Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biology (Basel) ; 11(10)2022 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-36290313

RESUMEN

During the first steps of sea urchin development, fertilization elicits a marked increase in protein synthesis essential for subsequent cell divisions. While the translation of mitotic cyclin mRNAs is crucial, we hypothesized that additional mRNAs must be translated to finely regulate the onset into mitosis. One of the maternal mRNAs recruited onto active polysomes at this stage codes for the initiation factor eIF4B. Here, we show that the sea urchin eIF4B orthologs present the four specific domains essential for eIF4B function and that Paracentrotus lividus eIF4B copurifies with eIF4E in a heterologous system. In addition, we investigated the role of eIF4B mRNA de novo translation during the two first embryonic divisions of two species, P. lividus and Sphaerechinus granularis. Our results show that injection of a morpholino directed against eIF4B mRNA results in a downregulation of translational activity and delays cell division in these two echinoids. Conversely, injection of an mRNA encoding for P. lividus eIF4B stimulates translation and significantly accelerates cleavage rates. Taken together, our findings suggest that eIF4B mRNA de novo translation participates in a conserved regulatory loop that contributes to orchestrating protein synthesis and modulates cell division rhythm during early sea urchin development.

2.
Cells ; 9(4)2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32272587

RESUMEN

The sea urchin embryo provides a valuable system to analyse the molecular mechanisms orchestrating cell cycle progression and mitosis in a developmental context. However, although it is known that the regulation of histone activity by post-translational modification plays an important role during cell division, the dynamics and the impact of these modifications have not been characterised in detail in a developing embryo. Using different immuno-detection techniques, we show that the levels of Histone 3 phosphorylation at Threonine 3 oscillate in synchrony with mitosis in Sphaerechinus granularis early embryos. We present, in addition, the results of a pharmacological study aimed at analysing the role of this key histone post-translational modification during sea urchin early development.


Asunto(s)
Erizos de Mar/metabolismo , Animales , División Celular/fisiología , Histonas/metabolismo , Mitosis/fisiología , Fosforilación , Erizos de Mar/citología , Erizos de Mar/embriología
3.
PLoS One ; 12(10): e0185897, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28977027

RESUMEN

The maintenance of paracellular barriers in invertebrate epithelia depends on the integrity of specific cell adhesion structures known as septate junctions (SJ). Multiple studies in Drosophila have revealed that these junctions have a stereotyped architecture resulting from the association in the lateral membrane of a large number of components. However, little is known about the dynamic organisation adopted by these multi-protein complexes in living tissues. We have used live imaging techniques to show that the Ly6 protein Boudin is a component of these adhesion junctions and can diffuse systemically to associate with the SJ of distant cells. We also observe that this protein and the claudin Kune-kune are endocytosed in epidermal cells during embryogenesis. Our data reveal that the SJ contain a set of components exhibiting a high membrane turnover, a feature that could contribute in a tissue-specific manner to the morphogenetic plasticity of these adhesion structures.


Asunto(s)
Uniones Adherentes/metabolismo , Proteínas de Drosophila/metabolismo , Endocitosis , Proteínas de la Membrana/metabolismo , Animales , Membrana Celular/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Morfogénesis , Transporte de Proteínas
4.
Mol Biol Evol ; 34(12): 3132-3147, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28961967

RESUMEN

Pairs of duplicated genes generally display a combination of conserved expression patterns inherited from their unduplicated ancestor and newly acquired domains. However, how the cis-regulatory architecture of duplicated loci evolves to produce these expression patterns is poorly understood. We have directly examined the gene-regulatory evolution of two tandem duplicates, the Drosophila Ly6 genes CG9336 and CG9338, which arose at the base of the drosophilids between 40 and 60 Ma. Comparing the expression patterns of the two paralogs in four Drosophila species with that of the unduplicated ortholog in the tephritid Ceratitis capitata, we show that they diverged from each other as well as from the unduplicated ortholog. Moreover, the expression divergence appears to have occurred close to the duplication event and also more recently in a lineage-specific manner. The comparison of the tissue-specific cis-regulatory modules (CRMs) controlling the paralog expression in the four Drosophila species indicates that diverse cis-regulatory mechanisms, including the novel tissue-specific enhancers, differential inactivation, and enhancer sharing, contributed to the expression evolution. Our analysis also reveals a surprisingly variable cis-regulatory architecture, in which the CRMs driving conserved expression domains change in number, location, and specificity. Altogether, this study provides a detailed historical account that uncovers a highly dynamic picture of how the paralog expression patterns and their underlying cis-regulatory landscape evolve. We argue that our findings will encourage studying cis-regulatory evolution at the whole-locus level to understand how interactions between enhancers and other regulatory levels shape the evolution of gene expression.


Asunto(s)
Proteínas de Drosophila/genética , Elementos de Facilitación Genéticos/genética , Duplicación de Gen/genética , Animales , Drosophila melanogaster/genética , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica/genética , Genes Duplicados/genética , Filogenia , Análisis de Secuencia de Proteína , Especificidad de la Especie
5.
Mol Biol Evol ; 32(7): 1730-47, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25743545

RESUMEN

Gene families often consist of members with diverse expression domains reflecting their functions in a wide variety of tissues. However, how the expression of individual members, and thus their tissue-specific functions, diversified during the course of gene family expansion is not well understood. In this study, we approached this question through the analysis of the duplication history and transcriptional evolution of a rapidly expanding subfamily of insect Ly6 genes. We analyzed different insect genomes and identified seven Ly6 genes that have originated from a single ancestor through sequential duplication within the higher Diptera. We then determined how the original embryonic expression pattern of the founding gene diversified by characterizing its tissue-specific expression in the beetle Tribolium castaneum, the butterfly Bicyclus anynana, and the mosquito Anopheles stephensi and those of its duplicates in three higher dipteran species, representing various stages of the duplication history (Megaselia abdita, Ceratitis capitata, and Drosophila melanogaster). Our results revealed that frequent neofunctionalization episodes contributed to the increased expression breadth of this subfamily and that these events occurred after duplication and speciation events at comparable frequencies. In addition, at each duplication node, we consistently found asymmetric expression divergence. One paralog inherited most of the tissue-specificities of the founder gene, whereas the other paralog evolved drastically reduced expression domains. Our approach attests to the power of combining a well-established duplication history with a comprehensive coverage of representative species in acquiring unequivocal information about the dynamics of gene expression evolution in gene families.


Asunto(s)
Regulación de la Expresión Génica , Genes de Insecto , Insectos/genética , Familia de Multigenes , Animales , Embrión no Mamífero/metabolismo , Evolución Molecular , Duplicación de Gen , Perfilación de la Expresión Génica , Insectos/embriología , Especificidad de Órganos/genética , Filogenia , Especificidad de la Especie
6.
Nature ; 479(7374): 487-92, 2011 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-22113690

RESUMEN

The spider mite Tetranychus urticae is a cosmopolitan agricultural pest with an extensive host plant range and an extreme record of pesticide resistance. Here we present the completely sequenced and annotated spider mite genome, representing the first complete chelicerate genome. At 90 megabases T. urticae has the smallest sequenced arthropod genome. Compared with other arthropods, the spider mite genome shows unique changes in the hormonal environment and organization of the Hox complex, and also reveals evolutionary innovation of silk production. We find strong signatures of polyphagy and detoxification in gene families associated with feeding on different hosts and in new gene families acquired by lateral gene transfer. Deep transcriptome analysis of mites feeding on different plants shows how this pest responds to a changing host environment. The T. urticae genome thus offers new insights into arthropod evolution and plant-herbivore interactions, and provides unique opportunities for developing novel plant protection strategies.


Asunto(s)
Adaptación Fisiológica/genética , Genoma/genética , Herbivoria/genética , Tetranychidae/genética , Tetranychidae/fisiología , Adaptación Fisiológica/fisiología , Animales , Ecdisterona/análogos & derivados , Ecdisterona/genética , Evolución Molecular , Fibroínas/genética , Regulación de la Expresión Génica , Transferencia de Gen Horizontal/genética , Genes Homeobox/genética , Genómica , Herbivoria/fisiología , Datos de Secuencia Molecular , Muda/genética , Familia de Multigenes/genética , Nanoestructuras/química , Plantas/parasitología , Seda/biosíntesis , Seda/química , Transcriptoma/genética
7.
Prog Mol Biol Transl Sci ; 100: 51-82, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21377624

RESUMEN

The basic cellular processes deregulated during carcinogenesis and the vast majority of the genes implicated in cancer appear conserved from humans to flies. This conservation, together with an ever-expanding fly genetic toolbox, has made of Drosophila melanogaster a remarkably profitable model to study many fundamental aspects of carcinogenesis. In particular, Drosophila has played a major role in the identification of genes and pathways implicated in cancer and in disclosing novel functional relationships between cancer genes. It has also proved to be a genetically tractable system where to mimic cancer-like situations and characterize the mode of action of human oncogenes. Here, we outline some advances in the study of cancer, both at the basic and more translational levels, which have benefited from research carried out in flies.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila melanogaster/fisiología , Neoplasias/patología , Animales , Diferenciación Celular , Polaridad Celular , Proliferación Celular
8.
PLoS One ; 6(3): e17763, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21423573

RESUMEN

BACKGROUND: Genetic analysis of the Drosophila septate junctions has greatly contributed to our understanding of the mechanisms controlling the assembly of these adhesion structures, which bear strong similarities with the vertebrate tight junctions and the paranodal septate junctions. These adhesion complexes share conserved molecular components and have a common function: the formation of paracellular barriers restraining the diffusion of solutes through epithelial and glial envelopes. METHODOLOGY/PRINCIPAL FINDINGS: In this work we characterise the function of the Drosophila cold gene, that codes for a protein belonging to the Ly6 superfamily of extracellular ligands. Analysis of cold mutants shows that this gene is specifically required for the organisation of the septate junctions in epithelial tissues and in the nervous system, where its contribution is essential for the maintenance of the blood-brain barrier. We show that cold acts in a cell autonomous way, and we present evidence indicating that this protein could act as a septate junction component. CONCLUSION/SIGNIFICANCE: We discuss the specific roles of cold and three other Drosophila members of the Ly6 superfamily that have been shown to participate in a non-redundant way in the process of septate junction assembly. We propose that vertebrate Ly6 proteins could fulfill analogous roles in tight junctions and/or paranodal septate junctions.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Uniones Estrechas/metabolismo , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Ectodermo/citología , Ectodermo/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Genes de Insecto/genética , Larva/citología , Larva/metabolismo , Datos de Secuencia Molecular , Morfogénesis , Neuroglía/citología , Neuroglía/metabolismo , Fenotipo , Transporte de Proteínas , Glándulas Salivales/citología , Glándulas Salivales/metabolismo , Fracciones Subcelulares/metabolismo , Tráquea/citología , Tráquea/embriología , Tráquea/metabolismo , Alas de Animales/citología , Alas de Animales/metabolismo
9.
BMC Dev Biol ; 10: 65, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20540764

RESUMEN

BACKGROUND: In metazoans, the hematopoietic system plays a key role both in normal development and in defense of the organism. In Drosophila, the cellular immune response involves three types of blood cells: plasmatocytes, crystal cells and lamellocytes. This last cell type is barely present in healthy larvae, but its production is strongly induced upon wasp parasitization or in mutant contexts affecting larval blood cell homeostasis. Notably, several zygotic mutations leading to melanotic mass (or "tumor") formation in larvae have been associated to the deregulated differentiation of lamellocytes. To gain further insights into the gene regulatory network and the mechanisms controlling larval blood cell homeostasis, we conducted a tissue-specific loss of function screen using hemocyte-specific Gal4 drivers and UAS-dsRNA transgenic lines. RESULTS: By targeting around 10% of the Drosophila genes, this in vivo RNA interference screen allowed us to recover 59 melanotic tumor suppressor genes. In line with previous studies, we show that melanotic tumor formation is associated with the precocious differentiation of stem-cell like blood progenitors in the larval hematopoietic organ (the lymph gland) and the spurious differentiation of lamellocytes. We also find that melanotic tumor formation can be elicited by defects either in the fat body, the embryo-derived hemocytes or the lymph gland. In addition, we provide a definitive confirmation that lymph gland is not the only source of lamellocytes as embryo-derived plasmatocytes can differentiate into lamellocytes either upon wasp infection or upon loss of function of the Friend of GATA cofactor U-shaped. CONCLUSIONS: In this study, we identify 55 genes whose function had not been linked to blood cell development or function before in Drosophila. Moreover our analyses reveal an unanticipated plasticity of embryo-derived plasmatocytes, thereby shedding new light on blood cell lineage relationship, and pinpoint the Friend of GATA transcription cofactor U-shaped as a key regulator of the plasmatocyte to lamellocyte transformation.


Asunto(s)
Drosophila melanogaster/citología , Drosophila melanogaster/genética , Redes Reguladoras de Genes , Homeostasis , Animales , Drosophila melanogaster/inmunología , Drosophila melanogaster/metabolismo , Genes Supresores de Tumor , Hematopoyesis , Hemocitos/citología , Hemocitos/inmunología , Interferencia de ARN
10.
J Cell Sci ; 123(Pt 12): 2058-67, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20519583

RESUMEN

The ezrin, radixin and moesin (ERM) proteins regulate cell membrane architecture in several cellular contexts. Current models propose that ERM activation requires a PtdIns(4,5)P(2)-induced conformational change, followed by phosphorylation of a conserved threonine. However, how these inputs contribute in vivo to orchestrate ERM activation is poorly understood. We addressed this issue by evaluating the contribution of PtdIns(4,5)P(2) and phosphorylation to the regulation of moesin during Drosophila development. Unexpectedly, we found that a form of moesin that cannot be phosphorylated displayed significant activity and could substitute for the endogenous product during wing morphogenesis. By contrast, we also show that PtdIns(4,5)P(2) binding is essential for moesin recruitment to the membrane and for its subsequent phosphorylation. Our data indicate that PtdIns(4,5)P(2) acts as a dosing mechanism that locally regulates ERM membrane recruitment and activation, whereas cycles of phosphorylation and dephosphorylation further control their activity once they have reached the cell cortex.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/crecimiento & desarrollo , Proteínas de Microfilamentos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Secuencia de Aminoácidos , Animales , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Drosophila/química , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Transporte de Proteínas , Alineación de Secuencia , Alas de Animales/química , Alas de Animales/crecimiento & desarrollo , Alas de Animales/metabolismo
11.
Development ; 136(13): 2199-209, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19502482

RESUMEN

The Ly6 superfamily, present in most metazoan genomes, codes for different cell-surface proteins and secreted ligands containing an extracellular motif called a Ly6 domain or three-finger domain. We report the identification of 36 novel genes coding for proteins of this family in Drosophila. One of these fly Ly6 proteins, coded by the gene boudin (bou), is essential for tracheal morphogenesis in the fly embryo and contributes to the maintenance of the paracellular barrier and the organisation of the septate junctions in this tissue. Bou, a glycosylphosphatidylinositol anchored membrane protein, is also required for septate junction organisation in epithelial tissues and in the chordotonal organ glial cells, but not in the central nervous system. Our study reveals interesting parallelisms between the Ly6 proteins of flies and vertebrates, such as the CD59 antigen. Similarly to this human protein, Bou travels from cell to cell associated with extracellular particles and, consistently, we show that it is required in a non-cell-autonomous fashion. Our work opens the way for future studies addressing the function of Ly6 proteins using Drosophila as a model system.


Asunto(s)
Antígenos Ly/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Uniones Intercelulares/metabolismo , Proteínas de la Membrana/metabolismo , Tráquea/embriología , Secuencia de Aminoácidos , Animales , Antígenos Ly/genética , Barrera Hematoencefálica/embriología , Barrera Hematoencefálica/ultraestructura , Membrana Celular/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Humanos , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Morfogénesis/fisiología , Tejido Nervioso/anatomía & histología , Tejido Nervioso/embriología , Alineación de Secuencia , Tráquea/anatomía & histología
12.
PLoS Biol ; 4(9): e290, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16933974

RESUMEN

It is well established that developmental programs act during embryogenesis to determine animal morphogenesis. How these developmental cues produce specific cell shape during morphogenesis, however, has remained elusive. We addressed this question by studying the morphological differentiation of the Drosophila epidermis, governed by a well-known circuit of regulators leading to a stereotyped pattern of smooth cells and cells forming actin-rich extensions (trichomes). It was shown that the transcription factor Shavenbaby plays a pivotal role in the formation of trichomes and underlies all examined cases of the evolutionary diversification of their pattern. To gain insight into the mechanisms of morphological differentiation, we sought to identify shavenbaby's downstream targets. We show here that Shavenbaby controls epidermal cell shape, through the transcriptional activation of different classes of cellular effectors, directly contributing to the organization of actin filaments, regulation of the extracellular matrix, and modification of the cuticle. Individual inactivation of shavenbaby's targets produces distinct trichome defects and only their simultaneous inactivation prevent trichome formation. Our data show that shavenbaby governs an evolutionarily conserved developmental module consisting of a set of genes collectively responsible for trichome formation, shedding new light on molecular mechanisms acting during morphogenesis and the way they can influence evolution of animal forms.


Asunto(s)
Tipificación del Cuerpo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/citología , Drosophila/embriología , Células Epidérmicas , Factores de Transcripción/metabolismo , Animales , Animales Modificados Genéticamente , Forma de la Célula , Proteínas de Unión al ADN/genética , Drosophila/anatomía & histología , Drosophila/genética , Proteínas de Drosophila/genética , Epidermis/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Pigmentación/genética , Factores de Transcripción/genética , Transcripción Genética
13.
J Cell Sci ; 116(Pt 7): 1199-207, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12615963

RESUMEN

We have characterised the function of two Drosophila genes, miniature and dusky, that are required for the morphological reorganisation of the apical membrane during wing epidermis differentiation. These genes encode transmembrane proteins containing a ZP (zona pellucida) domain and are homologous to several vertebrate and invertebrate apical matrix components. miniature and dusky are only expressed in tissues secreting a cuticle, and the Min protein localises to the apical membrane during the early stages of cuticle formation. We propose that Min and Dusky form a novel subfamily within the ZP domain proteins and are specifically involved in the interactions between the apical membrane, the cytoskeleton and the forming cuticle.


Asunto(s)
Citoesqueleto/metabolismo , Proteínas de Drosophila/deficiencia , Drosophila melanogaster/crecimiento & desarrollo , Proteínas de la Membrana/deficiencia , Alas de Animales/crecimiento & desarrollo , Secuencia de Aminoácidos/genética , Animales , Secuencia de Bases/genética , Citoesqueleto/genética , Citoesqueleto/ultraestructura , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestructura , Epidermis/crecimiento & desarrollo , Epidermis/metabolismo , Epidermis/ultraestructura , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Larva/crecimiento & desarrollo , Larva/metabolismo , Larva/ultraestructura , Proteínas de la Membrana/genética , Metamorfosis Biológica/genética , Microscopía Electrónica , Microscopía Electrónica de Rastreo , Datos de Secuencia Molecular , Morfogénesis , Mutación/fisiología , Estructura Terciaria de Proteína/genética , Alas de Animales/metabolismo , Alas de Animales/ultraestructura
14.
Development ; 129(4): 993-1002, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11861482

RESUMEN

Localised activation of the Ras/Raf pathway by Epidermal Growth Factor Receptor (EGFR) signalling specifies the formation of veins in the Drosophila wing. However, little is known about how the EGFR signal regulates transcriptional responses during the vein/intervein cell fate decision. We provide evidence that EGFR signalling induces expression of vein-specific genes by inhibiting the Capicua (Cic) HMG-box repressor, a known regulator of embryonic body patterning. Lack of Cic function causes ectopic expression of EGFR targets such as argos, ventral veinless and decapentaplegic and leads to formation of extra vein tissue. In vein cells, EGFR signalling downregulates Cic protein levels in the nucleus and relieves repression of vein-specific genes, whereas intervein cells maintain high levels of Cic throughout larval and pupal development, repressing the expression of vein-specific genes and allowing intervein differentiation. However, regulation of some EGFR targets such as rhomboid appears not to be under direct control of Cic, suggesting that EGFR signalling branches out in the nucleus and controls different targets via distinct mediator factors. Our results support the idea that localised inactivation of transcriptional repressors such as Cic is a rather general mechanism for regulation of target gene expression by the Ras/Raf pathway.


Asunto(s)
Proteínas de Drosophila , Receptores ErbB/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Animales , Drosophila/genética , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Genes de Insecto , Proteínas HMGB , Proteínas del Grupo de Alta Movilidad/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Procesamiento Postranscripcional del ARN , Proteínas Represoras/genética , Factores de Transcripción/genética , Alas de Animales/crecimiento & desarrollo , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...