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










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(5): 114167, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38691452

RESUMEN

Polycomb (Pc) group proteins are transcriptional regulators with key roles in development, cell identity, and differentiation. Pc-bound chromatin regions form repressive domains that interact in 3D to assemble repressive nuclear compartments. Here, we use multiplexed chromatin imaging to investigate whether Pc compartments involve the clustering of multiple Pc domains during Drosophila development. Notably, 3D proximity between Pc targets is rare and involves predominantly pairwise interactions. These 3D proximities are particularly enhanced in segments where Pc genes are co-repressed. In addition, segment-specific expression of Hox Pc targets leads to their spatial segregation from Pc-repressed genes. Finally, non-Hox Pc targets are more proximal in regions where they are co-expressed. These results indicate that long-range Pc interactions are temporally and spatially regulated during differentiation and development but do not induce frequent clustering of multiple distant Pc genes.


Asunto(s)
Cromatina , Proteínas de Drosophila , Proteínas del Grupo Polycomb , Animales , Cromatina/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica
2.
Methods Mol Biol ; 2784: 227-257, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38502490

RESUMEN

The simultaneous observation of three-dimensional (3D) chromatin structure and transcription in single cells is critical to understand how DNA is organized inside cells and how this organization influences or is affected by other processes, such as transcription. We have recently introduced an innovative technology known as Hi-M, which enables the sequential tagging, 3D visualization, and precise localization of multiple genomic DNA regions alongside RNA expression within individual cells. In this chapter, we present a comprehensive guide outlining the creation of probes, as well as sample preparation and labeling. Finally, we provide a step-by-step guide to conduct a complete Hi-M acquisition using our open-source software package, Qudi-HiM, which controls the robotic microscope handling the entire acquisition procedure.


Asunto(s)
Cromatina , Cromosomas , Cromatina/genética , Cromosomas/metabolismo , ADN/química , Conformación Molecular
3.
Genome Biol ; 25(1): 47, 2024 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-38351149

RESUMEN

Genome-wide ensemble sequencing methods improved our understanding of chromatin organization in eukaryotes but lack the ability to capture single-cell heterogeneity and spatial organization. To overcome these limitations, new imaging-based methods have emerged, giving rise to the field of spatial genomics. Here, we present pyHiM, a user-friendly python toolbox specifically designed for the analysis of multiplexed DNA-FISH data and the reconstruction of chromatin traces in individual cells. pyHiM employs a modular architecture, allowing independent execution of analysis steps and customization according to sample specificity and computing resources. pyHiM aims to facilitate the democratization and standardization of spatial genomics analysis.


Asunto(s)
Genómica , Programas Informáticos , Genómica/métodos , Cromatina , Cromosomas , ADN
4.
Open Res Eur ; 2: 46, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37645324

RESUMEN

Multiplexed sequential and combinatorial imaging enables the simultaneous detection of multiple biological molecules, e.g. proteins, DNA, or RNA, enabling single-cell spatial multi-omics measurements at sub-cellular resolution. Recently, we designed a multiplexed imaging approach (Hi-M) to study the spatial organization of chromatin in single cells. In order to enable Hi-M sequential imaging on custom microscope setups, we developed Qudi-HiM, a modular software package written in Python 3. Qudi-HiM contains modules to automate the robust acquisition of thousands of three-dimensional multicolor microscopy images, the handling of microfluidics devices, and the remote monitoring of ongoing acquisitions and real-time analysis. In addition, Qudi-HiM can be used as a stand-alone tool for other imaging modalities.

5.
Nat Genet ; 53(4): 477-486, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33795867

RESUMEN

Acquisition of cell fate is thought to rely on the specific interaction of remote cis-regulatory modules (CRMs), for example, enhancers and target promoters. However, the precise interplay between chromatin structure and gene expression is still unclear, particularly within multicellular developing organisms. In the present study, we employ Hi-M, a single-cell spatial genomics approach, to detect CRM-promoter looping interactions within topologically associating domains (TADs) during early Drosophila development. By comparing cis-regulatory loops in alternate cell types, we show that physical proximity does not necessarily instruct transcriptional states. Moreover, multi-way analyses reveal that multiple CRMs spatially coalesce to form hubs. Loops and CRM hubs are established early during development, before the emergence of TADs. Moreover, CRM hubs are formed, in part, via the action of the pioneer transcription factor Zelda and precede transcriptional activation. Our approach provides insight into the role of CRM-promoter interactions in defining transcriptional states, as well as distinct cell types.


Asunto(s)
Linaje de la Célula/genética , Cromatina/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Nucleares/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular , Cromatina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Embrión no Mamífero , Elementos de Facilitación Genéticos , Perfilación de la Expresión Génica , Genómica , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Análisis de la Célula Individual , Factores de Transcripción/clasificación , Factores de Transcripción/metabolismo , Transcripción Genética
6.
Nat Protoc ; 15(3): 840-876, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31969721

RESUMEN

Simultaneous observation of 3D chromatin organization and transcription at the single-cell level and with high spatial resolution may hold the key to unveiling the mechanisms regulating embryonic development, cell differentiation and even disease. We recently developed Hi-M, a technology that enables the sequential labeling, 3D imaging and localization of multiple genomic DNA loci, together with RNA expression, in single cells within whole, intact Drosophila embryos. Importantly, Hi-M enables simultaneous detection of RNA expression and chromosome organization without requiring sample unmounting and primary probe rehybridization. Here, we provide a step-by-step protocol describing the design of probes, the preparation of samples, the stable immobilization of embryos in microfluidic chambers, and the complete procedure for image acquisition. The combined RNA/DNA fluorescence in situ hybridization procedure takes 4-5 d, including embryo collection. In addition, we describe image analysis software to segment nuclei, detect genomic spots, correct for drift and produce Hi-M matrices. A typical Hi-M experiment takes 1-2 d to complete all rounds of labeling and imaging and 4 additional days for image analysis. This technology can be easily expanded to investigate cell differentiation in cultured cells or organization of chromatin within complex tissues.


Asunto(s)
Cromosomas , Regulación del Desarrollo de la Expresión Génica/fisiología , Procesamiento de Imagen Asistido por Computador , Transcripción Genética/fisiología , Animales , Cromatina , ADN/química , ADN/genética , ADN/metabolismo , Drosophila/embriología , Colorantes Fluorescentes , Hibridación Fluorescente in Situ/métodos , ARN/química , ARN/genética , ARN/metabolismo
7.
Mol Cell ; 74(1): 212-222.e5, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30795893

RESUMEN

Eukaryotic chromosomes are organized in multiple scales, from nucleosomes to chromosome territories. Recently, genome-wide methods identified an intermediate level of chromosome organization, topologically associating domains (TADs), that play key roles in transcriptional regulation. However, these methods cannot directly examine the interplay between transcriptional activation and chromosome architecture while maintaining spatial information. Here we present a multiplexed, sequential imaging approach (Hi-M) that permits simultaneous detection of chromosome organization and transcription in single nuclei. This allowed us to unveil the changes in 3D chromatin organization occurring upon transcriptional activation and homologous chromosome unpairing during awakening of the zygotic genome in intact Drosophila embryos. Excitingly, the ability of Hi-M to explore the multi-scale chromosome architecture with spatial resolution at different stages of development or during the cell cycle will be key to understanding the mechanisms and consequences of the 4D organization of the genome.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/genética , Cromosomas de Insectos/genética , Drosophila melanogaster/genética , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Microscopía Fluorescente/métodos , ARN/genética , Análisis de la Célula Individual/métodos , Transcripción Genética , Activación Transcripcional , Animales , Ciclo Celular/genética , Cromatina/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hibridación Fluorescente in Situ , ARN/biosíntesis
8.
Nat Commun ; 8(1): 1753, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-29170434

RESUMEN

At the kilo- to megabase pair scales, eukaryotic genomes are partitioned into self-interacting modules or topologically associated domains (TADs) that associate to form nuclear compartments. Here, we combine high-content super-resolution microscopies with state-of-the-art DNA-labeling methods to reveal the variability in the multiscale organization of the Drosophila genome. We find that association frequencies within TADs and between TAD borders are below ~10%, independently of TAD size, epigenetic state, or cell type. Critically, despite this large heterogeneity, we are able to visualize nanometer-sized epigenetic domains at the single-cell level. In addition, absolute contact frequencies within and between TADs are to a large extent defined by genomic distance, higher-order chromosome architecture, and epigenetic identity. We propose that TADs and compartments are organized by multiple, small-frequency, yet specific interactions that are regulated by epigenetics and transcriptional state.


Asunto(s)
Cromosomas/genética , Drosophila/genética , Animales , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Cromosomas/química , Cromosomas/metabolismo , Drosophila/química , Drosophila/metabolismo , Epigénesis Genética , Genoma , Análisis de la Célula Individual
9.
PLoS One ; 11(3): e0150997, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26974334

RESUMEN

BACKGROUND: Fine tuning of the Wnt/ß-catenin signaling pathway is essential for the proper development and function of the liver. Aberrant activation of this pathway is observed in 20%-40% of hepatocellular carcinomas (HCC). Notum encodes a secreted Wnt deacylase that inhibits Wnt activity and thereby restricts the zone of activation of Wnt/ß-catenin signaling. An important role of NOTUM has been described in development in drosophila, planaria and zebrafish, but its role in the mammalian liver is unknown. Notum is required for spatial control of the Wnt/ß-catenin signaling in several animal models and the Wnt/ß-catenin pathway contributes to liver patterning involved in metabolic zonation. Therefore, Notum may be involved in the liver patterning induced by the Wnt/ß-catenin signaling during the adult stage. METHODOLOGY/PRINCIPAL FINDINGS: We generated a conditional Notum knockout mouse mutant to study the effect of the deletion of Notum in the liver. We show that Notum is a direct target of the Wnt/ß-catenin signaling in the liver. Liver-specific deletion of Notum did not modify liver zonation, but Notum deletion had a long-term effect on mouse physiology. In particular, male mutant mice developed metabolic disorders. CONCLUSION: We show that Notum is not a key actor of Wnt/ß-catenin-dependent liver patterning of adult mice, but has role in liver glucose homeostasis. Male mice deficient in Notum specifically in the liver develop metabolic dysfunctions implicating Notum in the development of Type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Esterasas/genética , Eliminación de Gen , Hepatocitos/enzimología , Hígado/enzimología , Vía de Señalización Wnt/genética , Animales , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/genética , Femenino , Masculino , Ratones , Ratones Mutantes , Especificidad de Órganos
10.
J Cell Biol ; 198(5): 815-32, 2012 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-22945933

RESUMEN

Satellite cells (SCs) are stem cells that mediate skeletal muscle growth and regeneration. Here, we observe that adult quiescent SCs and their activated descendants expressed the homeodomain transcription factor Six1. Genetic disruption of Six1 specifically in adult SCs impaired myogenic cell differentiation, impaired myofiber repair during regeneration, and perturbed homeostasis of the stem cell niche, as indicated by an increase in SC self-renewal. Six1 regulated the expression of the myogenic regulatory factors MyoD and Myogenin, but not Myf5, which suggests that Six1 acts on divergent genetic networks in the embryo and in the adult. Moreover, we demonstrate that Six1 regulates the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway during regeneration via direct control of Dusp6 transcription. Muscles lacking Dusp6 were able to regenerate properly but showed a marked increase in SC number after regeneration. We conclude that Six1 homeoproteins act as a rheostat system to ensure proper regeneration of the tissue and replenishment of the stem cell pool during the events that follow skeletal muscle trauma.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Músculo Esquelético/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/fisiología , Células Madre/fisiología , Cicatrización de Heridas/fisiología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo , Proteínas de Homeodominio/genética , Homeostasis , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Proteína MioD/genética , Proteína MioD/metabolismo , Miofibrillas/metabolismo , Miofibrillas/fisiología , Factor 5 Regulador Miogénico/genética , Factor 5 Regulador Miogénico/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Miogenina/genética , Miogenina/metabolismo , Regeneración/genética , Células Satélite del Músculo Esquelético/metabolismo , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cicatrización de Heridas/genética
11.
J Biol Chem ; 284(50): 34769-76, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19776009

RESUMEN

Focal adhesion kinase (FAK) regulates numerous cellular functions and is critical for processes ranging from embryo development to cancer progression. Although autophosphorylation on Tyr-397 appears required for FAK functions in vitro, its role in vivo has not been established. We addressed this question using a mutant mouse (fakDelta) deleted of exon 15, which encodes Tyr-397. The resulting mutant protein FAKDelta is an active kinase expressed at normal levels. Our results demonstrate that the requirement for FAK autophosphorylation varies during development. FAK(Delta/Delta) embryos developed normally up to embryonic day (E) 12.5, contrasting with the lethality at E8.5 of FAK-null embryos. Thus, autophosphorylation on Tyr-397 is not required for FAK to achieve its functions until late mid-gestation. However, FAK(Delta/Delta) embryos displayed hemorrhages, edema, delayed artery formation, vascular remodeling defects, multiple organ abnormalities, and overall developmental retardation at E13.5-14.5, and died thereafter demonstrating that FAK autophosphorylation is also necessary for normal development. Fibroblasts derived from mutant embryos had a normal stellate morphology and expression of focal adhesion proteins, Src family members, p53, and Pyk2. In contrast, in FAK(Delta/Delta) fibroblasts and endothelial cells, spreading and lamellipodia formation were altered with an increased size and number of focal adhesions, enriched in FAKDelta. FAK mutation also decreased fibroblast proliferation. These results show that the physiological functions of FAK in vivo are achieved through both autophosphorylation-independent and autophosphorylation-dependent mechanisms.


Asunto(s)
Embrión de Mamíferos/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Mutación , Animales , Biomarcadores/metabolismo , Adhesión Celular/fisiología , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/anatomía & histología , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Integrinas/genética , Integrinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación
12.
Pflugers Arch ; 459(1): 115-30, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19756723

RESUMEN

We assessed the involvement of harmonin-b, a submembranous protein containing PDZ domains, in the mechanoelectrical transduction machinery of inner ear hair cells. Harmonin-b is located in the region of the upper insertion point of the tip link that joins adjacent stereocilia from different rows and that is believed to gate transducer channel(s) located in the region of the tip link's lower insertion point. In Ush1c (dfcr-2J/dfcr-2J) mutant mice defective for harmonin-b, step deflections of the hair bundle evoked transduction currents with altered speed and extent of adaptation. In utricular hair cells, hair bundle morphology and maximal transduction currents were similar to those observed in wild-type mice, but adaptation was faster and more complete. Cochlear outer hair cells displayed reduced maximal transduction currents, which may be the consequence of moderate structural anomalies of their hair bundles. Their adaptation was slower and displayed a variable extent. The latter was positively correlated with the magnitude of the maximal transduction current, but the cells that showed the largest currents could be either hyperadaptive or hypoadaptive. To interpret our observations, we used a theoretical description of mechanoelectrical transduction based on the gating spring theory and a motor model of adaptation. Simulations could account for the characteristics of transduction currents in wild-type and mutant hair cells, both vestibular and cochlear. They led us to conclude that harmonin-b operates as an intracellular link that limits adaptation and engages adaptation motors, a dual role consistent with the scaffolding property of the protein and its binding to both actin filaments and the tip link component cadherin-23.


Asunto(s)
Adaptación Fisiológica , Proteínas Portadoras/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Mecanotransducción Celular/fisiología , Potenciales de Acción/fisiología , Animales , Proteínas de Ciclo Celular , Proteínas del Citoesqueleto , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Mutantes , Microscopía Electrónica de Rastreo , Técnicas de Placa-Clamp
13.
J Neurosci ; 27(35): 9439-50, 2007 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-17728457

RESUMEN

Loss of oligophrenin1 (OPHN1) function in human causes X-linked mental retardation associated with cerebellar hypoplasia and, in some cases, with lateral ventricle enlargement. In vitro studies showed that ophn1 regulates dendritic spine through the control of Rho GTPases, but its in vivo function remains unknown. We generated a mouse model of ophn1 deficiency and showed that it mimics the ventricles enlargement without affecting the cerebellum morphoanatomy. The ophn1 knock-out mice exhibit behavioral defects in spatial memory together with impairment in social behavior, lateralization, and hyperactivity. Long-term potentiation and mGluR-dependent long-term depression are normal in the CA1 hippocampal area of ophn1 mutant, whereas paired-pulse facilitation is reduced. This altered short-term plasticity that reflects changes in the release of neurotransmitters from the presynaptic processes is associated with normal synaptic density together with a reduction in mature dendritic spines. In culture, inactivation of ophn1 function increases the density and proportion of immature spines. Using a conditional model of loss of ophn1 function, we confirmed this immaturity defect and showed that ophn1 is required at all the stages of the development. These studies show that, depending of the context, ophn1 controls the maturation of dendritic spines either by maintaining the density of mature spines or by limiting the extension of new filopodia. Altogether, these observations indicate that cognitive impairment related to OPHN1 loss of function is associated with both presynaptic and postsynaptic alterations.


Asunto(s)
Ventrículos Cerebrales/patología , Proteínas del Citoesqueleto/fisiología , Espinas Dendríticas/patología , Proteínas Activadoras de GTPasa/fisiología , Trastornos de la Memoria , Neuronas/patología , Proteínas Nucleares/fisiología , Conducta Espacial/fisiología , Análisis de Varianza , Animales , Conducta Animal , Células Cultivadas , Proteínas del Citoesqueleto/deficiencia , Espinas Dendríticas/ultraestructura , Conducta Exploratoria/fisiología , Femenino , GTP Fosfohidrolasas/metabolismo , Proteínas Activadoras de GTPasa/deficiencia , Hipocampo/citología , Masculino , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión/métodos , Neuronas/ultraestructura , Proteínas Nucleares/deficiencia , Fragmentos de Péptidos/metabolismo , Tinción con Nitrato de Plata/métodos , Trastorno de la Conducta Social/genética , Proteína de Unión al GTP rac1/metabolismo
14.
Mol Cell Biol ; 26(17): 6664-74, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16914747

RESUMEN

Serum response factor (SRF) is a crucial transcriptional factor for muscle-specific gene expression. We investigated SRF function in adult skeletal muscles, using mice with a postmitotic myofiber-targeted disruption of the SRF gene. Mutant mice displayed severe skeletal muscle mass reductions due to a postnatal muscle growth defect resulting in highly hypotrophic adult myofibers. SRF-depleted myofibers also failed to regenerate following injury. Muscles lacking SRF had very low levels of muscle creatine kinase and skeletal alpha-actin (SKA) transcripts and displayed other alterations to the gene expression program, indicating an overall immaturity of mutant muscles. This loss of SKA expression, together with a decrease in beta-tropomyosin expression, contributed to myofiber growth defects, as suggested by the extensive sarcomere disorganization found in mutant muscles. However, we observed a downregulation of interleukin 4 (IL-4) and insulin-like growth factor 1 (IGF-1) expression in mutant myofibers which could also account for their defective growth and regeneration. Indeed, our demonstration of SRF binding to interleukin 4 and IGF-1 promoters in vivo suggests a new crucial role for SRF in pathways involved in muscle growth and regeneration.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Interleucina-4/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/fisiología , Regeneración , Factor de Respuesta Sérica/metabolismo , Animales , Animales Recién Nacidos , Secuencia de Bases , Núcleo Celular/metabolismo , Tamaño de la Célula , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/genética , Integrasas/genética , Interleucina-4/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Músculo Esquelético/citología , Músculo Esquelético/ultraestructura , Tamaño de los Órganos , Fenotipo , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sarcómeros/patología , Sarcómeros/ultraestructura , Factor de Respuesta Sérica/deficiencia , Factor de Respuesta Sérica/genética
15.
Blood ; 108(4): 1402-5, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16574947

RESUMEN

We previously reported that mice made deficient for the transcriptional factor USF2 fail to express hepcidin 1 and hepcidin 2 genes as a consequence of targeted disruption of the Usf2 gene lying just upstream in the locus. These mice developed an iron overload phenotype with excess iron deposition in parenchymal cells and decreased reticuloendothelial iron. At that time, although the role of USF2 was still confounding, we proposed for the first time the role of hepcidin as a negative regulator of iron absorption and iron release from macrophages. Accordingly, we subsequently demonstrated that hyperexpression of hepcidin 1, but not hepcidin 2, resulted in a profound hyposideremic anemia. To analyze the consequences of hepcidin 1 deletion on iron metabolism without any disturbance due to USF2 deficiency, we disrupted the hepcidin 1 gene by targeting almost all the coding region. Confirming our prior results, Hepc1(-/-) mice developed early and severe multivisceral iron overload, with sparing of the spleen macrophages, and demonstrated increased serum iron and ferritin levels as compared with their controls.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/deficiencia , Eliminación de Gen , Hemocromatosis/genética , Sistemas de Lectura Abierta/genética , Sitios de Carácter Cuantitativo/genética , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Ferritinas/metabolismo , Hemocromatosis/metabolismo , Hemocromatosis/patología , Hepcidinas , Hierro/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Bazo/metabolismo , Bazo/patología , Factores Estimuladores hacia 5'/deficiencia , Factores Estimuladores hacia 5'/metabolismo
16.
Hum Mol Genet ; 15(9): 1387-400, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16571605

RESUMEN

Type I lissencephaly results from mutations in the doublecortin (DCX) and LIS1 genes. We generated Dcx knockout mice to further understand the pathophysiological mechanisms associated with this cortical malformation. Dcx is expressed in migrating interneurons in developing human and mouse brains. Video microscopy analyses of such tangentially migrating neuron populations derived from the medial ganglionic eminence show defects in migratory dynamics. Specifically, the formation and division of growth cones, leading to the production of new branches, are more frequent in knockout cells, although branches are less stable. Dcx-deficient cells thus migrate in a disorganized manner, extending and retracting short branches and making less long-distant movements of the nucleus. Despite these differences, migratory speeds and distances remain similar to wild-type cells. These novel data thus highlight a role for Dcx, a microtubule-associated protein enriched at the leading edge in the branching and nucleokinesis of migrating interneurons.


Asunto(s)
Movimiento Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Interneuronas/patología , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Neuropéptidos/deficiencia , Neuropéptidos/genética , Animales , Células Cultivadas , Técnicas de Cocultivo , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Femenino , Masculino , Eminencia Media/citología , Eminencia Media/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/fisiología , Neuropéptidos/fisiología , Técnicas de Cultivo de Órganos
17.
Nat Med ; 11(5): 499-506, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15834428

RESUMEN

Vascular endothelial growth factor (VEGF)-induced blood vessel growth is involved in both physiological and pathological angiogenesis and requires integrin-mediated signaling. We now show that an integrin-binding protein initially described in milk-fat globule, MFG-E8 (also known as lactadherin), is expressed in and around blood vessels and has a crucial role in VEGF-dependent neovascularization in the adult mouse. Using neutralizing antibodies and lactadherin-deficient animals, we show that lactadherin interacts with alphavbeta3 and alphavbeta5 integrins and alters both VEGF-dependent Akt phosphorylation and neovascularization. In the absence of VEGF, lactadherin administration induced alphavbeta3- and alphavbeta5-dependent Akt phosphorylation in endothelial cells in vitro and strongly improved postischemic neovascularization in vivo. These results show a crucial role for lactadherin in VEGF-dependent neovascularization and identify lactadherin as an important target for the modulation of neovascularization.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Antígenos de Superficie/metabolismo , Proteínas de la Leche/metabolismo , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Análisis de Varianza , Animales , Southern Blotting , Cruzamientos Genéticos , Femenino , Vectores Genéticos , Humanos , Integrina alfaVbeta3/metabolismo , Isquemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt
18.
Development ; 132(9): 2235-49, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15788460

RESUMEN

In mammals, Six5, Six4 and Six1 genes are co-expressed during mouse myogenesis. Six4 and Six5 single knockout (KO) mice have no developmental defects, while Six1 KO mice die at birth and show multiple organ developmental defects. We have generated Six1Six4 double KO mice and show an aggravation of the phenotype previously reported for the single Six1 KO. Six1Six4 double KO mice are characterized by severe craniofacial and rib defects, and general muscle hypoplasia. At the limb bud level, Six1 and Six4 homeogenes control early steps of myogenic cell delamination and migration from the somite through the control of Pax3 gene expression. Impaired in their migratory pathway, cells of the somitic ventrolateral dermomyotome are rerouted, lose their identity and die by apoptosis. At the interlimb level, epaxial Met expression is abolished, while it is preserved in Pax3-deficient embryos. Within the myotome, absence of Six1 and Six4 impairs the expression of the myogenic regulatory factors myogenin and Myod1, and Mrf4 expression becomes undetectable. Myf5 expression is correctly initiated but becomes restricted to the caudal region of each somite. Early syndetomal expression of scleraxis is reduced in the Six1Six4 embryo, while the myotomal expression of Fgfr4 and Fgf8 but not Fgf4 and Fgf6 is maintained. These results highlight the different roles played by Six proteins during skeletal myogenesis.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/metabolismo , Desarrollo de Músculos/fisiología , Proteínas Musculares/genética , Factores Reguladores Miogénicos/genética , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Animales , Apoptosis/fisiología , Huesos/anomalías , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/genética , Ratones , Proteínas Musculares/metabolismo , Músculos/anomalías , Músculos/embriología , Factor 5 Regulador Miogénico , Factores Reguladores Miogénicos/metabolismo , Miogenina , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box , Transactivadores/deficiencia , Factores de Transcripción/metabolismo
19.
Lab Invest ; 84(12): 1619-30, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15502862

RESUMEN

Murine models of familial adenomatous polyposis harbor a germinal heterozygous mutation on Apc tumor suppressor gene. They are valuable tools for studying intestinal carcinogenesis, as most human sporadic cancers contain inactivating mutations of APC. However, Apc(+/-) mice, such as the well-characterized Apc(Min/+) model, develop cancers principally in the small intestine, while humans develop mainly colorectal cancers. We used a Cre-loxP strategy to achieve a new model of germline Apc invalidation in which exon 14 is deleted. We compared the phenotype of these Apc(Delta14/+) mice to that of the classical Apc(Min/+). The main phenotypic difference is the shift of the tumors in the distal colon and rectum, often associated with a rectal prolapse. Thus, the severity of the colorectal phenotype is partly due to the particular mutation Delta14, but also to environmental parameters, as mice raised in conventional conditions developed more colon cancers than those raised in pathogen-free conditions. All lesions, including early lesions, revealed Apc LOH and loss of Apc gene expression. They accumulated beta-catenin, overexpressed the beta-catenin target genes cyclin D1 and c-Myc, and the distribution pattern of glutamine synthetase, a beta-catenin target gene recently identified in the liver, was mosaic in intestinal adenomas. The Apc(Delta14/+) model is thus a useful new tool for studies on the molecular mechanisms of colorectal tumorigenesis.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/fisiopatología , Eliminación de Gen , Genes APC/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células Clonales , Colon/patología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/patología , ADN de Neoplasias/genética , Modelos Animales de Enfermedad , Ambiente , Exones , Biblioteca de Genes , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , ARN Neoplásico/genética , Neoplasias del Recto/genética , Neoplasias del Recto/patología , Eliminación de Secuencia
20.
Development ; 130(10): 2239-52, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12668636

RESUMEN

Six homeoproteins are expressed in several tissues, including muscle, during vertebrate embryogenesis, suggesting that they may be involved in diverse differentiation processes. To determine the functions of the Six1 gene during myogenesis, we constructed Six1-deficient mice by replacing its first exon with the lacZ gene. Mice lacking Six1 die at birth because of severe rib malformations and show extensive muscle hypoplasia affecting most of the body muscles in particular certain hypaxial muscles. Six1(-/-) embryos have impaired primary myogenesis, characterized, at E13.5, by a severe reduction and disorganisation of primary myofibers in most body muscles. While Myf5, MyoD and myogenin are correctly expressed in the somitic compartment in early Six1(-/-) embryos, by E11.5 MyoD and myogenin gene activation is reduced and delayed in limb buds. However, this is not the consequence of a reduced ability of myogenic precursor cells to migrate into the limb buds or of an abnormal apoptosis of myoblasts lacking Six1. It appears therefore that Six1 plays a specific role in hypaxial muscle differentiation, distinct from those of other hypaxial determinants such as Pax3, cMet, Lbx1 or Mox2.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Desarrollo de Músculos/fisiología , Animales , Apoptosis/fisiología , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Femenino , Marcación de Gen , Proteínas de Homeodominio/genética , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Transgénicos , Desarrollo de Músculos/genética , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Proteína MioD/genética , Proteína MioD/metabolismo , Miogenina/genética , Miogenina/metabolismo , Fenotipo , Costillas/patología , Esternón/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...