Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Cell ; 154(3): 664-75, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23911328

RESUMEN

The risk of specific cancers increases in patients with metabolic dysfunction, including obesity and diabetes. Here, we use Drosophila as a model to explore the effects of diet on tumor progression. Feeding Drosophila a diet high in carbohydrates was previously demonstrated to direct metabolic dysfunction, including hyperglycemia, hyperinsulinemia, and insulin resistance. We demonstrate that high dietary sugar also converts Ras/Src-transformed tissue from localized growths to aggressive tumors with emergent metastases. Whereas most tissues displayed insulin resistance, Ras/Src tumors retained insulin pathway sensitivity, increased the ability to import glucose, and resisted apoptosis. High dietary sugar increased canonical Wingless/Wnt pathway activity, which upregulated insulin receptor gene expression to promote insulin sensitivity. The result is a feed-forward circuit that amplified diet-mediated malignant phenotypes within Ras/Src-transformed tumors. By targeting multiple steps in this circuit with rationally applied drug combinations, we demonstrate the potential of combinatorial drug intervention to treat diet-enhanced malignant tumors.


Asunto(s)
Carbohidratos de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Resistencia a la Insulina , Neoplasias/metabolismo , Transducción de Señal , Proteína Wnt1/metabolismo , Animales , Transformación Celular Neoplásica , Dieta Alta en Grasa , Glucosa/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo
2.
PLoS Genet ; 19(6): e1010792, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37267433

RESUMEN

Experimental models that capture the genetic complexity of human disease and allow mechanistic explorations of the underlying cell, tissue, and organ interactions are crucial to furthering our understanding of disease biology. Such models require combinatorial manipulations of multiple genes, often in more than one tissue at once. The ability to perform complex genetic manipulations in vivo is a key strength of Drosophila, where many tools for sophisticated and orthogonal genetic perturbations exist. However, combining the large number of transgenes required to establish more representative disease models and conducting mechanistic studies in these already complex genetic backgrounds is challenging. Here we present a design that pushes the limits of Drosophila genetics by allowing targeted combinatorial ectopic expression and knockdown of multiple genes from a single inducible transgene. The polycistronic transcript encoded by this transgene includes a synthetic short hairpin cluster cloned within an intron placed at the 5' end of the transcript, followed by two protein-coding sequences separated by the T2A sequence that mediates ribosome skipping. This technology is particularly useful for modeling genetically complex diseases like cancer, which typically involve concurrent activation of multiple oncogenes and loss of multiple tumor suppressors. Furthermore, consolidating multiple genetic perturbations into a single transgene further streamlines the ability to perform combinatorial genetic manipulations and makes it readily adaptable to a broad palette of transgenic systems. This flexible design for combinatorial genetic perturbations will also be a valuable tool for functionally exploring multigenic gene signatures identified from omics studies of human disease and creating humanized Drosophila models to characterize disease-associated variants in human genes. It can also be adapted for studying biological processes underlying normal tissue homeostasis and development that require simultaneous manipulation of many genes.


Asunto(s)
Drosophila , Técnicas Genéticas , Animales , Humanos , Drosophila/genética , Transgenes , Animales Modificados Genéticamente , Intrones
3.
Nucleic Acids Res ; 47(W1): W183-W190, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31069376

RESUMEN

High-throughput experiments produce increasingly large datasets that are difficult to analyze and integrate. While most data integration approaches focus on aligning metadata, data integration can be achieved by abstracting experimental results into gene sets. Such gene sets can be made available for reuse through gene set enrichment analysis tools such as Enrichr. Enrichr currently only supports gene sets compiled from human and mouse, limiting accessibility for investigators that study other model organisms. modEnrichr is an expansion of Enrichr for four model organisms: fish, fly, worm and yeast. The gene set libraries within FishEnrichr, FlyEnrichr, WormEnrichr and YeastEnrichr are created from the Gene Ontology, mRNA expression profiles, GeneRIF, pathway databases, protein domain databases and other organism-specific resources. Additionally, libraries were created by predicting gene function from RNA-seq co-expression data processed uniformly from the gene expression omnibus for each organism. The modEnrichr suite of tools provides the ability to convert gene lists across species using an ortholog conversion tool that automatically detects the species. For complex analyses, modEnrichr provides API access that enables submitting batch queries. In summary, modEnrichr leverages existing model organism databases and other resources to facilitate comprehensive hypothesis generation through data integration.


Asunto(s)
Bases de Datos Genéticas , Expresión Génica/genética , Biblioteca de Genes , Biblioteca Genómica , Programas Informáticos , Animales , Biología Computacional , Ontología de Genes , Humanos , Metadatos
4.
Nat Chem Biol ; 14(3): 291-298, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29355849

RESUMEN

Synthetic tailoring of approved drugs for new indications is often difficult, as the most appropriate targets may not be readily apparent, and therefore few roadmaps exist to guide chemistry. Here, we report a multidisciplinary approach for accessing novel target and chemical space starting from an FDA-approved kinase inhibitor. By combining chemical and genetic modifier screening with computational modeling, we identify distinct kinases that strongly enhance ('pro-targets') or limit ('anti-targets') whole-animal activity of the clinical kinase inhibitor sorafenib in a Drosophila medullary thyroid carcinoma (MTC) model. We demonstrate that RAF-the original intended sorafenib target-and MKNK kinases function as pharmacological liabilities because of inhibitor-induced transactivation and negative feedback, respectively. Through progressive synthetic refinement, we report a new class of 'tumor calibrated inhibitors' with unique polypharmacology and strongly improved therapeutic index in fly and human MTC xenograft models. This platform provides a rational approach to creating new high-efficacy and low-toxicity drugs.


Asunto(s)
Carcinoma Neuroendocrino/metabolismo , Carcinoma/metabolismo , Drosophila/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Tiroides/metabolismo , Animales , Animales Modificados Genéticamente , Línea Celular Tumoral , Movimiento Celular , Modelos Animales de Enfermedad , Diseño de Fármacos , Femenino , Células HCT116 , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Simulación del Acoplamiento Molecular , Trasplante de Neoplasias , Isoformas de Proteínas , Proteínas Proto-Oncogénicas c-raf/metabolismo , Transducción de Señal , Sorafenib/farmacología
5.
PLoS Comput Biol ; 15(4): e1006878, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31026276

RESUMEN

Drosophila provides an inexpensive and quantitative platform for measuring whole animal drug response. A complementary approach is virtual screening, where chemical libraries can be efficiently screened against protein target(s). Here, we present a unique discovery platform integrating structure-based modeling with Drosophila biology and organic synthesis. We demonstrate this platform by developing chemicals targeting a Drosophila model of Medullary Thyroid Cancer (MTC) characterized by a transformation network activated by oncogenic dRetM955T. Structural models for kinases relevant to MTC were generated for virtual screening to identify unique preliminary hits that suppressed dRetM955T-induced transformation. We then combined features from our hits with those of known inhibitors to create a 'hybrid' molecule with improved suppression of dRetM955T transformation. Our platform provides a framework to efficiently explore novel kinase inhibitors outside of explored inhibitor chemical space that are effective in inhibiting cancer networks while minimizing whole body toxicity.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Neuroendocrino , Evaluación Preclínica de Medicamentos/métodos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas , Neoplasias de la Tiroides , Animales , Carcinoma Neuroendocrino/enzimología , Carcinoma Neuroendocrino/metabolismo , Biología Computacional/métodos , Drosophila , Modelos Biológicos , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/metabolismo , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/metabolismo , Neoplasias de la Tiroides/enzimología , Neoplasias de la Tiroides/metabolismo
6.
Nature ; 486(7401): 80-4, 2012 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22678283

RESUMEN

The complexity of cancer has led to recent interest in polypharmacological approaches for developing kinase-inhibitor drugs; however, optimal kinase-inhibition profiles remain difficult to predict. Using a Ret-kinase-driven Drosophila model of multiple endocrine neoplasia type 2 and kinome-wide drug profiling, here we identify that AD57 rescues oncogenic Ret-induced lethality, whereas related Ret inhibitors imparted reduced efficacy and enhanced toxicity. Drosophila genetics and compound profiling defined three pathways accounting for the mechanistic basis of efficacy and dose-limiting toxicity. Inhibition of Ret plus Raf, Src and S6K was required for optimal animal survival, whereas inhibition of the 'anti-target' Tor led to toxicity owing to release of negative feedback. Rational synthetic tailoring to eliminate Tor binding afforded AD80 and AD81, compounds featuring balanced pathway inhibition, improved efficacy and low toxicity in Drosophila and mammalian multiple endocrine neoplasia type 2 models. Combining kinase-focused chemistry, kinome-wide profiling and Drosophila genetics provides a powerful systems pharmacology approach towards developing compounds with a maximal therapeutic index.


Asunto(s)
Terapia Molecular Dirigida , Neoplasia Endocrina Múltiple Tipo 2b/tratamiento farmacológico , Neoplasia Endocrina Múltiple Tipo 2b/genética , Polifarmacia , Animales , Bencenosulfonatos/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/genética , Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/efectos adversos , Compuestos Heterocíclicos de 4 o más Anillos/química , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Masculino , Neoplasia Endocrina Múltiple Tipo 2b/enzimología , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Piridinas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sorafenib , Tasa de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
7.
EMBO Rep ; 14(2): 172-7, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23306653

RESUMEN

Tumours evolve several mechanisms to evade apoptosis, yet many resected carcinomas show significantly elevated caspase activity. Moreover, caspase activity is positively correlated with tumour aggression and adverse patient outcome. These observations indicate that caspases might have a functional role in promoting tumour invasion and metastasis. Using a Drosophila model of invasion, we show that precise effector caspase activity drives cell invasion without initiating apoptosis. Affected cells express the matrix metalloprotinase Mmp1 and invade by activating Jnk. Our results link Jnk and effector caspase signalling during the invasive process and suggest that tumours under apoptotic stresses from treatment, immune surveillance or intrinsic signals might be induced further along the metastatic cascade.


Asunto(s)
Caspasa 3/metabolismo , Movimiento Celular , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Transducción de Señal , Alas de Animales/patología , Animales , Apoptosis , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Activación Enzimática , Humanos , Metaloproteinasa 1 de la Matriz/metabolismo , Invasividad Neoplásica , Neoplasias/patología , Neuropéptidos/metabolismo
8.
Oncogenesis ; 13(1): 1, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38172609

RESUMEN

Throughout an individual's life, somatic cells acquire cancer-associated mutations. A fraction of these mutations trigger tumour formation, a phenomenon partly driven by the interplay of mutant and wild-type cell clones competing for dominance; conversely, other mutations function against tumour initiation. This mechanism of 'cell competition', can shift clone dynamics by evaluating the relative status of clonal populations, promoting 'winners' and eliminating 'losers'. This review examines the role of cell competition in the context of tumorigenesis, tumour progression and therapeutic intervention.

9.
Dis Model Mech ; 17(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38721669

RESUMEN

Accounting for 10-20% of breast cancer cases, triple-negative breast cancer (TNBC) is associated with a disproportionate number of breast cancer deaths. One challenge in studying TNBC is its genomic profile: with the exception of TP53 loss, most breast cancer tumors are characterized by a high number of copy number alterations (CNAs), making modeling the disease in whole animals challenging. We computationally analyzed 186 CNA regions previously identified in breast cancer tumors to rank genes within each region by likelihood of acting as a tumor driver. We then used a Drosophila p53-Myc TNBC model to identify 48 genes as functional drivers. To demonstrate the utility of this functional database, we established six 3-hit models; altering candidate genes led to increased aspects of transformation as well as resistance to the chemotherapeutic drug fluorouracil. Our work provides a functional database of CNA-associated TNBC drivers, and a template for an integrated computational/whole-animal approach to identify functional drivers of transformation and drug resistance within CNAs in other tumor types.


Asunto(s)
Variaciones en el Número de Copia de ADN , Modelos Animales de Enfermedad , Neoplasias de la Mama Triple Negativas , Animales , Variaciones en el Número de Copia de ADN/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Femenino , Drosophila melanogaster/genética , Humanos , Resistencia a Antineoplásicos/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Transformación Celular Neoplásica/genética
10.
BMC Genomics ; 14: 136, 2013 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-23445342

RESUMEN

BACKGROUND: Genome-wide association studies (GWAS) identify regions of the genome that are associated with particular traits, but do not typically identify specific causative genetic elements. For example, while a large number of single nucleotide polymorphisms associated with type 2 diabetes (T2D) and related traits have been identified by human GWAS, only a few genes have functional evidence to support or to rule out a role in cellular metabolism or dietary interactions. Here, we use a recently developed Drosophila model in which high-sucrose feeding induces phenotypes similar to T2D to assess orthologs of human GWAS-identified candidate genes for risk of T2D and related traits. RESULTS: Disrupting orthologs of certain T2D candidate genes (HHEX, THADA, PPARG, KCNJ11) led to sucrose-dependent toxicity. Tissue-specific knockdown of the HHEX ortholog dHHEX (CG7056) directed metabolic defects and enhanced lethality; for example, fat-body-specific loss of dHHEX led to increased hemolymph glucose and reduced insulin sensitivity. CONCLUSION: Candidate genes identified in human genetic studies of metabolic traits can be prioritized and functionally characterized using a simple Drosophila approach. To our knowledge, this is the first large-scale effort to study the functional interaction between GWAS-identified candidate genes and an environmental risk factor such as diet in a model organism system.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Proteínas de Drosophila/genética , Estudio de Asociación del Genoma Completo , Proteínas de Homeodominio/genética , Proteínas Musculares/genética , Factores de Transcripción/genética , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Cuerpo Adiposo/metabolismo , Cuerpo Adiposo/patología , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Glucosa/genética , Glucosa/metabolismo , Humanos , Resistencia a la Insulina/genética , Especificidad de Órganos , Fenotipo , Polimorfismo de Nucleótido Simple
11.
Drug Discov Today Technol ; 10(1): e65-71, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24050232

RESUMEN

Thyroid neoplasias represent among the fastest growing cancers. While surgery has become the treatment of choice for most thyroid tumors, many require chemotherapy. In this review, we examine the contributions of work in the fruit fly Drosophila toward multiple endocrine neoplasia type 2 (MEN2), a Ret-based disease to which recent Drosophila models have proven useful both for understanding disease mechanism as well as helping identify new generation therapeutics.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila/genética , Proteínas Proto-Oncogénicas c-ret/genética , Neoplasias de la Tiroides/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Evaluación Preclínica de Medicamentos , Humanos , Piperidinas/farmacología , Piperidinas/uso terapéutico , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Neoplasias de la Tiroides/genética
12.
Dev Dyn ; 241(1): 107-18, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22038952

RESUMEN

Cancer is driven by complex genetic and cellular mechanisms. Recently, the Drosophila community has become increasingly interested in exploring cancer issues. The Drosophila field has made seminal contributions to many of the mechanisms that are fundamental to the cancer process; several of these mechanisms have already been validated in vertebrates. Less well known are the Drosophila field's early direct contributions to the cancer field: some of the earliest tumor suppressors were identified in flies. In this review, we identify major contributions that Drosophila studies have made toward dissecting the pathways and mechanisms underlying tumor progression. We also highlight areas, such as drug discovery, where we expect Drosophila studies to make a major scientific impact in the future.


Asunto(s)
Drosophila melanogaster/fisiología , Modelos Animales , Neoplasias/fisiopatología , Animales , Polaridad Celular , Cromatina/metabolismo , Progresión de la Enfermedad , Descubrimiento de Drogas , Humanos , Mamíferos , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/patología , Microambiente Tumoral
13.
Dev Dyn ; 241(12): 1933-43, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23027549

RESUMEN

BACKGROUND: Morphogenetic modeling of tissues requires coordinated regulation of adhesion. For its correct patterning, the Drosophila pupal eye requires several Immunoglobulin superfamily cell adhesion molecules (IgCAMs) and the adaptor protein Cindr. Orthologs of these proteins are essential components of specialized junctions of the vertebrate kidney; the Cindr ortholog Cd2ap is essential for the integrity of this structure. RESULTS: Reducing Cindr during fly eye development led to incorrect distribution of the IgCAMs Roughest (Rst) and Hibris (Hbs). Both bound Cindr. Disrupting endocytosis similarly led to Rst and Hbs mis-localization; our data suggests an additional early requirement for endocytosis in regulating Hbs localization or stability. Finally, Rst and Hbs localized correctly only when in stable membrane complexes and we propose that Cindr anchors these to the cytoskeleton. This regulation likely does not extend to IgCAMs Kin of irre (Kirre) and Sticks and stones (Sns) in the pupal eye; neither interacted with Cindr in in vitro assays. Nonetheless, Kirre and Sns partially mis-localized when Cindr was reduced, possibly due to interactions with Rst/Hbs. CONCLUSIONS: Our data suggests Cindr recapitulates both proposed functions of its mammalian orthologs Cd2ap and Cin85: targeting the IgCAMs Rst and Hbs for endocytosis and stabilizing these heterophilic IgCAM complexes.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas del Ojo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Endocitosis/fisiología , Proteínas del Ojo/genética , Proteínas de la Membrana/genética , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Proteínas Musculares/metabolismo
14.
J Invest Dermatol ; 143(8): 1378-1387, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37330719

RESUMEN

Neurofibromatosis type 1 (NF1) is caused by a nonfunctional copy of the NF1 tumor suppressor gene that predisposes patients to the development of cutaneous neurofibromas (cNFs), the skin tumor that is the hallmark of this condition. Innumerable benign cNFs, each appearing by an independent somatic inactivation of the remaining functional NF1 allele, form in nearly all patients with NF1. One of the limitations in developing a treatment for cNFs is an incomplete understanding of the underlying pathophysiology and limitations in experimental modeling. Recent advances in preclinical in vitro and in vivo modeling have substantially enhanced our understanding of cNF biology and created unprecedented opportunities for therapeutic discovery. We discuss the current state of cNF preclinical in vitro and in vivo model systems, including two- and three-dimensional cell cultures, organoids, genetically engineered mice, patient-derived xenografts, and porcine models. We highlight the models' relationship to human cNFs and how they can be used to gain insight into cNF development and therapeutic discovery.


Asunto(s)
Neurofibroma , Neurofibromatosis 1 , Neoplasias Cutáneas , Ratones , Humanos , Animales , Porcinos , Neurofibromatosis 1/genética , Neurofibromatosis 1/terapia , Mutación , Neurofibroma/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Alelos
15.
J Invest Dermatol ; 143(8): 1358-1368, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37245145

RESUMEN

Cutaneous neurofibromas (cNFs) are the most common tumor in people with the rasopathy neurofibromatosis type 1. They number in hundreds or even thousands throughout the body, and currently, there are no effective interventions to prevent or treat these skin tumors. To facilitate the identification of novel and effective therapies, essential studies including a more refined understanding of cNF biology and the role of RAS signaling and downstream effector pathways responsible for cNF initiation, growth, and maintenance are needed. This review highlights the current state of knowledge of RAS signaling in cNF pathogenesis and therapeutic development for cNF treatment.


Asunto(s)
Neurofibroma , Neurofibromatosis 1 , Neoplasias Cutáneas , Humanos , Neurofibroma/metabolismo , Neurofibroma/patología , Neurofibromatosis 1/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Transducción de Señal
16.
Dev Biol ; 344(2): 948-56, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20599904

RESUMEN

In the Drosophila eye, neighboring ommatidia are separated by inter-ommatidial cells (IOCs). How this ommatidial spacing emerges during eye development is not clear. Here we demonstrate that four adhesion molecules of the Irre cell recognition module (IRM) family play a redundant role in maintaining separation of ommatidia. The four IRM proteins are divided into two groups: Kirre and Rst are expressed in IOCs, and Hbs and Sns in primary pigment cells (1 degrees s). Kirre binds Hbs and Sns in vivo and in vitro. Reducing activity of either Rst or Kirre alone had minimal effects on ommatidial spacing, but reducing both together led to direct ommatidium:ommatidium contact. A similar phenotype was also observed when reducing both Hbs and Sns. Consistent with the role of these factors in sorting ommatidia, mis-expression of Hbs plus Sns within a single IOC led to complete separation of the cell from neighboring ommatidia. Our results indicate mutual preferential adhesion between ommatidia and IOCs mediated by four IRM proteins is both necessary and sufficient to maintain separation of ommatidia.


Asunto(s)
Drosophila/genética , Drosophila/metabolismo , Ojo/crecimiento & desarrollo , Ojo/metabolismo , Animales , Fenotipo
17.
Dev Cell ; 10(1): 33-44, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16399076

RESUMEN

The construction and maintenance of normal epithelia relies on local signals that guide cells into their proper niches and remove unwanted cells. Failure to execute this process properly may result in aberrant development or diseases, including cancer and associated metastasis. Here, we show that local environment influences the behavior of dCsk-deficient cells. Broad loss of dCsk led to enlarged and mispatterned tissues due to overproliferation, a block in apoptosis, and decreased cadherin-mediated adhesion. Loss of dCsk in discrete patches led to a different outcome: epithelial exclusion, invasive migration, and apoptotic death. These latter phenotypes required sharp differences in dCsk activity between neighbors; dE-cadherin, P120-catenin, Rho1, JNK, and MMP2 mediated this signal. Together, our data demonstrate how the cellular microenvironment plays a central role in determining the outcome of altered dCsk activity, and reveal a role for P120-catenin in a mechanism that protects epithelial integrity by removing abnormal cells.


Asunto(s)
Apoptosis/fisiología , Movimiento Celular/fisiología , Células Epiteliales/fisiología , Epitelio/metabolismo , Proteínas Tirosina Quinasas/deficiencia , Animales , Animales Modificados Genéticamente , Armadillos/metabolismo , Proteína Tirosina Quinasa CSK , Cateninas , Moléculas de Adhesión Celular/metabolismo , Drosophila , Proteínas de Drosophila , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica/métodos , Etiquetado Corte-Fin in Situ , MAP Quinasa Quinasa 4/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Microscopía Electrónica de Rastreo/métodos , Modelos Biológicos , Mutagénesis , Fosfoproteínas/metabolismo , Células Fotorreceptoras de Invertebrados/crecimiento & desarrollo , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/ultraestructura , Retina/crecimiento & desarrollo , Retina/metabolismo , Retina/ultraestructura , Factores de Tiempo , Alas de Animales/crecimiento & desarrollo , beta-Galactosidasa , Proteínas de Unión al GTP rho/metabolismo , Familia-src Quinasas , Catenina delta
18.
Dev Cell ; 10(4): 497-508, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16580994

RESUMEN

Cell migration within a natural context is tightly controlled, often by specific transcription factors. However, the switch from stationary to migratory behavior is poorly understood. Border cells perform a spatially and temporally controlled invasive migration during Drosophila oogenesis. Slbo, a C/EBP family transcriptional activator, is required for them to become migratory. We purified wild-type and slbo mutant border cells as well as nonmigratory follicle cells and performed comparative whole-genome expression profiling, followed by functional tests of the contributions of identified targets to migration. About 300 genes were significantly upregulated in border cells, many dependent on Slbo. Among these, the microtubule regulator Stathmin was strongly upregulated and was required for normal migration. Actin cytoskeleton regulators were also induced, including, surprisingly, a large cluster of "muscle-specific" genes. We conclude that Slbo induces multiple cytoskeletal effectors, and that each contributes to the behavioral changes in border cells.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/fisiología , Movimiento Celular/fisiología , Proteínas de Drosophila/fisiología , Perfilación de la Expresión Génica , Oogénesis/fisiología , Ovario/fisiología , Factores de Transcripción/fisiología , Transcripción Genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas del Citoesqueleto/fisiología , Drosophila , Proteínas de Drosophila/genética , Femenino , Oogénesis/genética , Ovario/citología , Ovario/metabolismo , Estatmina/fisiología , Factores de Transcripción/genética , Regulación hacia Arriba
19.
Curr Opin Nephrol Hypertens ; 20(4): 409-15, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21610496

RESUMEN

PURPOSE OF REVIEW: The functioning kidney requires proper organization in multiple cell types that mediate filtration and removal of wastes. Interest has increasingly focused on the podocyte as an important mediator of kidney function; defects in podocyte function likely mediate a broad palate of kidney dysfunctions. Here I explore recent work that establishes the Drosophila nephrocyte as a useful model for podocyte function and dysfunction. RECENT FINDINGS: Although described many decades in the past, recent evidence has emphasized important similarities in the molecules that construct the 'nephrocyte diaphragm' and its vertebrate cousin the 'podocyte diaphragm'. For example, loss of Nephrin and its associated proteins lead to collapse of these structures and loss of proper filtration. SUMMARY: These data emphasize both differences between the podocyte and nephrocyte and also useful similarities. These similarities provide the promise of bringing Drosophila genetics--strongly successful in other disciplines--to the complex problem of how podocyte dysfunction leads to disease. To further explore this point I discuss work on Nephrin in a better understood tissue, the Drosophila eye, in which the role of Nephrin and its connection to actin dynamics is under intense study.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Riñón/metabolismo , Podocitos/metabolismo , Animales , Drosophila/anatomía & histología , Drosophila/genética , Proteínas de Drosophila/genética , Genotipo , Humanos , Riñón/citología , Proteínas de la Membrana/metabolismo , Modelos Animales , Proteínas Musculares/metabolismo , Fenotipo
20.
PLoS Comput Biol ; 6: e1000841, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20617161

RESUMEN

We present a computer simulation and associated experimental validation of assembly of glial-like support cells into the interweaving hexagonal lattice that spans the Drosophila pupal eye. This process of cell movements organizes the ommatidial array into a functional pattern. Unlike earlier simulations that focused on the arrangements of cells within individual ommatidia, here we examine the local movements that lead to large-scale organization of the emerging eye field. Simulations based on our experimental observations of cell adhesion, cell death, and cell movement successfully patterned a tracing of an emerging wild-type pupal eye. Surprisingly, altering cell adhesion had only a mild effect on patterning, contradicting our previous hypothesis that the patterning was primarily the result of preferential adhesion between IRM-class surface proteins. Instead, our simulations highlighted the importance of programmed cell death (PCD) as well as a previously unappreciated variable: the expansion of cells' apical surface areas, which promoted rearrangement of neighboring cells. We tested this prediction experimentally by preventing expansion in the apical area of individual cells: patterning was disrupted in a manner predicted by our simulations. Our work demonstrates the value of combining computer simulation with in vivo experiments to uncover novel mechanisms that are perpetuated throughout the eye field. It also demonstrates the utility of the Glazier-Graner-Hogeweg model (GGH) for modeling the links between local cellular interactions and emergent properties of developing epithelia as well as predicting unanticipated results in vivo.


Asunto(s)
Ojo Compuesto de los Artrópodos , Simulación por Computador , Drosophila/crecimiento & desarrollo , Morfogénesis/fisiología , Pupa/crecimiento & desarrollo , Animales , Apoptosis/genética , Apoptosis/fisiología , Adhesión Celular , Movimiento Celular , Proliferación Celular , Ojo Compuesto de los Artrópodos/citología , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Ojo Compuesto de los Artrópodos/ultraestructura , Microscopía Electrónica de Transmisión , Modelos Biológicos , Mutación , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA