Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Biol Evol ; 33(10): 2621-32, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27401231

RESUMEN

Secreted ligands in the Dpp/BMP family drive dorsal-ventral (D/V) axis formation in all Bilaterian species. However, maternal factors regulating Dpp/BMP transcription in this process are largely unknown. We identified the BTB domain protein longitudinals lacking-like (lolal) as a modifier of decapentaplegic (dpp) mutations. We show that Lolal is evolutionarily related to the Trithorax group of chromatin regulators and that lolal interacts genetically with the epigenetic factor Trithorax-like during Dpp D/V signaling. Maternally driven Lolal(HA) is found in oocytes and translocates to zygotic nuclei prior to the point at which dpp transcription begins. lolal maternal and zygotic mutant embryos display significant reductions in dpp, pMad, and zerknullt expression, but they are never absent. The data suggest that lolal is required to maintain dpp transcription during D/V patterning. Phylogenetic data revealed that lolal is an evolutionarily new gene present only in insects and crustaceans. We conclude that Lolal is the first maternal protein identified with a role in dpp D/V transcriptional maintenance, that Lolal and the epigenetic protein Trithorax-like are essential for Dpp D/V signaling and that the architecture of the Dpp D/V pathway evolved in the arthropod lineage after the separation from vertebrates via the incorporation of new genes such as lolal.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Factores de Transcripción/genética , Animales , Evolución Biológica , Tipificación del Cuerpo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Epigenómica , Femenino , Masculino , Mutación , Fenotipo , Filogenia , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Vertebrados/embriología , Vertebrados/metabolismo
2.
PLoS One ; 6(2): e16799, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21364917

RESUMEN

Mutations in the human mitochondrial elongation factor G1 (EF-G1) are recessive lethal and cause death shortly after birth. We have isolated mutations in iconoclast (ico), which encodes the highly conserved Drosophila orthologue of EF-G1. We find that EF-G1 is essential during fly development, but its function is not required in every tissue. In contrast to null mutations, missense mutations exhibit stronger, possibly neomorphic phenotypes that lead to premature death during embryogenesis. Our experiments show that EF-G1 contains a secondary C-terminal nuclear localization signal. Expression of missense mutant forms of EF-G1 can accumulate in the nucleus and cause growth and patterning defects and animal lethality. We find that transgenes that encode mutant human EF-G1 proteins can rescue ico mutants, indicating that the underlying problem of the human disease is not just the loss of enzymatic activity. Our results are consistent with a model where EF-G1 acts as a retrograde signal from mitochondria to the nucleus to slow down cell proliferation if mitochondrial energy output is low.


Asunto(s)
Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Drosophila/embriología , Drosophila/genética , Señales de Localización Nuclear , Factor G de Elongación Peptídica/química , Factor G de Elongación Peptídica/genética , Factor G de Elongación Peptídica/fisiología , Factores de Elongación de Péptidos/química , Factores de Elongación de Péptidos/fisiología , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Regulación hacia Abajo/genética , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Proteínas de Drosophila/genética , Embrión no Mamífero , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Humanos , Masculino , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/fisiología , Datos de Secuencia Molecular , Señales de Localización Nuclear/química , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/fisiología , Factor G de Elongación Peptídica/metabolismo , Factores de Elongación de Péptidos/genética , Factores de Elongación de Péptidos/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal/genética
3.
Mech Dev ; 127(7-8): 358-70, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20381612

RESUMEN

The goal of this report is to elucidate the contributions of the Drosophila TGF-beta type I receptors TKV and SAX to the activity gradient formed by the two BMP family members DPP and GBB that play important roles in growth and patterning of imaginal discs. Binding studies display preferential interactions of DPP and GBB with homodimers of TKV or SAX, respectively, but also low affinities of both ligands to heterodimers. Inside the cell, constitutively activated forms of both TKV and SAX can ectopically phosphorylate the SMAD transcription factor MAD. However, MAD phosphorylated by homodimers of activated SAX or certain mutant forms of TKV localizes to the nucleus without changing the expression of downstream genes. Differences in signaling between SAX and TKV can be localized to amino acid residues within an area that has been shown to influence complexes formation between type I and type II receptors. The finding that the type II receptor PUT but not activated forms of SAX can enhance signaling of a pseudo-activated MAD-SDVD, which cannot be phosphorylated at the C-terminus, suggests a model, where activation of SMADs requires the presence of type II receptors and a second activation step in addition to C-terminal phosphorylation. Complete activation of MAD can only occur in tetrameric complexes of type II receptors in combination with SAX-TKV heterodimers or TKV homodimers but not SAX homodimers. Since TKV is not distributed equally in wing discs, heterodimers of SAX and TKV play an important role in extending the BMP activity gradient by facilitating DPP diffusion and assisting GBB signaling through functional complexes with type II receptors.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Smad/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Drosophila/química , Prueba de Complementación Genética , Ligandos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Fosforilación , Unión Proteica , Multimerización de Proteína , Proteínas Serina-Treonina Quinasas/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Superficie Celular/química , Receptores de Factores de Crecimiento Transformadores beta/química , Transducción de Señal , Alas de Animales/metabolismo
4.
Mech Dev ; 125(1-2): 117-29, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18077144

RESUMEN

Follistatin (FS) is one of several secreted proteins that modulate the activity of TGF-beta family members during development. The structural and functional analysis of Drosophila Follistatin (dFS) reveals important differences between dFS and its vertebrate orthologues: it is larger, more positively charged, and proteolytically processed. dFS primarily inhibits signaling of Drosophila Activin (dACT) but can also inhibit other ligands like Decapentaplegic (DPP). In contrast, the presence of dFS enhances signaling of the Activin-like protein Dawdle (DAW), indicating that dFS exhibits a dual function in promoting and inhibiting signaling of TGF-beta ligands. In addition, FS proteins may also function in facilitating ligand diffusion. We find that mutants of daw are rescued in significant numbers by expression of vertebrate FS proteins. Since two PiggyBac insertions in dfs are not lethal, it appears that the function of dFS is non-essential or functionally redundant.


Asunto(s)
Folistatina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Drosophila , Folistatina/química , Folistatina/genética , Hidrólisis , Inmunohistoquímica , Hibridación in Situ , Ligandos , Fenotipo , Unión Proteica , Conformación Proteica
5.
Fly (Austin) ; 1(4): 212-21, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18820452

RESUMEN

Growth factors of the TGF-beta family signal through type I/II receptor complexes that phosphorylate SMAD transcription factors. In this study, we analyzed signaling of all seven TGF-beta members to identify those that mediate growth through the Drosophila type I receptor BABO. We find that two potential ligands of BABO, Myoglianin (MYO) and Maverick (MAV), do not activate dSMAD2. Only Drosophila Activin (dACT) and the Activin-like ligand Dawdle (DAW) signal through BABO in combination with the type II receptor PUNT and activate dSMAD2. Surprisingly, we find that activation of BABO can also lead to the phosphorylation of the "BMP-specific" MAD. In wing discs, expression of an activated form of dSMAD2 promotes growth similar to dACT and activated BABO. By itself, activated dSMAD2 does not affect DPP/GBB target genes. However, coexpression of activated forms of dSMAD2 and MAD additively induces the expression of spalt. In contrast to dACT, we find that DAW does not promote growth when expressed in wings. In fact, coexpression of DAW with MAD or dSMAD2 decreases growth. daw mutants die primarily during larval stages and exhibit anal pad phenotypes reminiscent of babo mutants. The rescue of daw mutants by restricted expression in neuroendocrine cells indicates that Activin-type ligands are likely distributed through the endocrine system. The distinct signaling of dACT, DAW and MYO through BABO suggests the existence of co-receptors that modulate the canonical SMAD pathway.


Asunto(s)
Activinas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Receptores de Activinas/metabolismo , Animales , Proteínas de Unión al ADN/metabolismo , Drosophila melanogaster/embriología , Proteína Smad2/metabolismo , Factores de Transcripción/metabolismo , Alas de Animales/embriología , Alas de Animales/metabolismo
6.
Cell ; 112(3): 303-15, 2003 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-12581521

RESUMEN

Metamorphosis of the Drosophila brain involves pruning of many larval-specific dendrites and axons followed by outgrowth of adult-specific processes. From a genetic mosaic screen, we recovered two independent mutations that block neuronal remodeling in the mushroom bodies (MBs). These phenotypically indistinguishable mutations affect Baboon function, a Drosophila TGF-beta/activin type I receptor, and dSmad2, its downstream transcriptional effector. We also show that Punt and Wit, two type II receptors, act redundantly in this process. In addition, knocking out dActivin around the mid-third instar stage interferes with remodeling. Binding of the insect steroid hormone ecdysone to distinct ecdysone receptor isoforms induces different metamorphic responses in various larval tissues. Interestingly, expression of the ecdysone receptor B1 isoform (EcR-B1) is reduced in activin pathway mutants, and restoring EcR-B1 expression significantly rescues remodeling defects. We conclude that the Drosophila Activin signaling pathway mediates neuronal remodeling in part by regulating EcR-B1 expression.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Diferenciación Celular/genética , Drosophila/crecimiento & desarrollo , Vías Nerviosas/crecimiento & desarrollo , Plasticidad Neuronal/genética , Proteínas , Receptores de Esteroides/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Receptores de Activinas Tipo I/deficiencia , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo II/deficiencia , Receptores de Activinas Tipo II/genética , Animales , Encéfalo/citología , Encéfalo/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Drosophila/citología , Drosophila/metabolismo , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Femenino , Genes Letales/genética , Larva/citología , Larva/crecimiento & desarrollo , Larva/metabolismo , Masculino , Cuerpos Pedunculados/citología , Cuerpos Pedunculados/crecimiento & desarrollo , Cuerpos Pedunculados/metabolismo , Mutación Missense/genética , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptores de Esteroides/genética , Transducción de Señal/genética , Proteína Smad2 , Transactivadores/deficiencia , Transactivadores/genética
7.
Gene ; 291(1-2): 85-93, 2002 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-12095682

RESUMEN

With the genomic sequence of multicellular organisms such as Caenorhabditis elegans, Drosophila melanogaster, and Homo sapiens completed and others to be finished in the near future, the focus has shifted from accumulating sequence information to the prediction and analysis of genes within the completed genomes. Unfortunately, presently available computer programs do not always accurately predict gene structure such as mRNA and translation start sites or intron/exon boundaries. The only way to be certain about a gene's structure is to isolate and characterize its cDNA. Since the screening of libraries is a time-consuming, labor-intensive process that sometimes fails to yield the desired clone, we searched for faster, more efficient ways to isolate cDNAs. In this study, we describe two methods for amplification and isolation of cDNAs from plasmid libraries that requires no hybridization (MACH). With the polymerase chain reaction-based MACH-2 protocol, we present a strategy that requires little DNA sequence information to selectively isolate the longest cDNA variant from plasmid libraries in about 3 days. Our protocols were used to isolate cDNAs for the Drosophila activin and follistatin genes.


Asunto(s)
Activinas/genética , ADN Complementario/aislamiento & purificación , Drosophila/genética , Técnicas de Amplificación de Ácido Nucleico/métodos , Secuencia de Aminoácidos , Animales , Clonación Molecular , ADN Complementario/química , ADN Complementario/genética , Folistatina , Datos de Secuencia Molecular , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido
8.
Neuron ; 33(4): 529-43, 2002 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-11856528

RESUMEN

Proper synaptic development is critical for establishing all aspects of neural function including learning, memory, and locomotion. Here, we describe the phenotypic consequences of mutations in the wishful thinking (wit) gene, the Drosophila homolog of the vertebrate BMP type II receptor. Mutations in wit result in pharate lethality that can be rescued by expression of a wit transgene in motor neurons but not in muscles. Mutant larvae exhibit small synapses, severe defects in evoked junctional potentials, a lower frequency of spontaneous vesicle release, and an alteration in the ultrastructure of synaptic active zones. These results reveal a novel role for BMP signaling in regulating Drosophila neuromuscular junction synapse assembly and activity and may indicate that similar pathways could govern vertebrate synapse development.


Asunto(s)
Sistema Nervioso Central/anomalías , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Mutación/fisiología , Unión Neuromuscular/anomalías , Proteínas Serina-Treonina Quinasas/genética , Receptores de Superficie Celular/genética , Factores de Transcripción , Animales , Tipificación del Cuerpo/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/genética , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/ultraestructura , ADN Complementario/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/aislamiento & purificación , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/ultraestructura , Femenino , Genes Letales/genética , Inmunohistoquímica , Masculino , Datos de Secuencia Molecular , Neuronas Motoras/metabolismo , Neuronas Motoras/ultraestructura , Unión Neuromuscular/crecimiento & desarrollo , Unión Neuromuscular/ultraestructura , Plasticidad Neuronal/genética , Neurotransmisores/genética , Neurotransmisores/metabolismo , Fenotipo , Proteínas Serina-Treonina Quinasas/aislamiento & purificación , Receptores de Superficie Celular/aislamiento & purificación , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA