Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Development ; 150(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37681291

RESUMEN

Vertebrate podocytes and Drosophila nephrocytes display slit diaphragms, specialised cell junctions that are essential for the execution of the basic excretory function of ultrafiltration. To elucidate the mechanisms of slit diaphragm assembly we have studied their formation in Drosophila embryonic garland nephrocytes. These cells of mesenchymal origin lack overt apical-basal polarity. We find that their initial membrane symmetry is broken by an acytokinetic cell division that generates PIP2-enriched domains at their equator. The PIP2-enriched equatorial cortex becomes a favourable domain for hosting slit diaphragm proteins and the assembly of the first slit diaphragms. Indeed, when this division is either prevented or forced to complete cytokinesis, the formation of diaphragms is delayed to larval stages. Furthermore, although apical polarity determinants also accumulate at the equatorial cortex, they do not appear to participate in the recruitment of slit diaphragm proteins. The mechanisms we describe allow the acquisition of functional nephrocytes in embryos, which may confer on them a biological advantage similar to the formation of the first vertebrate kidney, the pronephros.


Asunto(s)
Citocinesis , Drosophila , Animales , División Celular , Corteza Cerebral , Diafragma
2.
Development ; 150(10)2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-37213082
3.
PLoS Genet ; 11(8): e1005463, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26305360

RESUMEN

During development, proper differentiation and final organ size rely on the control of territorial specification and cell proliferation. Although many regulators of these processes have been identified, how both are coordinated remains largely unknown. The homeodomain Iroquois/Irx proteins play a key, evolutionarily conserved, role in territorial specification. Here we show that in the imaginal discs, reduced function of Iroquois genes promotes cell proliferation by accelerating the G1 to S transition. Conversely, their increased expression causes cell-cycle arrest, down-regulating the activity of the Cyclin E/Cdk2 complex. We demonstrate that physical interaction of the Iroquois protein Caupolican with Cyclin E-containing protein complexes, through its IRO box and Cyclin-binding domains, underlies its activity in cell-cycle control. Thus, Drosophila Iroquois proteins are able to regulate cell-autonomously the growth of the territories they specify. Moreover, our results provide a molecular mechanism for a role of Iroquois/Irx genes as tumour suppressors.


Asunto(s)
Proteínas de Drosophila/fisiología , Proteínas de Homeodominio/fisiología , Animales , Ciclo Celular , Línea Celular , Proliferación Celular , Ojo Compuesto de los Artrópodos/citología , Ojo Compuesto de los Artrópodos/metabolismo , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Discos Imaginales/citología , Discos Imaginales/metabolismo , Unión Proteica , Mapas de Interacción de Proteínas
4.
Genome Res ; 22(4): 642-55, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22234889

RESUMEN

Developmental genes are regulated by complex, distantly located cis-regulatory modules (CRMs), often forming genomic regulatory blocks (GRBs) that are conserved among vertebrates and among insects. We have investigated GRBs associated with Iroquois homeobox genes in 39 metazoans. Despite 600 million years of independent evolution, Iroquois genes are linked to ankyrin-repeat-containing Sowah genes in nearly all studied bilaterians. We show that Iroquois-specific CRMs populate the Sowah locus, suggesting that regulatory constraints underlie the maintenance of the Iroquois-Sowah syntenic block. Surprisingly, tetrapod Sowah orthologs are intronless and not associated with Iroquois; however, teleost and elephant shark data demonstrate that this is a derived feature, and that many Iroquois-CRMs were ancestrally located within Sowah introns. Retroposition, gene, and genome duplication have allowed selective elimination of Sowah exons from the Iroquois regulatory landscape while keeping associated CRMs, resulting in large associated gene deserts. These results highlight the importance of CRMs in imposing constraints to genome architecture, even across large phylogenetic distances, and of gene duplication-mediated genetic redundancy to disentangle these constraints, increasing genomic plasticity.


Asunto(s)
Genoma/genética , Proteínas de Homeodominio/genética , Invertebrados/genética , Vertebrados/genética , Secuencia de Aminoácidos , Animales , Evolución Molecular , Duplicación de Gen/genética , Regulación del Desarrollo de la Expresión Génica , Genómica/métodos , Proteínas de Homeodominio/clasificación , Insectos/clasificación , Insectos/embriología , Insectos/genética , Invertebrados/clasificación , Invertebrados/embriología , Datos de Secuencia Molecular , Filogenia , Retroelementos/genética , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Vertebrados/clasificación , Vertebrados/embriología
5.
Development ; 139(6): 1110-4, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22318229

RESUMEN

Dorsoventral (DV) axis formation in Drosophila begins with selective activation of EGFR, a receptor tyrosine kinase (RTK), in dorsal-anterior (DA) ovarian follicle cells. A critical event regulated by EGFR signaling is the repression of the sulfotransferase-encoding gene pipe in dorsal follicle cells, but how this occurs remains unclear. Here we show that Mirror (Mirr), a homeodomain transcription factor induced by EGFR signaling in DA follicle cells, directly represses pipe expression by binding to a conserved element in the pipe regulatory region. In addition, we find that the HMG-box protein Capicua (Cic) supports pipe expression in ventral follicle cells by repressing Mirr in this region. Interestingly, this role of Cic resembles its function in regulating anteroposterior (AP) body patterning, where Cic supports gap gene expression in central regions of the embryo by repressing Tailless, a repressor induced by RTK signaling at the embryonic poles. Thus, related RTK-Cic repressor circuits regulate the early stages of Drosophila DV and AP body axis formation.


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Receptores ErbB/metabolismo , Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/metabolismo , Proteínas de Homeodominio/metabolismo , Receptores de Péptidos de Invertebrados/metabolismo , Proteínas Represoras/metabolismo , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia Conservada , Proteínas de Drosophila/biosíntesis , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Embrión no Mamífero/metabolismo , Femenino , Folículo Ovárico/citología , Folículo Ovárico/embriología , Folículo Ovárico/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Transducción de Señal , Sulfotransferasas/biosíntesis
6.
PLoS Genet ; 7(7): e1002186, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21811416

RESUMEN

A central issue of myogenesis is the acquisition of identity by individual muscles. In Drosophila, at the time muscle progenitors are singled out, they already express unique combinations of muscle identity genes. This muscle code results from the integration of positional and temporal signalling inputs. Here we identify, by means of loss-of-function and ectopic expression approaches, the Iroquois Complex homeobox genes araucan and caupolican as novel muscle identity genes that confer lateral transverse muscle identity. The acquisition of this fate requires that Araucan/Caupolican repress other muscle identity genes such as slouch and vestigial. In addition, we show that Caupolican-dependent slouch expression depends on the activation state of the Ras/Mitogen Activated Protein Kinase cascade. This provides a comprehensive insight into the way Iroquois genes integrate in muscle progenitors, signalling inputs that modulate gene expression and protein activity.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas de Homeodominio/genética , Músculos/metabolismo , Factores de Transcripción/genética , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Línea Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Hibridación in Situ , Microscopía Confocal , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Desarrollo de Músculos/genética , Músculos/embriología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Homología de Secuencia de Ácido Nucleico , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Proteínas ras/metabolismo
7.
J Cell Sci ; 124(Pt 2): 240-51, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21172808

RESUMEN

Many epithelial tissues undergo extensive remodelling during morphogenesis. How their epithelial features, such as apicobasal polarity or adhesion, are maintained and remodelled and how adhesion and polarity proteins contribute to morphogenesis are two important questions in development. Here, we approach these issues by investigating the role of the apical determinant protein Crumbs (Crb) during the morphogenesis of the embryonic Drosophila tracheal system. Crb accumulates differentially throughout tracheal development and is required for different tracheal events. The earliest requirement for Crb is for tracheal invagination, which is preceded by an enhanced accumulation of Crb in the invagination domain. There, Crb, acting in parallel with the epidermal growth factor receptor (Egfr) pathway, is required for tracheal cell apical constriction and for organising an actomyosin complex, which we propose is mediated by Crb recruitment of moesin (Moe). The ability of a Crb isoform unable to rescue polarity in crb mutants to otherwise rescue their invagination phenotype, and the converse inability of a FERM-binding domain mutant Crb to rescue faulty invagination, support our hypothesis that it is the absence of Crb-dependent Moe enrichment, and not the polarity defect, that mainly underlies the crb invagination phenotype. This hypothesis is supported by the phenotype of lethal giant larvae (lgl); crb double mutants. These results unveil a link between Crb and the organisation of the actin cytoskeleton during morphogenesis.


Asunto(s)
Polaridad Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Proteínas de la Membrana/metabolismo , Morfogénesis , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Proteínas de la Membrana/genética , Tráquea/citología , Tráquea/embriología , Tráquea/metabolismo
8.
J Cell Biol ; 166(4): 549-57, 2004 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-15302858

RESUMEN

Both in Drosophila and vertebrate epithelial cells, the establishment of apicobasal polarity requires the apically localized, membrane-associated Par-3-Par-6-aPKC protein complex. In Drosophila, this complex colocalizes with the Crumbs-Stardust (Sdt)-Pals1-associated TJ protein (Patj) complex. Genetic and molecular analyses suggest a functional relationship between them. We show, by overexpression of a kinase-dead Drosophila atypical PKC (DaPKC), the requirement for the kinase activity of DaPKC to maintain the position of apical determinants and to restrict the localization of basolateral ones. We demonstrate a novel physical interaction between the apical complexes, via direct binding of DaPKC to both Crb and Patj, and identify Crumbs as a phosphorylation target of DaPKC. This phosphorylation of Crumbs is functionally significant. Thus, a nonphosphorylatable Crumbs protein behaves in vivo as a dominant negative. Moreover, the phenotypic effect of overexpressing wild-type Crumbs is suppressed by reducing DaPKC activity. These results provide a mechanistic framework for the functional interaction between the Par-3-Par-6-aPKC and Crumbs-Sdt-Patj complexes based in the posttranslational modification of Crb by DaPKC.


Asunto(s)
Proteínas de Drosophila/metabolismo , Células Epiteliales/citología , Proteínas de la Membrana/metabolismo , Proteína Quinasa C/metabolismo , Secuencia de Aminoácidos , Animales , Tipificación del Cuerpo , Polaridad Celular , ADN Complementario/metabolismo , Drosophila , Regulación del Desarrollo de la Expresión Génica , Biblioteca de Genes , Genes Dominantes , Glutatión Transferasa/metabolismo , Inmunohistoquímica , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión/metabolismo , Uniones Estrechas , Alas de Animales/metabolismo
9.
Fly (Austin) ; 9(3): 126-31, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26760760

RESUMEN

Drosophila Iroquois (Iro) proteins are components of the TALE homeodomain family of transcriptional regulators. They play key roles in territorial specification and pattern formation. A recent study has disclosed a novel developmental function of the Iro proteins. In the eye and wing imaginal discs, they can regulate the size of the territories that they specify. They do so by cell-autonomously controlling cell cycle progression. Indeed, Iro proteins down-regulate the activity of the CyclinE/Cdk2 complex by a transcription-independent mechanism. This novel function is executed mainly through 2 evolutionarily conserved domains of the Iro proteins: the Cyclin Binding Domain and the IRO-box, which mediate their binding to CyclinE-containing protein complexes. Here we discuss the functional implications of the control of the cell cycle by Iro proteins for development and oncogenesis.


Asunto(s)
Ciclo Celular/genética , Proteínas de Drosophila/fisiología , Drosophila/citología , Proteínas de Homeodominio/fisiología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Secuencia Conservada , Ciclina E/genética , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Regulación hacia Abajo , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Dominios Proteicos , Alineación de Secuencia
10.
Fly (Austin) ; 6(4): 234-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22878648

RESUMEN

Dorsoventral (DV) axis formation in Drosophila begins during oogenesis through the graded activation of the EGF receptor (EGFR)-Ras-MAPK signaling pathway in the follicle cell layer of the egg chamber. EGFR signaling, which is higher in dorsal follicle cells, represses expression of the sulfotransferase-encoding gene pipe, thereby delimiting a ventral domain of Pipe activity that is critical for the subsequent induction of ventral embryonic fates. We have characterized the transcriptional circuit that links EGFR signaling to pipe repression: in dorsal follicle cells, the homeodomain transcription factor Mirror (Mirr), which is induced by EGFR signaling, directly represses pipe transcription, whereas in ventral follicle cells, the HMG-box protein Capicua (Cic) supports pipe expression by repressing mirr. Although Cic is under negative post-transcriptional regulation by Ras-MAPK signaling in different contexts, the relevance of this mechanism for the interpretation of the EGFR signal during DV pattern formation remains unclear. Here, we consider a model where EGFR-mediated downregulation of Cic modulates the spatial distribution of Mirr protein in lateral follicle cells, thereby contributing to define the position at which the pipe expression border is formed.


Asunto(s)
Tipificación del Cuerpo/genética , Regulación hacia Abajo , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila/citología , Receptores ErbB/fisiología , Proteínas HMGB/genética , Receptores de Péptidos de Invertebrados/fisiología , Proteínas Represoras/genética , Animales , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/metabolismo , Proteínas HMGB/fisiología , Modelos Biológicos , Proteínas Represoras/metabolismo , Proteínas Represoras/fisiología , Transducción de Señal , Sulfotransferasas/genética , Sulfotransferasas/metabolismo
11.
Development ; 134(7): 1337-46, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17329358

RESUMEN

In Drosophila, restricted expression of the Iroquois complex (Iro-C) genes in the proximal region of the wing imaginal disc contributes to its territorial subdivision, specifying first the development of the notum versus the wing hinge, and subsequently, that of the lateral versus medial notum. Iro-C expression is under the control of the EGFR and Dpp signalling pathways. To analyze how both pathways cooperate in the regulation of Iro-C, we isolated several wing disc-specific cis-regulatory elements of the complex. One of these (IroRE(2)) integrates competing inputs of the EGFR and Dpp pathways, mediated by the transcription factors Pointed (downstream of EGFR pathway) and Pannier/U-shaped and Mothers against Dpp (Mad), in the case of Dpp. By contrast, a second element (IroRE(1)) mediates activation by both the EGFR and Dpp pathways, thus promoting expression of Iro-C in a region of elevated levels of Dpp signalling, the prospective lateral notum near the anterior-posterior compartment boundary. These results help define the molecular mechanisms of the interplay between the EGFR and Dpp pathways in the specification and patterning of the notum.


Asunto(s)
Tipificación del Cuerpo/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/embriología , Receptores ErbB/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteínas Quinasas/metabolismo , Receptores de Péptidos de Invertebrados/metabolismo , Transducción de Señal/fisiología , Tórax/embriología , Animales , Cartilla de ADN , Proteínas de Unión al ADN/metabolismo , Drosophila/metabolismo , Inmunohistoquímica , Mutagénesis , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Análisis de Secuencia de ADN , Factores de Transcripción/metabolismo
12.
Mol Cell ; 23(5): 631-40, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16949360

RESUMEN

The PB1-domain-containing proteins p62, aPKC, MEKK2/MEKK3, MEK5, and Par-6 play roles in critical cell processes like osteoclastogenesis, angiogenesis, and early cardiovascular development or cell polarity. PB1 domains are scaffold modules that adopt the topology of ubiquitin-like beta-grasp folds that interact with each other in a front-to-back mode to arrange heterodimers or homo-oligomers. The different PB1 domain adaptors provide specificity for PB1 kinases to ensure the effective transmission of cellular signals. Also, recent data suggest that PB1 domains may serve to orchestrate signaling cascades not involving other PB1 domains, such as the MEK5-ERK5 and p62-ERK1 interactions.


Asunto(s)
Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Transducción de Señal , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al ADN , Humanos , Inflamación , Unión Proteica , Estructura Terciaria de Proteína , Proteínas/metabolismo , Proteína Sequestosoma-1
13.
EMBO J ; 21(13): 3327-36, 2002 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12093734

RESUMEN

Grim encodes a protein required for programmed cell death in Drosophila. The Grim N-terminus induces apoptosis by disrupting IAP blockage of caspases; however, N-terminally-deleted Grim retains pro apoptotic activity. We describe GH3, a 15 amino acid internal Grim domain absolutely required for its proapoptotic activity and sufficient to induce cell death when fused to heterologous carrier proteins. A GH3 homology region is present in the Drosophila proapoptotic proteins Reaper and Sickle. The GH3 domain and the homologous regions in Reaper and Sickle are predicted to be structured as amphipathic alpha-helixes. During apoptosis induction, Grim colocalizes with mitochondria and cytochrome c in a GH3-dependent but N-terminal- and caspase activity-independent manner. When Grim is overexpressed in vivo, both the N-terminal and the GH3 domains are equally necessary, and cooperate for apoptosis induction. The N-terminal and GH3 Grim domains thus activate independent apoptotic pathways that synergize to induce programmed cell death efficiently.


Asunto(s)
Apoptosis/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/citología , Mitocondrias/fisiología , Neuropéptidos/fisiología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Animales Modificados Genéticamente , Caspasas/fisiología , Células Cultivadas , Grupo Citocromo c/fisiología , Proteínas de Drosophila/química , Femenino , Masculino , Datos de Secuencia Molecular , Neuropéptidos/química , Fenotipo , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/fisiología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
14.
Eur J Biochem ; 270(2): 293-306, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12605680

RESUMEN

Four distinct eukaryotic initiation factor 2alpha (eIF2alpha) kinases phosphorylate eIF2alpha at S51 and regulate protein synthesis in response to various environmental stresses. These are the hemin-regulated inhibitor (HRI), the interferon-inducible dsRNA-dependent kinase (PKR), the endoplasmic reticulum (ER)-resident kinase (PERK) and the GCN2 protein kinase. Whereas HRI and PKR appear to be restricted to mammalian cells, GCN2 and PERK seem to be widely distributed in eukaryotes. In this study, we have characterized the second eIF2alpha kinase found in Drosophila, a PERK homologue (DPERK). Expression of DPERK is developmentally regulated. During embryogenesis, DPERK expression becomes concentrated in the endodermal cells of the gut and in the germ line precursor cells. Recombinant wild-type DPERK, but not the inactive DPERK-K671R mutant, exhibited an autokinase activity, specifically phosphorylated Drosophila eIF2alpha at S50, and functionally replaced the endogenous Saccharomyces cerevisiae GCN2. The full length protein, when expressed in 293T cells, located in the ER-enriched fraction, and its subcellular localization changed with deletion of different N-terminal fragments. Kinase activity assays with these DPERK deletion mutants suggested that DPERK localization facilitates its in vivo function. Similar to mammalian PERK, DPERK forms oligomers in vivo and DPERK activity appears to be regulated by ER stress. Furthermore, the stable complexes between wild-type DPERK and DPERK-K671R mutant were mediated through the N terminus of the proteins and exhibited an in vitro eIF2alpha kinase activity.


Asunto(s)
Drosophila melanogaster/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , eIF-2 Quinasa/metabolismo , Secuencia de Aminoácidos , Animales , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Retículo Endoplásmico/metabolismo , Perfilación de la Expresión Génica , Datos de Secuencia Molecular , Fosforilación , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Estructura Terciaria de Proteína , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae , Análisis de Secuencia de ADN , Análisis de Secuencia de Proteína , eIF-2 Quinasa/genética
15.
Genetica ; 122(3): 311-24, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15609554

RESUMEN

The 3A and 60E regions of Drosophila melanogaster polytene chromosomes containing inserted copies of the P[1ArB] transposon have been subjected to an electron microscopic (EM) analysis. We show that both inserts led to formation of new bands within the interband regions 3A4/A6 and 60E8-9/E10. This allowed us to clone DNA of these interbands. Their sequences, as well as those of DNA from other four interbands described earlier, have been analyzed. We have found that, with the exception of 60E8-9/E10 interband, all other five regions under study corresponded to 5' or 3' ends of genes. We have further obtained the evidence for 60E8-9/E10 interband to harbor the 'housekeeping' RpL19 gene, which is transcribed in many tissues, including salivary glands. Based upon the genetic heterogeneity of the interbands observed a revised model of polytene chromosome organization is discussed.


Asunto(s)
Cromosomas/genética , Drosophila melanogaster/genética , Animales , Bandeo Cromosómico , Cromosomas/fisiología , Cromosomas/ultraestructura , Clonación Molecular , Elementos Transponibles de ADN , Microscopía Electrónica , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA