potential for treating various diseases and for scar reduction in adult wound healing. In the current study, a Chinese hamster ovary (CHO) cell line overexpressing recombinant human TGFß3 (rhTGFß3) was established. Through a 15-day fed-batch culture process in a 7.5-l bioreactor (5-l working volume) using chemically defined medium, the established cells could produce over 133mg/l of rhTGFß3 protein. The rhTGFß3 was purified to homogeneity from the culture supernatant using a two-step chromatographic procedure, resulting in a recovery rate of approximately 65%, with protein purity greater than 97%. The N-terminal amino acid sequences of the purified rhTGFß3 were confirmed by N-terminal sequencing analysis. The purified rhTGFß3 was further demonstrated to be functionally active by measuring the inhibition of growth of HT-2 cells, revealing a half-maximal effective concentration of 42.11pg/ml and specific activity of 1.84×10(7)U/mg.


Asunto(s)
Plásmidos/química , Proteínas Recombinantes/genética , Factor de Crecimiento Transformador beta3/genética , Secuencia de Aminoácidos , Animales , Técnicas de Cultivo Celular por Lotes , Reactores Biológicos , Células CHO , Proliferación Celular/efectos de los fármacos , Clonación Molecular , Cricetulus , Medios de Cultivo/química , Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Plásmidos/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta3/química , Factor de Crecimiento Transformador beta3/aislamiento & purificación , Factor de Crecimiento Transformador beta3/farmacología
13.
Dev Biol ; 381(2): 491-501, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23792023

RESUMEN

The SRY-related HMG box transcription factor Sox2 plays critical roles throughout embryogenesis. Haploinsufficiency for SOX2 results in human developmental defects including anophthalmia, microphthalmia and septo-optic dysplasia, a congenital forebrain defect. To understand how Sox2 plays a role in neurogenesis, we combined genomic and in vivo transgenic approaches to characterize genomic regions occupied by Sox2 in the developing forebrain. Six3, a homeobox gene associated with holoprosencephaly, a forebrain midline defect, was identified as a Sox2 transcriptional target. This study shows that Sox2 directly regulates a previously unidentified long-range forebrain enhancer to activate Six3 expression in the rostral diencephalon. Further biochemical and genetic evidences indicated a direct regulatory link between Sox2 and Six3 during forebrain development, providing a better understanding of a common molecular mechanism underlying these forebrain defects.


Asunto(s)
Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Prosencéfalo/metabolismo , Factores de Transcripción SOXB1/genética , Animales , Secuencia de Bases , Sitios de Unión , Secuencia Conservada , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Elementos de Facilitación Genéticos , Evolución Molecular , Proteínas del Ojo/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Placa Neural/embriología , Placa Neural/metabolismo , Prosencéfalo/citología , Prosencéfalo/embriología , Unión Proteica , Factores de Transcripción SOXB1/metabolismo , Activación Transcripcional , Proteína Homeobox SIX3
14.
Development ; 138(3): 531-41, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21205797

RESUMEN

In caudal regions of the diencephalon, sonic hedgehog (Shh) is expressed in the ventral midline of prosomeres 1-3 (p1-p3), which underlie the pretectum, thalamus and prethalamus, respectively. Shh is also expressed in the zona limitans intrathalamica (zli), a dorsally projecting spike that forms at the p2-p3 boundary. The presence of two Shh signaling centers in the thalamus has made it difficult to determine the specific roles of either one in regional patterning and neuronal fate specification. To investigate the requirement of Shh from a focal source of expression in the ventral midline of the diencephalon, we used a newly generated mouse line carrying a targeted deletion of the 525 bp intronic sequence mediating Shh brain enhancer-1 (SBE1) activity. In SBE1 mutant mice, Shh transcription was initiated but not maintained in the ventral midline of the rostral midbrain and caudal diencephalon, yet expression in the zli was unaffected. In the absence of ventral midline Shh, rostral thalamic progenitors (pTH-R) adopted the molecular profile of a more caudal thalamic subtype (pTH-C). Surprisingly, despite their early mis-specification, neurons derived from the pTH-R domain continued to migrate to their proper thalamic nucleus, extended axons along their normal trajectory and expressed some, but not all, of their terminal differentiation markers. Our results, and those of others, suggest a model whereby Shh signaling from distinct spatial and temporal domains in the diencephalon exhibits unique and overlapping functions in the development of discrete classes of thalamic interneurons.


Asunto(s)
Proteínas Hedgehog/metabolismo , Interneuronas/citología , Interneuronas/metabolismo , Tálamo/citología , Tálamo/metabolismo , Animales , Diencéfalo/citología , Diencéfalo/embriología , Diencéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Mutantes , Tálamo/embriología
15.
Front Cell Dev Biol ; 12: 1336308, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38344749

RESUMEN

The mechanism underlying the differentiation of the dorsal midbrain into two morphologically and functionally distinct compartments, the inferior colliculus (IC) and superior colliculus (SC), which process auditory and visual information, respectively, remains largely unexplored. By using null and conditional alleles, we uncover the roles of a homeodomain transcription factor Dbx1 in the regulation of IC and SC differentiation. We show that Dbx1 regulates GABAergic neuron development in the dorsal midbrain. In the absence of Dbx1 function, the dorsal-most m1-m2 progenitor domains in the midbrain fail to activate GABAergic neuron-specific gene expression and instead switch to a glutamatergic phenotype. These results identify Dbx1 as a dorsal midbrain-specific GABAergic determinant that regulates the selector genes, Helt, Gata2, and Tal2. Furthermore, we demonstrate that maturation of the dorsal midbrain into the IC and SC is dependent on Dbx1. Null mutation of Dbx1 impairs the identity and fate of IC and SC neurons. Surprisingly, Dbx1 is required for preventing IC into SC fate switch and thus Dbx1-deficient IC neurons undergo acquisition of SC identity. Conditional inactivation of Dbx1 at late developmental phase leads to alteration in the identity and fate of the IC, but not the SC. These results suggest that SC differentiation is dependent on the early function of Dbx1, and that the IC requires the prolonged action for its normal formation. Furthermore, we uncover that Tcf7l2 acts downstream of Dbx1 selectively to promote IC differentiation. Altogether, our study identifies a molecular mechanism underlying spatial and temporal control of dorsal midbrain development.

16.
Dev Biol ; 366(2): 393-403, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22561201

RESUMEN

Six6, a sine oculis homeobox protein, plays a crucial and conserved role in the development of the forebrain and eye. To understand how the expression of Six6 is regulated during embryogenesis, we screened ~250 kb of genomic DNA encompassing the Six6 locus for cis-regulatory elements capable of directing reporter gene expression to sites of Six6 transcription in transgenic mouse embryos. Here, we describe two novel enhancer elements, that are highly conserved in vertebrate species and whose activities recapitulate Six6 expression in the ventral forebrain and eye, respectively. Cross-species comparisons of the Six6 forebrain enhancer sequences revealed highly conserved binding sites matching the consensus for homeodomain and SoxB1 transcription factors. Deletion of either of the binding sites resulted in loss of the forebrain enhancer activity in the ventral forebrain. Moreover, our studies show that members of the SoxB1 family, including Sox2 and Sox3, are expressed in the overlapping region of the ventral forebrain with Six6 and can bind to the Six6 forebrain enhancer. Loss of function of SoxB1 genes in vivo further emphasizes their role in regulating Six6 forebrain enhancer activity. Thus, our data strongly suggest that SoxB1 transcription factors are direct activators of Six6 expression in the ventral forebrain.


Asunto(s)
Elementos de Facilitación Genéticos , Proteínas de Homeodominio/genética , Prosencéfalo/fisiología , Factores de Transcripción SOXB1/genética , Transactivadores/genética , Animales , Ojo/embriología , Proteínas del Ojo/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Prosencéfalo/embriología , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción SOXB1/metabolismo
17.
Protein Expr Purif ; 88(1): 1-6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23201283

RESUMEN

Mannose-binding lectin (MBL) is an important serum protein that functions in the innate immune system and has been considered to have therapeutic potential in MBL replacement therapies for patients with deficient or low levels of MBL. In this study, we established a Chinese hamster ovary (CHO) cell line that overexpresses the recombinant human MBL (rhMBL) protein. In an 11-day batch culture process using a 30-L bioreactor (20-L working volume) and serum-free medium, these cells could produce over 226 mg/L of rhMBL protein. The recombinant protein was then purified to homogeneity from the culture supernatant using a three-step chromatographic procedure that resulted in a recovery rate of approximately 55%. This purified rhMBL protein adopted oligomeric bouquet-like structures that were similar to those of native MBL present in human blood, and these oligomeric structures were reported to be critical in MBL functions. We further demonstrated in carbohydrate binding and complementation activation assays that this rhMBL protein was functionally active with very similar dissociation constants and half maximal effective concentrations to those of native MBL.


Asunto(s)
Lectina de Unión a Manosa/biosíntesis , Lectina de Unión a Manosa/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Animales , Reactores Biológicos , Células CHO , Cricetinae , Cricetulus , Humanos , Lectina de Unión a Manosa/química , Proteínas Recombinantes/química
18.
Biotechnol Lett ; 35(7): 987-93, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23468139

RESUMEN

The gradual loss of recombinant protein expression in CHO cell lines during prolonged subculture is a common issue, referred to as instability, which seriously affects the industrial production processes of therapeutic proteins. Loss of recombinant gene copies, due to the genetic instability of CHO cells, and epigenetic silencing of transgene sequences, are the main reported causes of production instability. To increase our understanding on the molecular mechanisms inherent to CHO cells involved in production instability, we explored the molecular features of stable and unstable antibody producing cell lines obtained without gene amplification, to exclude the genetic instability induced by the gene amplification process. The instability of recombinant antibody production during long-term culture was caused by a 48-53% decrease in recombinant mRNA levels without significant loss of recombinant gene copies, but accompanied by a ~45% decrease in histone H3 acetylation (H3ac). Thus, our results suggest a critical role of H3ac in the stability of recombinant protein production.


Asunto(s)
Anticuerpos/metabolismo , Epigénesis Genética , Expresión Génica , Histonas/metabolismo , Acetilación , Animales , Anticuerpos/genética , Células CHO , Técnicas de Cultivo de Célula/métodos , Cricetulus , Inestabilidad Genómica , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Cell Death Differ ; 30(6): 1563-1574, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37081114

RESUMEN

At the top of the midbrain is the inferior colliculus (IC), which functions as the major hub for processing auditory information. Despite the functional significance of neurons in the IC, our understanding of their formation is limited. In this study, we identify the embryonic patterning gene Dbx1 as a key molecular player that governs genetic programs for IC survival. We find that Dbx1 plays a critical role in preventing apoptotic cell death in postnatal IC by transcriptionally repressing c-Jun and pro-apoptotic BH3 only factors. Furthermore, by employing combined approaches, we uncover that Tcf7l2 functions downstream of Dbx1. Loss of Tcf7l2 function causes IC phenotypes with striking similarity to those of Dbx1 mutant mice, which include defective embryonic maturation and postnatal deletion of the IC. Finally, we demonstrate that the Dbx1-Tcf7l2 cascade functions upstream of Ap-2δ, which is essential for IC development and survival. Together, these results unravel a novel molecular mechanism for IC maintenance, which is indispensable for normal brain development.


Asunto(s)
Colículos Inferiores , Mesencéfalo , Animales , Ratones , Proteínas de Homeodominio/metabolismo , Colículos Inferiores/metabolismo , Mesencéfalo/metabolismo , Neuronas/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo
20.
Immune Netw ; 22(5): e41, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36381960

RESUMEN

The human antimicrobial peptide LL-37 has chemotactic and modulatory activities in various immune cells, including dendritic cells. Because of its characteristics, LL-37 can be considered an adjuvant for vaccine development. In this study, we confirmed the possible adjuvant activity of LL-37 in mucosal vaccine development against Middle East respiratory syndrome-coronavirus (MERS-CoV) by means of intranasal immunization in C57BL/6 and human dipeptidyl peptidase 4 (hDPP4)-transgenic (hDPP4-Tg) mice. Intranasal immunization using the receptor-binding domain (RBD) of MERS-CoV spike protein (S-RBD) recombined with LL-37 (S-RBD-LL-37) induced an efficient mucosal IgA and systemic IgG response with virus-neutralizing activity, compared with S-RBD. Ag-specific CTL stimulation was also efficiently induced in the lungs of mice that had been intranasally immunized with S-RBD-LL-37, compared with S-RBD. Importantly, intranasal immunization of hDPP4-Tg mice with S-RBD-LL-37 led to reduced immune cell infiltration into the lungs after infection with MERS-CoV. Finally, intranasal immunization of hDPP4-Tg mice with S-RBD-LL-37 led to enhanced protective efficacy, with increased survival and reduced body weight loss after challenge infection with MERS-CoV. Collectively, these results suggest that S-RBD-LL-37 is an effective intranasal vaccine candidate molecule against MERS-CoV infection.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA