Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr Biol ; 33(4): 727-736.e6, 2023 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-36669498

RESUMEN

Cilia are highly conserved organelles critical for animal development and perception. Dysfunction of cilia has been linked to a wide spectrum of human genetic diseases, termed ciliopathies.1,2 Transition fibers (TFs) are striking ciliary base structures essential for cilia assembly. Vertebrates' TFs that originate from centriole distal appendages (DAs) mediate basal body docking to ciliary vesicles to initiate ciliogenesis and regulate the entry of ciliary proteins for axoneme assembly via intraflagellar transport (IFT) machinery.3 Although no distal appendages can be observed on Drosophila centrioles,4,5 three key TF proteins, FBF1, CEP164, and CEP89, have obvious homologs in Drosophila. We aimed to compare their functions with their mammalian counterparts in Drosophila ciliogenesis. Here, we show that all three proteins are localized like TF proteins at the ciliary base in both sensory neurons and spermatocytes, the only two types of ciliated cells in flies. Fbf1 and Cep89 are essential for the formation of IFT-dependent neuronal cilia, but Cep164 is dispensable for ciliogenesis in flies. Strikingly, none are required for basal body docking and transition zone (TZ) assembly in IFT-dependent neuronal cilia or IFT-independent spermatocyte cilia. Furthermore, we demonstrate that Unc is essential to recruit all three TF proteins and establish a hierarchical order, with Cep89 acting on Fbf1. Collectively, our results not only demonstrate that TF proteins are required for IFT-dependent ciliogenesis in Drosophila, in agreement with an evolutionarily conserved function of these proteins in regulating ciliary protein entry, but also that the basal body docking function of TFs has diverged during evolution.


Asunto(s)
Cilios , Drosophila , Animales , Humanos , Cilios/metabolismo , Transporte Biológico/fisiología , Centriolos/metabolismo , Orgánulos/metabolismo , Mamíferos
2.
Elife ; 82019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31821146

RESUMEN

Cilia and flagella are conserved eukaryotic organelles essential for cellular signaling and motility. Cilia dysfunctions cause life-threatening ciliopathies, many of which are due to defects in the transition zone (TZ), a complex structure of the ciliary base. Therefore, understanding TZ assembly, which relies on ordered interactions of multiprotein modules, is of critical importance. Here, we show that Drosophila Dzip1 and Fam92 form a functional module which constrains the conserved core TZ protein, Cep290, to the ciliary base. We identify cell type specific roles of this functional module in two different tissues. While it is required for TZ assembly in all Drosophila ciliated cells, it also regulates basal-body growth and docking to the plasma membrane during spermatogenesis. We therefore demonstrate a novel regulatory role for Dzip1 and Fam92 in mediating membrane/basal-body interactions and show that these interactions exhibit cell type specific functions in basal-body maturation and TZ organization.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Cilios/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Alelos , Animales , Cuerpos Basales/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Transporte de Catión/genética , Membrana Celular/metabolismo , Cilios/genética , Cilios/ultraestructura , Drosophila/genética , Proteínas de Drosophila/genética , Flagelos/genética , Flagelos/metabolismo , Flagelos/ultraestructura , Células Germinativas , Masculino , Proteínas Nucleares/metabolismo , Células Receptoras Sensoriales , Espermatogénesis/fisiología
3.
Mol Biol Cell ; 30(5): 636-645, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30601696

RESUMEN

Producing mature spermatozoa is essential for sexual reproduction in metazoans. Spermiogenesis involves dramatic cell morphological changes going from sperm tail elongation and nuclear reshaping to cell membrane remodeling during sperm individualization and release. The sperm manchette plays a critical scaffolding function during nuclear remodeling by linking the nuclear lamina to the cytoskeleton. Here, we describe the role of an uncharacterized protein in Drosophila, salto/CG13164, involved in nuclear shaping and spermatid individualization. Salto has dynamic localization during spermatid differentiation, being progressively relocated from the sperm-nuclear dense body, which is equivalent to the mammalian sperm manchette, to the centriolar adjunct and acrosomal cap during spermiogenesis. salto-null male flies are sterile and exhibit complete spermatid individualization defects. salto-deficient spermatids show coiled spermatid nuclei at late maturation stages and stalled individualization complexes. Our work sheds light on a novel component involved in cytoskeleton-based cell-morphological changes during spermiogenesis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Morfogénesis , Cabeza del Espermatozoide/metabolismo , Animales , Caspasa 3/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Masculino , Mutación/genética , Especificidad de Órganos , Cabeza del Espermatozoide/ultraestructura , Espermatogénesis , Testículo/metabolismo
4.
J Cell Biol ; 214(7): 875-89, 2016 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-27646273

RESUMEN

The ciliary transition zone (TZ) is a complex structure found at the cilia base. Defects in TZ assembly are associated with human ciliopathies. In most eukaryotes, three protein complexes (CEP290, NPHP, and MKS) cooperate to build the TZ. We show that in Drosophila melanogaster, mild TZ defects are observed in the absence of MKS components. In contrast, Cby and Azi1 cooperate to build the TZ by acting upstream of Cep290 and MKS components. Without Cby and Azi1, centrioles fail to form the TZ, precluding sensory cilia assembly, and no ciliary membrane cap associated with sperm ciliogenesis is made. This ciliary cap is critical to recruit the tubulin-depolymerizing kinesin Klp59D, required for regulation of axonemal growth. Our results show that Drosophila TZ assembly in sensory neurons and male germ cells involves cooperative actions of Cby and Dila. They further reveal that temporal control of membrane cap assembly by TZ components and microtubule elongation by kinesin-13 is required for axoneme formation in male germ cells.


Asunto(s)
Axonema/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Espermatozoides/citología , Espermatozoides/metabolismo , Animales , Axonema/ultraestructura , Centriolos/metabolismo , Cilios/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/ultraestructura , Fertilidad , Masculino , Espermatogénesis , Espermatozoides/ultraestructura
5.
Methods Cell Biol ; 127: 279-302, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25837397

RESUMEN

Drosophila melanogaster is a powerful genetic model organism to understand the function of proteins in specific cellular processes. Cilia have been extensively studied in Drosophila playing various sensory functions that are essential for fly survival. Indeed, flies defective in cilia formation cannot walk, fly, or feed properly. Drosophila harbors different types of cilia that can be motile or immotile or that can show compartimentalized (intraflagellar transport (IFT)-dependent) or cytoplasmic (IFT-independent) mode of assembly. Therefore, Drosophila represents an advantageous model organism to study the function of novel ciliary candidates and to address specific questions such as their requirement for IFT-dependent processes versus other aspects of cilia-associated functions. This chapter describes protocols to visualize cilia by direct or indirect fluorescent labeling and protocols to analyze ciliary ultrastructure by electron microscopy.


Asunto(s)
Cilios/fisiología , Cilios/ultraestructura , Células Receptoras Sensoriales/fisiología , Animales , Axonema/metabolismo , Drosophila melanogaster/embriología , Técnica del Anticuerpo Fluorescente/métodos , Técnica del Anticuerpo Fluorescente Directa/métodos , Técnica del Anticuerpo Fluorescente Indirecta/métodos , Microscopía Electrónica de Transmisión/métodos , Coloración y Etiquetado/métodos
6.
Mol Biol Cell ; 25(8): 1276-86, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24554765

RESUMEN

Cilia play major functions in physiology and development, and ciliary dysfunctions are responsible for several diseases in humans called ciliopathies. Cilia motility is required for cell and fluid propulsion in organisms. In humans, cilia motility deficiencies lead to primary ciliary dyskinesia, with upper-airways recurrent infections, left-right asymmetry perturbations, and fertility defects. In Drosophila, we identified hemingway (hmw) as a novel component required for motile cilia function. hmw encodes a 604-amino acid protein characterized by a highly conserved coiled-coil domain also found in the human orthologue, KIAA1430. We show that HMW is conserved in species with motile cilia and that, in Drosophila, hmw is expressed in ciliated sensory neurons and spermatozoa. We created hmw-knockout flies and found that they are hearing impaired and male sterile. hmw is implicated in the motility of ciliated auditory sensory neurons and, in the testis, is required for elongation and maintenance of sperm flagella. Because HMW is absent from mature flagella, we propose that HMW is not a structural component of the motile axoneme but is required for proper acquisition of motile properties. This identifies HMW as a novel, evolutionarily conserved component necessary for motile cilium function and flagella assembly.


Asunto(s)
Cilios/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Cola del Espermatozoide/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Sitios de Unión , Trastornos de la Motilidad Ciliar , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Técnicas de Inactivación de Genes , Pérdida Auditiva/genética , Infertilidad Masculina , Masculino , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Factores de Transcripción del Factor Regulador X , Alineación de Secuencia , Espermatogénesis/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Hum Mol Genet ; 23(3): 563-77, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24067530

RESUMEN

Cilia are evolutionarily conserved organelles endowed with essential physiological and developmental functions. In humans, disruption of cilia motility or signaling leads to complex pleiotropic genetic disorders called ciliopathies. Cilia motility requires the assembly of multi-subunit motile components such as dynein arms, but mechanisms underlying their assembly pathway and transport into the axoneme are still largely unknown. We identified a previously uncharacterized coiled-coil domain containing protein CCDC151, which is evolutionarily conserved in motile ciliated species and shares ancient features with the outer dynein arm-docking complex 2 of Chlamydomonas. In Drosophila, we show that CG14127/CCDC151 is associated with motile intraflagellar transport (IFT)-dependent cilia and required for geotaxis behavior of adult flies. In zebrafish, Ccdc151 is expressed in tissues with motile cilia, and morpholino-induced depletion of Ccdc151 leads to left-right asymmetry defects and kidney cysts. We demonstrate that Ccdc151 is required for proper motile function of cilia in the Kupffer's vesicle and in the pronephros by controlling dynein arm assembly, showing that Ccdc151 is a novel player in the control of IFT-dependent dynein arm assembly in animals. However, we observed that CCDC151 is also implicated in other cellular functions in vertebrates. In zebrafish, ccdc151 is involved in proper orientation of cell divisions in the pronephros and genetically interacts with prickle1 in this process. Furthermore, knockdown experiments in mammalian cells demonstrate that CCDC151 is implicated in the regulation of primary cilium length. Hence, CCDC151 is required for motile cilia function in animals but has acquired additional non-motile functions in vertebrates.


Asunto(s)
Cilios/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Axonema/metabolismo , Transporte Biológico , Polaridad Celular , Cilios/genética , Secuencia Conservada , Drosophila/embriología , Drosophila/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Embrión no Mamífero/citología , Epéndimo/citología , Flagelos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Enfermedades Renales/genética , Enfermedades Renales/patología , Ratones , Filogenia , Estructura Terciaria de Proteína , Proteínas/química , Proteínas/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética
8.
J Cell Biol ; 197(2): 313-25, 2012 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-22508513

RESUMEN

Centriole-to-basal body conversion, a complex process essential for ciliogenesis, involves the progressive addition of specific proteins to centrioles. CHIBBY (CBY) is a coiled-coil domain protein first described as interacting with ß-catenin and involved in Wg-Int (WNT) signaling. We found that, in Drosophila melanogaster, CBY was exclusively expressed in cells that require functional basal bodies, i.e., sensory neurons and male germ cells. CBY was associated with the basal body transition zone (TZ) in these two cell types. Inactivation of cby led to defects in sensory transduction and in spermatogenesis. Loss of CBY resulted in altered ciliary trafficking into neuronal cilia, irregular deposition of proteins on spermatocyte basal bodies, and, consequently, distorted axonemal assembly. Importantly, cby(1/1) flies did not show Wingless signaling defects. Hence, CBY is essential for normal basal body structure and function in Drosophila, potentially through effects on the TZ. The function of CBY in WNT signaling in vertebrates has either been acquired during vertebrate evolution or lost in Drosophila.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Nucleares/metabolismo , Células Receptoras Sensoriales/metabolismo , Espermatozoides/metabolismo , Proteína Wnt1/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/química , Proteínas Portadoras/genética , Células Cultivadas , Centriolos/metabolismo , Cilios/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Infertilidad Masculina , Masculino , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/química , Proteínas Nucleares/genética , Transporte de Proteínas , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/metabolismo , Vía de Señalización Wnt
9.
PLoS One ; 7(2): e30482, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22347378

RESUMEN

Methods that use homologous recombination to engineer the genome of C. elegans commonly use strains carrying specific insertions of the heterologous transposon Mos1. A large collection of known Mos1 insertion alleles would therefore be of general interest to the C. elegans research community. We describe here the optimization of a semi-automated methodology for the construction of a substantial collection of Mos1 insertion mutant strains. At peak production, more than 5,000 strains were generated per month. These strains were then subject to molecular analysis, and more than 13,300 Mos1 insertions characterized. In addition to targeting directly more than 4,700 genes, these alleles represent the potential starting point for the engineered deletion of essentially all C. elegans genes and the modification of more than 40% of them. This collection of mutants, generated under the auspices of the European NEMAGENETAG consortium, is publicly available and represents an important research resource.


Asunto(s)
Caenorhabditis elegans/genética , Elementos Transponibles de ADN , Proteínas de Unión al ADN , Ingeniería Genética/métodos , Genoma/genética , Recombinación Genética , Transposasas , Animales , Animales Modificados Genéticamente , Recombinación Homóloga , Mutagénesis Insercional , Investigación
10.
Curr Biol ; 20(23): 2090-9, 2010 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-21093267

RESUMEN

BACKGROUND: A critical function of telomeres is to prevent fusion of chromosome ends by the DNA repair machinery. In Drosophila somatic cells, assembly of the protecting capping complex at telomeres notably involves the recruitment of HOAP, HP1, and their recently identified partner, HipHop. We previously showed that the hiphop gene was duplicated before the radiation of the melanogaster subgroup of species, giving birth to K81, a unique paternal effect gene specifically expressed in the male germline. RESULTS: Here we show that K81 specifically associates with telomeres during spermiogenesis, along with HOAP and HP1, and is retained on paternal chromosomes until zygote formation. In K81 mutant testes, capping proteins are not maintained at telomeres in differentiating spermatids, resulting in the transmission of uncapped paternal chromosomes that fail to properly divide during the first zygotic mitosis. Despite the apparent similar capping roles of K81 and HipHop in their respective domain of expression, we demonstrate by in vivo reciprocal complementation analyses that they are not interchangeable. Strikingly, HipHop appeared to be unable to maintain capping proteins at telomeres during the global chromatin remodeling of spermatid nuclei. CONCLUSIONS: Our data demonstrate that K81 is essential for the maintenance of capping proteins at telomeres in postmeiotic male germ cells. In species of the melanogaster subgroup, HipHop and K81 have not only acquired complementary expression domains, they have also functionally diverged following the gene duplication event. We propose that K81 specialized in the maintenance of telomere protection in the highly peculiar chromatin environment of differentiating male gametes.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Espermatozoides/fisiología , Telómero/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Drosophila/genética , Epigénesis Genética , Femenino , Masculino , Familia de Multigenes , Filogenia , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
11.
Genome Biol ; 8(9): R195, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17875208

RESUMEN

BACKGROUND: Regulatory factor X (RFX) transcription factors play a key role in ciliary assembly in nematode, Drosophila and mouse. Using the tremendous advantages of comparative genomics in closely related species, we identified novel genes regulated by dRFX in Drosophila. RESULTS: We first demonstrate that a subset of known ciliary genes in Caenorhabditis elegans and Drosophila are regulated by dRFX and have a conserved RFX binding site (X-box) in their promoters in two highly divergent Drosophila species. We then designed an X-box consensus sequence and carried out a genome wide computer screen to identify novel genes under RFX control. We found 412 genes that share a conserved X-box upstream of the ATG in both species, with 83 genes presenting a more restricted consensus. We analyzed 25 of these 83 genes, 16 of which are indeed RFX target genes. Two of them have never been described as involved in ciliogenesis. In addition, reporter construct expression analysis revealed that three of the identified genes encode proteins specifically localized in ciliated endings of Drosophila sensory neurons. CONCLUSION: Our X-box search strategy led to the identification of novel RFX target genes in Drosophila that are involved in sensory ciliogenesis. We also established a highly valuable Drosophila cilia and basal body dataset. These results demonstrate the accuracy of the X-box screen and will be useful for the identification of candidate genes for human ciliopathies, as several human homologs of RFX target genes are known to be involved in diseases, such as Bardet-Biedl syndrome.


Asunto(s)
Proteínas de Unión al ADN/genética , Drosophila/genética , Regulación de la Expresión Génica , Factores de Transcripción/genética , Secuencias de Aminoácidos , Animales , Sitios de Unión , Caenorhabditis elegans , Secuencia Conservada , Drosophila melanogaster/genética , Perfilación de la Expresión Génica , Genes Reporteros , Genómica/métodos , Humanos , Regiones Promotoras Genéticas , Factores de Transcripción del Factor Regulador X , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...