Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr Top Dev Biol ; 121: 287-309, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28057303

RESUMEN

Cancer is a complex disease that affects multiple organs. Whole-body animal models provide important insights into oncology that can lead to clinical impact. Here, we review novel concepts that Drosophila studies have established for cancer biology, drug discovery, and patient therapy. Genetic studies using Drosophila have explored the roles of oncogenes and tumor-suppressor genes that when dysregulated promote cancer formation, making Drosophila a useful model to study multiple aspects of transformation. Not limited to mechanism analyses, Drosophila has recently been showing its value in facilitating drug development. Flies offer rapid, efficient platforms by which novel classes of drugs can be identified as candidate anticancer leads. Further, we discuss the use of Drosophila as a platform to develop therapies for individual patients by modeling the tumor's genetic complexity. Drosophila provides both a classical and a novel tool to identify new therapeutics, complementing other more traditional cancer tools.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila/fisiología , Neoplasias/patología , Animales , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Humanos , Microambiente Tumoral
2.
Oncogene ; 33(21): 2801-6, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-23831567

RESUMEN

Current models of tumor cell invasion propose that oncogenic signaling converges upon key orchestrators of cytoskeletal dynamics, including c-Jun N-terminal kinase (Jnk) and RhoGTPase family members; these signals dynamically direct Actin remodeling proteins (ARPs) to catalyze the cytoskeletal changes required for migration. Src is a key driver of tumor aggression, metastasis and patient mortality. To clarify how Src regulates Actin dynamics to promote invasive migration, we performed a genetic modifier screen in a Drosophila model of invasion. Nine genes linked to Actin dynamics were identified that mediate invasion in situ. We found that ARPs were required for many oncogenic effects of Src including Mmp1 expression and initiation of apoptosis. Surprisingly, they were also regulators of Jnk pathway activity: both Src and the small GTPase Rho1 activated Jnk in a manner dependent on ARPs during invasion. Our results suggest that ARPs are not simply downstream executors of signal transduction pathways. Rather, they participate in a positive feedback network involving canonical oncogenic signaling pathways that promote tumor invasion.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Familia-src Quinasas/metabolismo , Animales , Apoptosis , Movimiento Celular , Drosophila , Activación Enzimática , Retroalimentación Fisiológica , Invasividad Neoplásica , Transducción de Señal
3.
Oncogenesis ; 2: e69, 2013 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-24018644

RESUMEN

Centrosomal kinase Nek2 is overexpressed in different cancers, yet how it contributes toward tumorigenesis remains poorly understood. dNek2 overexpression in a Drosophila melanogaster model led to upregulation of Drosophila Wnt ortholog wingless (Wg), and alteration of cell migration markers-Rho1, Rac1 and E-cadherin (Ecad)-resulting in changes in cell shape and tissue morphogenesis. dNek2 overexpression cooperated with receptor tyrosine kinase and mitogen-activated protein kinase signaling to upregulate activated Akt, Diap1, Mmp1 and Wg protein to promote local invasion, distant seeding and metastasis. In tumor cell injection assays, dNek2 cooperated with Ras and Src signaling to promote aggressive colonization of tumors into different adult fly tissues. Inhibition of the PI3K pathway suppressed the cooperation of dNek2 with other growth pathways. Consistent with our fly studies, overexpression of human Nek2 in A549 lung adenocarcinoma and HEK293T cells led to activation of the Akt pathway and increase in ß-catenin protein levels. Our computational approach identified a class of Nek2-inhibitory compounds and a novel drug-like pharmacophore that reversed the Nek2 overexpression phenotypes in flies and human cells. Our finding posits a novel role for Nek2 in promoting metastasis in addition to its currently defined role in promoting chromosomal instability. It provides a rationale for the selective advantage of centrosome amplification in cancer.

4.
Oncogene ; 32(26): 3184-97, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-22890320

RESUMEN

Chromatin remodeling proteins regulate multiple aspects of cell homeostasis, making them ideal candidates for misregulation in transformed cells. Here, we explore Sin3A, a member of the Sin3 family of proteins linked to tumorigenesis that are thought to regulate gene expression through their role as histone deacetylases (HDACs). We identified Drosophila Sin3a as an important mediator of oncogenic Ret receptor in a fly model of Multiple Endocrine Neoplasia Type 2. Reducing Drosophila Sin3a activity led to metastasis-like behavior and, in the presence of Diap1, secondary tumors distant from the site of origin. Genetic and Chip-Seq analyses identified previously undescribed Sin3a targets including genes involved in cell motility and actin dynamics, as well as signaling pathways including Src, Jnk and Rho. A key Sin3a oncogenic target, PP1B, regulates stability of ß-Catenin/Armadillo: the outcome is to oppose T-cell factor (TCF) function and Wg/Wnt pathway signaling in both fly and mammalian cancer cells. Reducing Sin3A strongly increased the invasive behavior of A549 human lung adenocarcinoma cells. We show that Sin3A is downregulated in a variety of human tumors and that Src, JNK, RhoA and PP1B/ß-Catenin are regulated in a manner analogous to our Drosophila models. Our data suggest that Sin3A influences a specific step of tumorigenesis by regulating a module of genes involved in cell invasion. Tumor progression may commonly rely on such 'modules of invasion' under the control of broad transcriptional regulators.


Asunto(s)
Neoplasias Pulmonares/patología , Neoplasia Endocrina Múltiple Tipo 2a/patología , Proteínas Represoras/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Transformación Celular Neoplásica , Drosophila , Proteínas de Drosophila/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasia Endocrina Múltiple Tipo 2a/metabolismo , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-ret/metabolismo , Proteínas Represoras/genética , Transducción de Señal , Complejo Correpresor Histona Desacetilasa y Sin3 , Activación Transcripcional , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Familia-src Quinasas/metabolismo
5.
Nat Cell Biol ; 3(8): E175-8, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11483971

RESUMEN

For the cell biologist, identifying changes in gene expression using DNA microarrays is just the start of a long journey from tissue to cell. We discuss how chip users can first filter noise (false-positives) from daunting microarray datasets. Combining laser capture microdissection with real-time polymerase chain reaction and reverse transcription is a helpful follow-up step that allows expression of selected genes to be quantified using sensitive new in situ hybridization and immunohistochemical methods based on tyramide signal amplification.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Animales , Artefactos , Fenómenos Fisiológicos Celulares , Humanos , Hibridación in Situ , Rayos Láser , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Nature ; 409(6820): 626-30, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11214322

RESUMEN

The mechanisms that establish and sharpen pattern across epithelia are poorly understood. In the developing nervous system, the first pattern elements appear as 'proneural clusters' In the morphogenetic furrow of the immature Drosophila retina proneural clusters emerge in a wave as a patterned array of 6-10-cell groups, which are recognizable by expression of Atonal, a basic helix-loop-helix transcription factor that is required to establish and pattern the first cell fate. The establishment and subsequent patterning of Atonal expression requires activity of the signalling transmembrane receptor Notch. Here we present in vivo and biochemical evidence that the secreted protein Scabrous associates with Notch, and can stabilize Notch protein at the surface. The result is a regulation of Notch activity that sharpens proneural cluster boundaries and ensures establishment of single pioneer neurons.


Asunto(s)
Proteínas de Drosophila , Glicoproteínas/metabolismo , Proteínas de Insectos/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Western Blotting , Tipificación del Cuerpo , Células Cultivadas , Drosophila , Respuesta al Choque Térmico , Sistema Nervioso/citología , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Receptores Notch , Retina/metabolismo
7.
Genetics ; 156(1): 205-17, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10978286

RESUMEN

Programmed cell death (PCD) in the Drosophila retina requires activity of the irregular chiasmC-roughest (irreC-rst) gene. Loss-of-function mutations in irreC-rst block PCD during retinal development and lead to a rough eye phenotype in the adult. To identify genes that interact with irreC-rst and may be involved in PCD, we conducted a genetic screen for dominant enhancers and suppressors of the adult rough eye phenotype. We screened 150,000 mutagenized flies and recovered 170 dominant modifiers that localized primarily to the second and third chromosomes. At least two allelic groups correspond to previously identified death regulators, Delta and dRas1. Examination of retinae from homozygous viable mutants indicated two major phenotypic classes. One class exhibited pleiotropic defects while the other class exhibited defects specific to the cell population that normally undergoes PCD.


Asunto(s)
Apoptosis/genética , Proteínas de Drosophila , Drosophila/citología , Drosophila/genética , Genes de Insecto , Retina/citología , Alelos , Animales , Moléculas de Adhesión Celular Neuronal/genética , Mapeo Cromosómico , Cruzamientos Genéticos , Drosophila/crecimiento & desarrollo , Elementos de Facilitación Genéticos , Proteínas del Ojo/genética , Femenino , Prueba de Complementación Genética , Proteínas de Insectos/genética , Masculino , Mutación , Fenotipo , Retina/crecimiento & desarrollo , Supresión Genética
8.
Curr Biol ; 10(9): 547-50, 2000 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-10801447

RESUMEN

As with all metazoans, the fly makes extensive use of selective programmed cell death (PCD) to remove excess cells and properly sculpt developing tissues. Several core components of the cell death machinery have been identified in flies, including caspases and an Apaf-1 ortholog [1] [2] [3] [4]. One missing component has been a member of the Bcl-2 family of proteins, which act either pro- or anti-apoptotically as upstream regulatory proteins. Here, we report the identification of Bcl-2 family members in Drosophila - dBorg-1 (Drosophila Bcl-2 ortholog), also identified by Igaki et al. [5], and dBorg-2. Removal of dBorg-1 function during Drosophila embryonic development resulted in excess glial cells, demonstrating its pro-apoptotic function. In cell culture assays, dBorg-1 efficiently induced apoptosis but, remarkably, also demonstrated protective activity when death stimuli were introduced. Finally, ectopic expression of dBorg-1 in the eye led to subtle defects that were strongly potentiated by ultra violet (UV) irradiation, resulting in a dramatic loss of retinal cells.


Asunto(s)
Apoptosis , Proteínas de Drosophila , Proteínas de Insectos/fisiología , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Animales , Apoptosis/efectos de la radiación , Secuencia de Bases , Células CHO , Línea Celular , Supervivencia Celular , Cricetinae , ADN Complementario , Drosophila , Ojo/citología , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas de Insectos/genética , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Neuroglía , Proteínas Proto-Oncogénicas c-bcl-2/genética , Rayos Ultravioleta
9.
Gene ; 243(1-2): 1-9, 2000 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-10675607

RESUMEN

The fine modulation of tyrosine phosphorylation by protein tyrosine phosphatases and protein tyrosine kinases is a key regulatory mechanism for many cell signaling pathways active during development. In a screen for genes with interesting expression patterns in the developing Drosophila pupal retina, we identified a novel pair of protein tyrosine phosphatases that exhibit an expression pattern suggesting a role in multiple steps of Drosophila neurogenesis. Together, these phosphatases define the primo locus. Their sequence is approx. 50% identical to each other and to low-molecular-weight protein tyrosine phosphatases (LMW-PTPs) identified in other species. Little is understood of the biological role of LMW-PTPs, and the powerful tools available in Drosophila should provide important insight into their role in signaling and development.


Asunto(s)
Drosophila/genética , Genes de Insecto/genética , Proteínas Tirosina Fosfatasas/genética , Secuencia de Aminoácidos , Animales , Northern Blotting , Mapeo Cromosómico , Drosophila/embriología , Drosophila/enzimología , Embrión no Mamífero/enzimología , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Hibridación in Situ , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Datos de Secuencia Molecular , Peso Molecular , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Distribución Tisular
10.
Mol Cell Neurosci ; 15(1): 88-98, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10662508

RESUMEN

Mutations in two genes, presenilin 1 (PS1) and presenilin 2, are linked to early onset cases of familial Alzheimer's disease. The presenilins are thought to contribute to the pathogenesis of Alzheimer's disease by directly or indirectly affecting the proteolytic processing of the amyloid precursor protein. They have also been implicated in the proteolytic processing of Notch. In PS1-deficient mammalian cells, the proteolytic release of the Notch intracellular domain is reduced. Likewise, loss-of-function mutations in Drosophila presenilin (Psn) prevent the production of the intracellular Notch signaling fragment and lead to phenotypes resembling Notch mutants. Here we characterize the Drosophila Psn protein and demonstrate that it undergoes a proteolytic cleavage. We describe Psn expression at different developmental stages of the fly and show Psn localization near both apical and basal plasma membranes. Furthermore, we demonstrate that portions of the Psn protein span the plasma membrane in S2 cells.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Procesamiento Proteico-Postraduccional , Empalme Alternativo , Enfermedad de Alzheimer/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Caenorhabditis elegans , Línea Celular , Membrana Celular/metabolismo , Secuencia Conservada , Proteínas de Drosophila , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Humanos , Proteínas de la Membrana/química , Ratones , Datos de Secuencia Molecular , Presenilina-1 , Receptores Notch , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Transactivadores/metabolismo
11.
Cell Death Differ ; 7(11): 1063-70, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11139279

RESUMEN

Selective cell death provides developing tissues with the means to precisely sculpt emerging structures. By imposing patterned cell death across a tissue, boundaries can be created and tightened. As such, programmed cell death is becoming recognized as a major mechanism for patterning of a variety of complex structures. Typically, cell types are initially organized into a fairly loose pattern; selective death then removes cells between pattern elements to create correct structures. In this review, we examine the role of selective cell death across the course of Drosophila development, including the tightening of embryonic segmental boundaries, head maturation, refining adult structures such as the eye and the wing, and the ability of cell death to correct for pattern defects introduced by gene mutation. We also review what is currently known of the relationship between signals at the cell surface that are responsible for tissue patterning and the basal cell death machinery, an issue that remains poorly understood.


Asunto(s)
Apoptosis , Tipificación del Cuerpo , Drosophila melanogaster/crecimiento & desarrollo , Metamorfosis Biológica , Animales , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Ecdisona/metabolismo , Retina/anatomía & histología , Retina/crecimiento & desarrollo
13.
Development ; 125(23): 4777-90, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9806926

RESUMEN

Developing epithelia use a variety of patterning mechanisms to place individual cells into their correct positions. However, the means by which pattern elements are established are poorly understood. Here, we report evidence that regulation of Drosophila EGF receptor (DER) activity plays a central role in propagating the evenly spaced array of ommatidia across the developing Drosophila retina. DER activity is essential for establishing the first ommatidial cell fate, the R8 photoreceptor neuron. R8s in turn appear to signal through Rhomboid and Vein to create a patterned array of 'proneural clusters' which contain high levels of phosphorylated ERKA and the bHLH protein Atonal. Finally, secretion by the proneural clusters of Argos represses DER activity in less mature regions to create a new pattern of R8s. Propagation of this process anteriorly results in a retina with a precise array of maturing ommatidia.


Asunto(s)
Tipificación del Cuerpo/fisiología , Proteínas de Drosophila , Drosophila melanogaster/embriología , Receptores ErbB/genética , Regulación del Desarrollo de la Expresión Génica , Neurregulinas , Epitelio Pigmentado Ocular/embriología , Retina/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas de Unión al ADN/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/efectos de la radiación , Embrión no Mamífero/fisiología , Receptores ErbB/fisiología , Rayos gamma , Genes ras , Secuencias Hélice-Asa-Hélice , Proteínas de Insectos/metabolismo , Proteínas de la Membrana/metabolismo , Mitosis , Mutagénesis , Fosforilación , Células Fotorreceptoras/citología , Células Fotorreceptoras/embriología , Epitelio Pigmentado Ocular/citología , Recombinación Genética , Retina/citología , Transducción de Señal , Factores de Transcripción/metabolismo
14.
Development ; 125(12): 2327-35, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9584131

RESUMEN

Local cell signaling can pattern the nervous system by directing cell fates, including programmed cell death. In the developing Drosophila retina, programmed cell death is used to remove excess cells between ommatidia. Cell ablation revealed the source and position of signals required for regulating the pattern of programmed cell death among these interommatidial cells. Two types of signals regulate this patterning event. Notch-mediated signals between interommatidial precursors result in removal of unneeded cells. Cone cells and primary pigment cells oppose this signal by supplying a 'life'-promoting activity; evidence is provided that this signal occurs through localized activation of the EGF Receptor/Ras pathway. Together, these signals refine the highly regular pattern observed in the adult retina.


Asunto(s)
Apoptosis , Tipificación del Cuerpo , Proteínas de Drosophila , Células Fotorreceptoras de Invertebrados/citología , Células Fotorreceptoras de Invertebrados/crecimiento & desarrollo , Proteínas Quinasas , Animales , Drosophila , Receptores ErbB/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Pupa , Receptores de Péptidos de Invertebrados/metabolismo , Receptores Notch , Retina/citología , Retina/crecimiento & desarrollo , Transducción de Señal , Proteínas ras/fisiología
15.
Development ; 122(12): 4139-47, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9012533

RESUMEN

In the developing Drosophila retina, the proneural gene for photoreceptor neurons is atonal, a basic helix-loop-helix transcription factor. Using atonal as a marker for proneural maturation, we examine the stepwise resolution of proneural clusters during the initiation of ommatidial differentiation in the developing eye disc. In addition, evidence is provided that atonal is negatively regulated by rough, a homeobox-containing transcription factor expressed exclusively in the retina. This interaction leads to the refinement of proneural clusters to specify R8, the first neuron to emerge in the retinal neuroepithelium. Ectopic expression of atonal or removal of rough results in the transformation of a discrete 'equivalence group' of cells into R8s. In addition, ectopic expression of rough blocks atonal expression and proneural cluster formation within the morphogenetic furrow. Thus, rough provides retina-specific regulation to the more general atonal-mediated proneural differentiation pathway. The opposing roles of atonal and rough are not mediated through the Notch pathway, as their expression remains complementary when Notch activity is reduced. These observations suggest that homeobox-containing genes can provide tissue-specific regulation to bHLH factors.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN/aislamiento & purificación , Proteínas de Drosophila , Regulación del Desarrollo de la Expresión Génica , Proteínas Asociadas a Microtúbulos , Células Fotorreceptoras de Invertebrados/citología , Retina/citología , Factores de Transcripción/aislamiento & purificación , Animales , Antígenos de Diferenciación , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Tipificación del Cuerpo , Diferenciación Celular , Regulación hacia Abajo , Drosophila/embriología , Genes Homeobox , Secuencias Hélice-Asa-Hélice , Inmunohistoquímica , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Proteínas del Tejido Nervioso , Células Fotorreceptoras de Invertebrados/embriología , Receptores Notch , Retina/embriología
16.
Curr Opin Neurobiol ; 4(1): 14-20, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8173320

RESUMEN

Cell-cell communication directs the development of photoreceptor cells in the Drosophila retina. A recent set of studies has provided a genetic dissection of one of these interactions, the induction of the R7 photoreceptor cell by the neighboring R8 photoreceptor cell. The results from these experiments shape our understanding of both the molecular basis of signal transduction mediated through receptor tyrosine kinases, as well as the developmental strategies used to generate different cell types in neuronal systems.


Asunto(s)
Drosophila/crecimiento & desarrollo , Drosophila/genética , Células Fotorreceptoras de Invertebrados/fisiología , Retina/citología , Retina/crecimiento & desarrollo , Animales , Comunicación Celular , Transducción de Señal
17.
Perspect Dev Neurobiol ; 2(2): 183-90, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-7728502

RESUMEN

The development of complex tissues, such as the nervous system, requires coordinated interactions between cells, and defining the molecular basis of these interactions is a primary goal of developmental neurobiology. Classic experimental embryological studies of the developmental potential of embryonic neural tissues have more recently been augmented by genetic and molecular approaches to yield a picture of the mechanisms by which such complex neural structures arise during development. Perhaps coincidentally, two of the most well-studied structures in this regard have been the eyes of Drosophila and vertebrates. In the past few years, a number of laboratories have focused on the development of the eye, of both Drosophila and vertebrates in order to understand the role of cell-cell interactions in the generation of organized neural structures. Recent evidence from both the vertebrate and Drosophila models suggests that common mechanisms apply in both systems, particularly in the role of tyrosine kinases in regulating phenotypic choices during histogenesis. Thus, although the eyes of flies and vertebrates have very different appearances, we examine the possibility that similar developmental processes are used to produce tissues with analogous functions.


Asunto(s)
Drosophila/embriología , Transducción de Señal , Vertebrados/embriología , Animales , Humanos , Fenotipo , Proteínas Tirosina Quinasas Receptoras/fisiología , Retina/embriología
18.
Cell ; 69(3): 393-9, 1992 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-1316239

RESUMEN

During Drosophila retinal development, the R8 photo-receptor neuron induces a neighboring cell to assume an R7 cell fate through cell contact. This is mediated by the transmembrane protein bride of sevenless (boss) on the surface of the R8 cell, which binds the sevenless tyrosine kinase receptor (sev) on the surface of the R7 precursor cell. The boss protein, which contains a large extracellular domain, seven transmembrane segments, and a C-terminal cytoplasmic domain, has an exceptional structure for a ligand of a receptor tyrosine kinase. Using a panel of antibodies directed to various cytoplasmic and extracellular epitopes, we demonstrate that the entire boss protein from its extreme N-terminus to its extreme C-terminus is internalized by sev-expressing tissue culture cells and by the R7 precursor cell in the developing eye imaginal disc. The receptor-mediated transfer of a transmembrane ligand represents a novel mechanism for protein transfer between developing cells.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster/fisiología , Proteínas del Ojo/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras , Receptores de Superficie Celular/fisiología , Receptores de Péptidos , Retina/embriología , Animales , Endocitosis , Proteínas del Ojo/química , Proteínas del Ojo/inmunología , Ligandos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/inmunología , Proteínas de la Membrana/química , Proteínas de la Membrana/inmunología , Microscopía Electrónica , Mapeo Peptídico , Unión Proteica
19.
Cell ; 67(6): 1145-55, 1991 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-1760842

RESUMEN

The development of the Drosophila R7 photoreceptor cell is determined by a specific inductive interaction between the R8 photoreceptor cell and a single neighboring precursor cell. This process is mediated by bride of sevenless (boss), a cell-surface bound ligand, and the sevenless (sev) tyrosine kinase receptor. The boss ligand is expressed specifically on the surface of the R8 cell, whereas the sev receptor is expressed on 5 cells contacting the developing R8 cell and other cells not in contact with R8. By altering the spatial and temporal expression of boss, we demonstrate that sev-expressing cells that do not contact R8 can assume an R7 cell fate. By contrast, the sev-expressing precursor cells to the R1-R6 photoreceptor cells that do contact R8 are nonresponsive to the inductive cue. Using the rough and Nspl mutations, we demonstrate that an early commitment to an R1-R6 cell fate blocks the pathway of sev activation in these cells.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster/embriología , Proteínas del Ojo/fisiología , Glicoproteínas de Membrana/fisiología , Proteínas de la Membrana/fisiología , Células Fotorreceptoras/embriología , Proteínas Tirosina Quinasas Receptoras , Receptores de Péptidos , Animales , Clonación Molecular , Inducción Embrionaria , Ojo/embriología , Expresión Génica , Ligandos , Neuronas/citología , Células Fotorreceptoras/citología , Proteínas Tirosina Quinasas/fisiología
20.
Nature ; 352(6332): 207-12, 1991 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-1857416

RESUMEN

During development of the Drosophila retina, the R8 photoreceptor neuron induces a neighbouring cell to assume an R7 cell fate. Genetic data suggest that the induction is mediated by two transmembrane proteins encoded by bride of sevenless and sevenless. A direct interaction between these two proteins was demonstrated by the heterotypic aggregation of cell lines expressing them. In the developing eye the sevenless-dependent internalization of bride of sevenless by the R7 precursor cell provides evidence for a direct interaction between these two proteins in vivo.


Asunto(s)
Proteínas de Drosophila , Drosophila/fisiología , Proteínas del Ojo/fisiología , Glicoproteínas de Membrana , Proteínas de la Membrana/fisiología , Neuronas/fisiología , Células Fotorreceptoras/fisiología , Proteínas Tirosina Quinasas/fisiología , Proteínas Tirosina Quinasas Receptoras , Animales , Agregación Celular , ADN/genética , Drosophila/genética , Proteínas del Ojo/genética , Ligandos , Proteínas de la Membrana/genética , Mosaicismo , Mutación , Neuronas/citología , Células Fotorreceptoras/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA