Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Mol Med (Berl) ; 100(7): 973-983, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35670836

RESUMEN

Coregulator proteins interact with signal-dependent transcription factors to modify their transcriptional activity. ZMIZ1 and ZMIZ2 (zinc finger MIZ-type containing 1 and 2) are coregulators with nonredundant functions that share unique structural characteristics. Among other interacting domains, they possess a MIZ (Msx-interacting zinc finger) that relates them to members of the protein inhibitor of activated STAT (PIAS) family and provides them the capacity to function as SUMO E3 ligases. The ZMIZ proteins stimulate the activity of various signaling pathways, including the androgen receptor (AR), P53, SMAD3/4, WNT/ß-catenin, and NOTCH1 pathways, and interact with the BAF chromatin remodeling complex. Due to their molecular versatility, ZMIZ proteins have pleiotropic effects and thus are important for embryonic development and for human diseases. Both have been widely associated with cancer, and ZMIZ1 has been very frequently identified as a risk allele for several autoimmune conditions and other disorders. Moreover, mutations in the coding region of the ZMIZ1 gene are responsible for a severe syndromic neurodevelopmental disability. Because the actions of coregulators are highly gene-specific, a better knowledge of the associations that exist between the function of the ZMIZ coregulators and human pathologies is expected to potentiate the use of ZMIZ1 and ZMIZ2 as new drug targets for diseases such as hormone-dependent cancers.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Inhibidoras de STAT Activados , Factores de Transcripción/metabolismo , Humanos , Proteínas Inhibidoras de STAT Activados/genética , Proteínas Inhibidoras de STAT Activados/metabolismo , Factores de Riesgo
2.
Life Sci ; 284: 119900, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34453946

RESUMEN

ZMIZ1 is a transcriptional coactivator that is related to members of the protein inhibitor of activated STAT (PIAS) family. ZMIZ1 regulates the activity of various transcription factors including the androgen receptor, p53, and Smad3. ZMIZ1 also interacts with Notch1 and selectively regulates Notch1 target genes relevant for T cell development and leukemogenesis in mammals. Human ZMIZ1 is additionally characterized as a latitude-dependent autoimmune disease (LDAD) risk gene, as it is responsive to vitamin D and has been associated with at least eleven blood cell traits. To address the function of ZMIZ1 in fish, we introduced CRISPR/Cas9 mutations in the zmiz1a gene in zebrafish. We observed that inactivation of zmiz1a in developing zebrafish larvae results in lethality at 15 days post fertilization (dpf) and delayed erythroid maturation. Differential gene expression analysis indicated that 15 dpf zmiz1a-null larvae had altered expression of autophagy genes, and erythrocytes that lacked Zmiz1a function exhibited an accumulation of mitochondrial DNA. Furthermore, we observed that autophagy gene expression was dysregulated at earlier stages of development, which suggests the involvement of Zmiz1a in the regulation of autophagy genes beyond the process of red blood cell differentiation. Finally, we showed that the loss of Zmiz1a decreased the capacity of the embryos to respond to vitamin D, indicating additional participation of Zmiz1a as a mediator of vitamin D activity.


Asunto(s)
Autofagia/genética , Eritropoyesis/genética , Regulación de la Expresión Génica/efectos de los fármacos , Mutación/genética , Vitamina D/farmacología , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Autofagia/efectos de los fármacos , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Embrión no Mamífero/patología , Eritrocitos/efectos de los fármacos , Eritrocitos/patología , Eritropoyesis/efectos de los fármacos , Dosificación de Gen , Hemoglobinas/metabolismo , Inflamación/genética , Transcriptoma/genética , Pez Cebra/embriología , Proteínas de Pez Cebra/metabolismo
3.
Dev Dyn ; 249(11): 1302-1317, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32996661

RESUMEN

The hematopoietic stem cell (HSC) is able to give rise to all blood cell lineages in vertebrates. HSCs are generated in the early embryo after two precedent waves of primitive hematopoiesis. Canonical Notch signaling is at the center of the complex mechanism that controls the development of the definitive HSC. The successful in vitro generation of hematopoietic cells from pluripotent stem cells with the capacity for multilineage hematopoietic reconstitution after transplantation requires the recapitulation of the most important process that takes place in the hemogenic endothelium during definitive hematopoiesis, that is the endothelial-to-hematopoietic transition (EHT). To meet this challenge, it is necessary to thoroughly understand the molecular mechanisms that modulate Notch signaling during the HSC differentiation process considering different temporal and spatial dimensions. In recent years, there have been important advances in this field. Here, we review relevant contributions describing different genes, factors, environmental cues, and signaling cascades that regulate the EHT through Notch interactions at multiple levels. The evolutionary conservation of the hematopoietic program has made possible the use of diverse model systems. We describe the contributions of the zebrafish model and the most relevant ones from transgenic mouse studies and from in vitro differentiated pluripotent cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/metabolismo , Receptores Notch , Transducción de Señal/fisiología , Animales , Humanos , Receptores Notch/genética , Receptores Notch/metabolismo
4.
Dev Biol ; 466(1-2): 22-35, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32828730

RESUMEN

Metamorphosis is a postembryonic developmental process that involves morphophysiological and behavioral changes, allowing organisms to adapt into a novel environment. In some amphibians, aquatic organisms undergo metamorphosis to adapt in a terrestrial environment. In this process, these organisms experience major changes in their circulatory, respiratory, digestive, excretory and reproductive systems. We performed a transcriptional global analysis of heart, lung and gills during diverse stages of Ambystoma velasci to investigate its metamorphosis. In our analyses, we identified eight gene clusters for each organ, according to the expression patterns of differentially expressed genes. We found 4064 differentially expressed genes in the heart, 4107 in the lung and 8265 in the gills. Among the differentially expressed genes in the heart, we observed genes involved in the differentiation of cardiomyocytes in the interatrial zone, vasculogenesis and in the maturation of coronary vessels. In the lung, we found genes differentially expressed related to angiogenesis, alveolarization and synthesis of the surfactant protein. In the case of the gills, the most prominent biological processes identified are degradation of extracellular matrix, apoptosis and keratin production. Our study sheds light on the transcriptional responses and the pathways modulation involved in the transformation of the facultative metamorphic salamander A. velasci in an organ-specific manner.


Asunto(s)
Proteínas Anfibias/biosíntesis , Embrión no Mamífero/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Metamorfosis Biológica/fisiología , Transcriptoma/fisiología , Ambystoma , Animales , Especificidad de Órganos/fisiología
5.
Zebrafish ; 16(6): 505-507, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31408407

RESUMEN

Characterization of a protein of interest during development is essential for functional studies. A general strategy for understanding the function of a particular protein involves the generation of null mutations, or treatment with drugs, that interfere with its activity. To demonstrate that the synthesis, stability, or activity of a protein has been affected, accurate and efficient detection of low amounts of protein is essential. This can be achieved by immunohistochemistry or by western blot. Here we describe a method for the detection of proteins from single de-yolked zebrafish embryos. This procedure includes a fixation step and the concomitant elimination of lipids from the yolk cell. We show that this approach allows the rapid analysis of proteins in embryos without having to manually remove the yolk. This method provides a convenient alternative for genotyping of mutant embryos as early as the 128 cell stage. In addition, in drug- or morpholino-treated embryos, the correlation between the penetrance of a phenotype and the concentration of a protein present may be established.


Asunto(s)
Western Blotting/métodos , Embrión no Mamífero/química , Proteínas de Peces/aislamiento & purificación , Técnicas de Genotipaje/métodos , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
6.
Free Radic Biol Med ; 130: 82-98, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30342187

RESUMEN

Cell movements are essential for morphogenesis during animal development. Epiboly is the first morphogenetic process in zebrafish in which cells move en masse to thin and spread the deep and enveloping cell layers of the blastoderm over the yolk cell. While epiboly has been shown to be controlled by complex molecular networks, the contribution of reactive oxygen species (ROS) to this process has not previously been studied. Here, we show that ROS are required for epiboly in zebrafish. Visualization of ROS in whole embryos revealed dynamic patterns during epiboly progression. Significantly, inhibition of NADPH oxidase activity leads to a decrease in ROS formation, delays epiboly, alters E-cadherin and cytoskeleton patterns and, by 24 h post-fertilization, decreases embryo survival, effects that are rescued by hydrogen peroxide treatment. Our findings suggest that a delicate ROS balance is required during early development and that disruption of that balance interferes with cell adhesion, leading to defective cell motility and epiboly progression.


Asunto(s)
Blastodermo/metabolismo , Citoesqueleto/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Pez Cebra/fisiología , Animales , Cadherinas/metabolismo , Adhesión Celular , Movimiento Celular , Embrión no Mamífero , Morfogénesis , Proteínas de Pez Cebra/metabolismo
7.
Sci Rep ; 8(1): 15369, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30337622

RESUMEN

SWI/SNF or BAF chromatin-remodeling complexes are polymorphic assemblies of homologous subunit families that remodel nucleosomes and facilitate tissue-specific gene regulation during development. BAF57/SMARCE1 is a BAF complex subunit encoded in animals by a single gene and is a component of all mammalian BAF complexes. In vivo, the loss of SMARCE1 would lead to the formation of deficient combinations of the complex which might present limited remodeling activities. To address the specific contribution of SMARCE1 to the function of the BAF complex, we generated CRISPR/Cas9 mutations of smarce1 in zebrafish. Smarce1 mutants showed visible defects at 72 hpf, including smaller eyes, abnormal body curvature and heart abnormalities. Gene expression analysis revealed that the mutant embryos displayed defects in endocardial development since early stages, which led to the formation of a misshapen heart tube. The severe morphological and functional cardiac problems observed at 4 dpf were correlated with the substantially increased expression of different cardiac transcription factors. Additionally, we showed that Smarce1 binds to cis-regulatory regions of the gata5 gene and is necessary for the recruitment of the BAF complex to these regions.


Asunto(s)
Proteínas Cromosómicas no Histona , Endocardio/embriología , Regulación del Desarrollo de la Expresión Génica , Mutación , Factores de Transcripción , Proteínas de Pez Cebra , Pez Cebra , Animales , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Embrión no Mamífero/embriología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
8.
Dev Biol ; 421(1): 27-42, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27836552

RESUMEN

Zebrafish germ plasm is composed of mRNAs such as vasa and nanos and of proteins such as Bucky ball, all of which localize symmetrically in four aggregates at the distal region of the first two cleavage furrows. The coordination of actin microfilaments, microtubules and kinesin is essential for the correct localization of the germ plasm. Rho-GTPases, through their effectors, coordinate cytoskeletal dynamics. We address the participation of RhoA and its effector ROCK in germ plasm localization during the transition from two- to eight-cell embryos. We found that active RhoA is enriched along the cleavage furrow during the first two division cycles, whereas ROCK localizes at the distal region of the cleavage furrows in a similar pattern as the germ plasm mRNAs. Specific inhibition of RhoA and ROCK affected microtubules organization at the cleavage furrow; these caused the incorrect localization of the germ plasm mRNAs. The incorrect localization of the germ plasm led to a dramatic change in the number of germ cells during the blastula and 24hpf embryo stages without affecting any other developmental processes. We demonstrate that the Rho/ROCK pathway is intimately related to the determination of germ cells in zebrafish embryos.


Asunto(s)
Embrión no Mamífero/metabolismo , Transducción de Señal , Pez Cebra/embriología , Pez Cebra/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Desarrollo Embrionario/genética , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Microtúbulos/metabolismo , Miosinas/metabolismo , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteína de Unión al GTP rhoA/antagonistas & inhibidores
9.
FEBS Lett ; 590(11): 1555-69, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27149204

RESUMEN

Mammalian SWI/SNF or BAF chromatin-remodeling complexes are polymorphic assemblies of homologous subunit families that remodel nucleosomes. BAF57 is a subunit of the BAF complexes; it is encoded only in higher eukaryotes and is present in all mammalian assemblies. Its main structural feature is a high-mobility group domain, the DNA-binding properties of which suggest that BAF57 may play topological roles as the BAF complex enters or exits the nucleosome. BAF57 displays specific interactions with a number of proteins outside the BAF complex. Through these interactions, it can accomplish specific functions. In the embryo, BAF57 is responsible for the silencing of the CD4 gene during T-cell differentiation, and during the repression of neuronal genes in non-neuronal cells, BAF57 interacts with the transcriptional corepressor, Co-REST, and facilitates repression. Extensive work has demonstrated a specific role of BAF57 in regulating the interactions between BAF and nuclear hormone receptors. Despite its involvement in oncogenic pathways, new generation sequencing studies do not support a prominent role for BAF57 in the initiation of cancer. On the other hand, evidence has emerged to support a role for BAF57 as a metastasis factor, a prognosis marker and a therapeutic target. In humans, BAF57 is associated with disease, as mutations in this gene predispose to important congenital disorders, including menigioma disease or the Coffin-Siris syndrome. In this article, we present an exhaustive analysis of the BAF57 molecular and biochemical properties, cellular functions, loss-of-function phenotypes in living organisms and pathological manifestations in cases of human mutations.


Asunto(s)
Proteínas Cromosómicas no Histona/fisiología , Proteínas de Unión al ADN/fisiología , Enfermedad/genética , Crecimiento y Desarrollo/genética , Animales , Ensamble y Desensamble de Cromatina/genética , Humanos , Complejos Multiproteicos/fisiología , Subunidades de Proteína
10.
Gene Expr Patterns ; 19(1-2): 98-107, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26315538

RESUMEN

Antioxidant cellular mechanisms are essential for cell redox homeostasis during animal development and in adult life. Previous in situ hybridization analyses of antioxidant enzymes in zebrafish have indicated that they are ubiquitously expressed. However, spatial information about the protein distribution of these enzymes is not available. Zebrafish embryos are particularly suitable for this type of analysis due to their small size, transparency and fast development. The main objective of the present work was to analyze the spatial and temporal gene expression pattern of the two reported zebrafish glutathione peroxidase 4 (GPx4) genes during the first day of zebrafish embryo development. We found that the gpx4b gene shows maternal and zygotic gene expression in the embryo proper compared to gpx4a that showed zygotic gene expression in the periderm covering the yolk cell only. Following, we performed a GPx4 protein immunolocalization analysis during the first 24-h of development. The detection of this protein suggests that the antibody recognizes GPx4b in the embryo proper during the first 24 h of development and GPx4a at the periderm covering the yolk cell after 14-somite stage. Throughout early cleavages, GPx4 was located in blastomeres and was less abundant at the cleavage furrow. Later, from the 128-cell to 512-cell stages, GPx4 remained in the cytoplasm but gradually increased in the nuclei, beginning in marginal blastomeres and extending the nuclear localization to all blastomeres. During epiboly progression, GPx4b was found in blastoderm cells and was excluded from the yolk cell. After 24 h of development, GPx4b was present in the myotomes particularly in the slow muscle fibers, and was excluded from the myosepta. These results highlight the dynamics of the GPx4 localization pattern and suggest its potential participation in fundamental developmental processes.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Glutatión Peroxidasa/genética , Proteínas de Pez Cebra/genética , Animales , Blastodermo/metabolismo , Citocinesis , Embrión no Mamífero , Desarrollo Embrionario , Glutatión Peroxidasa/biosíntesis , Glutatión Peroxidasa/metabolismo , Hibridación in Situ , Isoenzimas , Mesodermo/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Pez Cebra , Proteínas de Pez Cebra/biosíntesis
11.
Dev Biol ; 403(1): 89-100, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25912688

RESUMEN

The Zmiz2 (Zimp7) protein and its homolog Zmiz1 (Zimp10) were initially identified in humans as androgen receptor co-activators. Sequence analysis revealed the presence of an SP-RING/Miz domain, which is highly conserved in members of the PIAS family and confers SUMO-conjugating activity. Zimp7 has been shown to interact with components of the Wnt/ß-Catenin signaling pathway and with Brg1 and BAF57, components of the ATP-dependent mammalian SWI/SNF-like BAF chromatin-remodeling complexes. In this work, we analyze the role of zygotic Zimp7 in zebrafish development. We describe evidence indicating that Zimp7 is required for mesoderm development and dorsoventral patterning. Morpholino-mediated reduction of zygotic Zimp7 produced axial mesodermal defects that were preceded by up-regulation of organizer genes such as bozozok, goosecoid and floating head at the onset of gastrulation and by down-regulation of the ventral markers vox, vent and eve1 indicating loss of the ventrolateral mesoderm. Consistently, embryos overexpressing zimp7 RNA exhibited midline defects such as loss of forebrain and cyclopia accompanied by transcriptional changes directly opposite of those found in the morphants. In addition, the patterning of ventralized embryos produced by the overexpression of vox and vent was restored by a reduction of Zimp7 activity. Altogether, our findings indicate that Zimp7 is involved in transcriptional regulation of factors that are essential for patterning in the dorsoventral axis.


Asunto(s)
Tipificación del Cuerpo/genética , Regulación del Desarrollo de la Expresión Génica , Organizadores Embrionarios/embriología , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Dedos de Zinc/genética , Animales , Blástula/metabolismo , Gastrulación/genética , Técnicas de Silenciamiento del Gen , Proteína Goosecoide/biosíntesis , Proteínas de Homeodominio/biosíntesis , Mesodermo/embriología , Morfolinos/genética , Proteínas Inhibidoras de STAT Activados/genética , ARN Mensajero/biosíntesis , Proteínas Represoras/biosíntesis , Transactivadores/genética , Factores de Transcripción/biosíntesis , Transcripción Genética/genética , Proteínas de Pez Cebra/biosíntesis , Proteínas de Pez Cebra/genética
12.
Anat Rec (Hoboken) ; 296(5): 759-73, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23554225

RESUMEN

Although cell proliferation is an essential cell behavior for animal development, a detailed analysis of spatial and temporal patterns of proliferation in whole embryos are still lacking for most model organisms. Zebrafish embryos are particularly suitable for this type of analysis due to their transparency and size. Therefore, the main objective of the present work was to analyze the spatial and temporal patterns of proliferation during the first day of zebrafish embryo development by indirect immunofluorescence against phosphorylated histone H3, a commonly used mitotic marker. Several interesting findings were established. First, we found that mitosis metasynchrony among blastomeres could begin at the 2- to 4-cell stage embryos. Second, mitosis synchrony was lost before the midblastula transition (MBT). Third, we observed a novel pattern of mitotic clusters that coincided in time with the mitotic pseudo "waves" described to occur before the MBT. Altogether, our findings indicate that early development is less synchronic than anticipated and that synchrony is not a requirement for proper development in zebrafish.


Asunto(s)
Proliferación Celular , Mitosis , Pez Cebra/embriología , Animales , Blastómeros/fisiología , Gástrula/citología , Índice Mitótico
13.
Cell Mol Life Sci ; 68(24): 4045-64, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21892772

RESUMEN

Sumoylation is a reversible post-translational modification that targets a variety of proteins mainly within the nucleus, but also in the plasma membrane and cytoplasm of the cell. It controls diverse cellular mechanisms such as subcellular localization, protein-protein interactions, or transcription factor activity. In recent years, the use of several developmental model systems has unraveled many critical functions for the sumoylation system in the early life of diverse species. In particular, detailed analyses of mutant organisms in both the components of the SUMO pathway and their targets have established the importance of the SUMO system in early developmental processes, such as cell division, cell lineage commitment, specification, and/or differentiation. In addition, an increasing number of developmental proteins, including transcription factors and epigenetic regulators, have been identified as sumoylation substrates. Sumoylation acts on these targets through various mechanisms. For example, this modification has been involved in converting a transcription factor from an activator to a repressor or in regulating the localization and/or stability of numerous transcription factors. This review will summarize current information on the function of sumoylation in embryonic development in different species from yeast to mammals.


Asunto(s)
Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/fisiología , Sumoilación/fisiología , Animales , Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Desarrollo Embrionario , Epigénesis Genética , Células Germinativas/metabolismo , Meiosis , Modelos Biológicos , Estabilidad Proteica , Transducción de Señal , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/química , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Enzimas Ubiquitina-Conjugadoras/fisiología , Xenopus
14.
Cell Tissue Res ; 345(1): 137-48, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21647563

RESUMEN

The mammalian SWI/SNF chromatin remodeling complexes play essential roles in cell cycle control through the transcriptional regulation of cell-cycle-specific genes. These complexes depend on the energy of ATP hydrolysis provided by the BRG1 or BRM catalytic subunit. They contain seven or more noncatalytic subunits, some being constitutive components, with others having paralogs that assemble in a combinatory manner producing different SWI/SNF-related complexes with specific functions. ARID1A and ARID1B are mutually exclusive subunits of the BAF complex. The specific presence of these subunits in the complex has been demonstrated to determine whether SWI/SNF functions as a corepressor (ARID1A) or as a coactivator (ARID1B) of the cell cycle genes. Our aim has been to analyze the relevance of the ARID1 subunits in development. We have compared the patterns of expression of these two genes through various mouse embryonic stages. Arid1a is expressed widely and intensively, whereas Arid1b is poorly transcribed and expressed in selected regions. Moreover, ARID1A and ARID1B present different kinetics of expression in the cell cycle. ARID1A accumulates in G0 and is downregulated throughout the cell cycle phases but is completely eliminated during mitosis, whereas ARID1B is expressed at comparable levels at all phases, even during mitosis. These kinetics probably affect the incorporation patterns of the ARID1 proteins to the complex and hence modulate SWI/SNF activity during proliferation and arrest.


Asunto(s)
Ciclo Celular , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Proteínas Nucleares/metabolismo , Subunidades de Proteína/metabolismo , Factores de Transcripción/metabolismo , Animales , Blastocisto/citología , Blastocisto/metabolismo , Ciclo Celular/genética , Línea Celular , Proteínas de Unión al ADN/genética , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Immunoblotting , Interfase , Ratones , Proteínas Nucleares/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Subunidades de Proteína/genética , Factores de Transcripción/genética , Ubiquitina/metabolismo
15.
Glia ; 59(4): 577-89, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21319224

RESUMEN

Bioactive lipids serve as intracellular and extracellular mediators in cell signaling in normal and pathological conditions. Here we describe that an important regulator of some of these lipids, the lipid phosphate phosphatase-3 (LPP3), is abundantly expressed in specific plasma membrane domains of Bergmann glia (BG), a specialized type of astrocyte with key roles in cerebellum development and physiology. Mice selectively lacking expression of LPP3/Ppap2b in the nervous system are viable and fertile but exhibit defects in postnatal cerebellum development and modifications in the cytoarchitecture and arrangement of BG with a mild non-progressive motor coordination defect. Lipid and gene profiling studies in combination with pharmacological treatments suggest that most of these effects are associated with alterations in sphingosine-1-phosphate (S1P) metabolism and signaling. Altogether our data indicate that LPP3 participates in several aspects of neuron-glia communication required for proper cerebellum development.


Asunto(s)
Astrocitos/metabolismo , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Lisofosfolípidos/metabolismo , Fosfatidato Fosfatasa/metabolismo , Transducción de Señal/fisiología , Esfingosina/análogos & derivados , Animales , Recuento de Células , Cerebelo/citología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Destreza Motora/fisiología , Técnicas de Cultivo de Órganos , Fosfatidato Fosfatasa/genética , Prueba de Desempeño de Rotación con Aceleración Constante , Esfingosina/metabolismo
16.
Gene Expr Patterns ; 10(1): 16-23, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19931425

RESUMEN

Members of the PIAS (protein inhibitor of activated STAT) family perform essential functions in modulating the activity of transcriptional regulators. Zimp7 and Zimp10 are two proteins that together form a subfamily of the PIAS. Like the other members of this family, they contain the zinc-binding SP-RING/Miz domain, which confers SUMO-conjugating activity. Both proteins have been shown to stimulate androgen receptor-mediated transcription. Previously, we reported that both Zimp7 and Zimp10 genes are extensively expressed and dynamically regulated in the developing mouse embryo. In this work, we investigated the expression of these genes during gonadal development. We found that their expression is sex-specific. Both genes initiate their transcription at early stages in the embryonic male gonad, reaching their peak at 13.5days post coitum, which coincides with the process of sex-specific germ cell mitotic arrest. Zimp7 is expressed in germ cells of the embryonic gonad and the adult testis. Immunofluorescence of spermatogenic cells revealed that Zimp7 protein localizes to nuclear territories in meiotic spermatocytes, including the XY bodies. On the other hand, Zimp10 is found in somatic cells, outside the testis cords and ceases to be expressed in the adult testis.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Gónadas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Caracteres Sexuales , Espermatogénesis/fisiología , Animales , Western Blotting , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Gónadas/embriología , Hibridación in Situ , Masculino , Ratones , Microscopía Confocal , Proteínas Inhibidoras de STAT Activados , Proteínas de Unión al ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermatocitos/metabolismo , Espermatogénesis/genética
17.
BMC Res Notes ; 2: 22, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-19284699

RESUMEN

BACKGROUND: Members of the Snail gene family, which encode zinc finger proteins that function as transcriptional repressors, play essential roles during embryonic development in vertebrates. Mouse embryos with conditional deletion of the Snail1 (Snai1) gene in the epiblast, but not in most extraembryonic membranes, exhibit defects in left-right asymmetry specification and migration of mesoderm cells through the posterior primitive streak. Here we describe phenotypic defects that result in death of the mutant embryos by 9.5 days of gestation. FINDINGS: Endothelial cells differentiated in epiblast-specific Snai1-deficient embryos, but formation of an interconnected vascular network was abnormal. To determine whether the observed vascular defects were dependent on disruption of blood flow, we analyzed vascular remodeling in cultured allantois explants from the mutant embryos. Similar vascular defects were observed in the mutant allantois explants. CONCLUSION: These studies demonstrate that lethality in the Snai1-conditional mutant embryos is caused by multiple defects in the cardiovascular system.

18.
Gene Expr Patterns ; 8(3): 206-13, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18053775

RESUMEN

ZIMP7 and ZIMP10 are two novel human PIAS-like proteins that share a similarity beyond the SP-RING Zn-finger domain that characterizes the PIAS family. This extended similarity is conserved in proteins of several other species and define an independent subfamily. ZIMP10 has been shown to increase the sumoylation of the androgen receptor (AR) leading to a stimulation of AR-mediated transcription. The Drosophila tonalli (tna) is the ortholog gene of ZIMP7 and ZIMP10 and presents genetic interactions with the SWI-SNF complex. Mutations in the tna gene produce flies with homeotic phenotypes. In this study, we determined the spatial-temporal expression pattern of Zimp7 and Zimp10 in mouse embryos from embryonic day 7.5 (E7.5), to mid-gestation. We found that these two genes are extensively expressed during these embryonic days and present partially overlapping patterns with a predomination of the transcripts in the neural tissues at early stages and a drop of expression at E12.5. Unlike other PIAS proteins, the tonalli-related Zimp genes might be essential for development. Comparison of conserved motifs in Zimp7 and Zimp10 protein sequences identified characteristic family domains that might be related to their specific biological roles, besides their common role previously identified in the sumoylation pathway.


Asunto(s)
Perfilación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Especificidad de Órganos/genética , Proteínas Inhibidoras de STAT Activados/genética , Secuencia de Aminoácidos , Animales , Drosophila melanogaster/genética , Evolución Molecular , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Datos de Secuencia Molecular , Familia de Multigenes , Proteínas Inhibidoras de STAT Activados/biosíntesis , Proteínas de Unión al ARN , Factores de Tiempo , Dedos de Zinc/genética
19.
Dev Dyn ; 233(1): 29-40, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15736269

RESUMEN

Kit is a receptor tyrosine kinase that plays a fundamental role during the development of germ cells. Additionally, a truncated product, tr-kit, expressed in haploid spermatids and mature spermatozoa can induce parthenogenetic activation when microinjected into mouse eggs, through the activation of PLCgamma-1. In this work, we induced ectopic expression of a mutated Kit protein, Kit(D814Y) during germ cell development. The in vivo expression of this mutant in spermatids produced malformations in mature spermatozoa, and in the most severe cases, sterility. Ultrastructural analysis indicated that condensing spermatids in the transgenic mouse presented a mislocalization of the manchette; a structure that has a crucial role during the elongation steps of spermiogenesis. This morphogenetic phenotype was accompanied by an increased phosphorylation of PLCgamma-1 in spermatogenic cells. Interestingly, we also found that, in wild-type testis, PLCgamma-1 is specifically phosphorylated in condensing spermatids, coincident with the timing of expression of tr-kit in spermiogenesis. We propose that alterations of PLCgamma-1 activity artificially promoted by ectopic Kit(D814Y) expression are related to the abnormalities of spermiogenesis. Our observations suggest that PLCgamma-1 activity could be involved in the shaping of spermatozoa.


Asunto(s)
Proteínas Proto-Oncogénicas c-kit/genética , Espermátides/metabolismo , Espermatogénesis/genética , Animales , Regulación de la Expresión Génica/fisiología , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Integrasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Transgénicos , Mutación , Fosfolipasa C gamma , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Espermátides/citología , Espermatogénesis/fisiología , Espermatozoides/citología , Espermatozoides/fisiología , Fosfolipasas de Tipo C/metabolismo
20.
Dev Dyn ; 232(1): 180-90, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15580630

RESUMEN

Oct4 is a transcription factor that has been associated with pluripotency and fate determination in the initial cell lineages of mammals. On the other hand, Pou2, the ortholog of Oct4 in zebrafish, serves additional later functions during brain development acting as a differentiation switch. In mice, Oct4 is expressed throughout the neural plate of embryos until embryonic day (E) 8.0. In this study, we produced transgenic mouse embryos that ubiquitously express Oct4 and analyzed the consequences during development. We show that, at E8.0, a higher dosage of Oct4 in the neuroectoderm is sufficient to transiently alter mid-hindbrain patterning and produced a strong up-regulation of Pax2, indicating that Oct4 can regulate this gene in vivo. After E9.5, ectopic Oct4 in this region produced cell death and affected the development of the forebrain, suggesting that, at these later stages, Oct4 down-regulation is necessary for normal development to proceed. The phenotype of the transgenic embryos was also accompanied with an increase of Fgf8 expression in several of its endogenous domains, suggesting the possibility that Oct4 can participate in the regulation of expression of this ligand. Our observations support the hypothesis that Oct4, like zebrafish Pou2, has a conserved function during early brain patterning in mouse.


Asunto(s)
Encéfalo/embriología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Rombencéfalo/embriología , Rombencéfalo/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Animales , Western Blotting , Tipificación del Cuerpo , Muerte Celular , Diferenciación Celular , Linaje de la Célula , Regulación hacia Abajo , Factor 8 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/biosíntesis , Genotipo , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Ligandos , Ratones , Ratones Transgénicos , Microscopía Electrónica de Rastreo , Modelos Genéticos , Datos de Secuencia Molecular , Factor 3 de Transcripción de Unión a Octámeros , Factor de Transcripción PAX2 , Fenotipo , Estructura Terciaria de Proteína , Recombinación Genética , Factores de Tiempo , Transcripción Genética , Transgenes , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA