Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Development ; 149(16)2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35899600

RESUMEN

Phosphatidylinositol (PI) 4,5-bisphosphate (PIP2) is involved in many biological functions. However, the mechanisms of PIP2 in collective cell migration remain elusive. This study highlights the regulatory role of cytidine triphosphate synthase (CTPsyn) in collective border cell migration through regulating the asymmetrical distribution of PIP2. We demonstrated that border cell clusters containing mutant CTPsyn cells suppressed migration. CTPsyn was co-enriched with Actin at the leading edge of the Drosophila border cell cluster where PIP2 was enriched, and this enrichment depended on the CTPsyn activity. Genetic interactions of border cell migration were found between CTPsyn mutant and genes in PI biosynthesis. The CTPsyn reduction resulted in loss of the asymmetric activity of endocytosis recycling. Also, genetic interactions were revealed between components of the exocyst complex and CTPsyn mutant, indicating that CTPsyn activity regulates the PIP2-related asymmetrical exocytosis activity. Furthermore, CTPsyn activity is essential for RTK-polarized distribution in the border cell cluster. We propose a model in which CTPsyn activity is required for the asymmetrical generation of PIP2 to enrich RTK signaling through endocytic recycling in collective cell migration.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Ligasas de Carbono-Nitrógeno , Movimiento Celular/genética , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo
2.
Sci Adv ; 8(29): eabm2411, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35867785

RESUMEN

Collective migration is important to embryonic development and cancer metastasis, but migratory and nonmigratory cell fate discrimination by differential activity of signal pathways remains elusive. In Drosophila oogenesis, Jak/Stat signaling patterns the epithelial cell fates in early egg chambers but later renders motility to clustered border cells. How Jak/Stat signal spatiotemporally switches static epithelia to motile cells is largely unknown. We report that a nuclear protein, Dysfusion, resides on the inner nuclear membrane and interacts with importin α/ß and Nup153 to modulate Jak/Stat signal by attenuating Stat nuclear import. Dysfusion is ubiquitously expressed in oogenesis but specifically down-regulated in border cells when migrating. Increase of nuclear Stat by Dysfusion down-regulation triggers invasive cell behavior and maintains persistent motility. Mammalian homolog of Dysfusion (NPAS4) also negatively regulates the nuclear accumulation of STAT3 and cancer cell migration. Thus, our finding demonstrates that Dysfusion-dependent gating mechanism is conserved and may serve as a therapeutic target for Stat-mediated cancer metastasis.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Movimiento Celular/fisiología , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Mamíferos/metabolismo , Factores de Transcripción STAT/metabolismo
3.
PLoS Negl Trop Dis ; 16(1): e0010084, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35015769

RESUMEN

Dengue fever is one of the most severe viral diseases transmitted by Aedes mosquitoes, with traditional approaches of disease control proving insufficient to prevent significant disease burden. Release of Wolbachia-transinfected mosquitoes offers a promising alternative control methodologies; Wolbachia-transinfected female Aedes aegypti demonstrate reduced dengue virus transmission, whilst Wolbachia-transinfected males cause zygotic lethality when crossed with uninfected females, providing a method for suppressing mosquito populations. Although highly promising, the delicate nature of population control strategies and differences between local species populations means that controlled releases of Wolbachia-transinfected mosquitoes cannot be performed without extensive testing on specific local Ae. aegypti populations. In order to investigate the potential for using Wolbachia to suppress local Ae. aegypti populations in Taiwan, we performed lab-based and semi-field fitness trials. We first transinfected the Wolbachia strain wAlbB into a local Ae. aegypti population (wAlbB-Tw) and found no significant changes in lifespan, fecundity and fertility when compared to controls. In the laboratory, we found that as the proportion of released male mosquitoes carrying Wolbachia was increased, population suppression could reach up to 100%. Equivalent experiments in semi-field experiments found suppression rates of up to 70%. The release of different ratios of wAlbB-Tw males in the semi-field system provided an estimate of the optimal size of male releases. Our results indicate that wAlbB-Tw has significant potential for use in vector control strategies aimed at Ae. aegypti population suppression in Taiwan. Open field release trials are now necessary to confirm that wAlbB-Tw mediated suppression is feasible in natural environments.


Asunto(s)
Aedes/microbiología , Dengue/prevención & control , Control de Mosquitos/métodos , Control Biológico de Vectores/métodos , Wolbachia/metabolismo , Animales , Agentes de Control Biológico/administración & dosificación , Dengue/transmisión , Virus del Dengue/aislamiento & purificación , Femenino , Masculino , Mosquitos Vectores/virología , Taiwán , Wolbachia/clasificación , Cigoto/microbiología
4.
Front Mol Biosci ; 6: 157, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32118029

RESUMEN

The evolutionarily conserved Hippo kinase signaling cascade governs cell proliferation, tissue differentiation and organ size, and can promote tumor growth and cancer metastasis when dysregulated. Unlike conventional signaling pathways driven by ligand-receptor binding to initiate downstream cascades, core Hippo kinases are activated not only by biochemical cues but also by mechanical ones generated from altered cell shape, cell polarity, cell-cell junctions or cell-extracellular matrix adhesion. In this review, we focus on recent advances showing how mechanical force acts through the actin cytoskeleton to regulate the Hippo pathway during cell movement and cancer invasion. We also discuss how this force affects YAP-dependent tissue growth and cell proliferation, and how disruption of that homeostatic relationship contributes to cancer metastasis.

5.
Cell Rep ; 22(8): 2160-2175, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29466741

RESUMEN

In collective cell migration, directional protrusions orient cells in response to external cues, which requires coordinated polarity among the migrating cohort. However, the molecular mechanism has not been well defined. Drosophila border cells (BCs) migrate collectively and invade via the confined space between nurse cells, offering an in vivo model to examine how group polarity is organized. Here, we show that the front/back polarity of BCs requires Rap1, hyperactivation of which disrupts cluster polarity and induces misoriented protrusions and loss of asymmetry in the actin network. Conversely, hypoactive Rap1 causes fewer protrusions and cluster spinning during migration. A forward genetic screen revealed that downregulation of the Hippo (Hpo) pathway core components hpo or mats enhances the Rap1V12-induced migration defect and misdirected protrusions. Mechanistically, association of Rap1V12 with the kinase domain of Hpo suppresses its activity, which releases Hpo signaling-mediated suppression of F-actin elongation, promoting cellular protrusions in collective cell migration.


Asunto(s)
Movimiento Celular , Polaridad Celular , Extensiones de la Superficie Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas de Unión a Telómeros/metabolismo , Actomiosina/metabolismo , Animales , Epistasis Genética , Modelos Biológicos , Complejo Shelterina
6.
Proc Natl Acad Sci U S A ; 110(19): E1734-42, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23610413

RESUMEN

Asymmetric division of stem cells results in both self-renewal and differentiation of daughters. Understanding the molecules and mechanisms that govern differentiation of specific cell types from adult tissue stem cells is a major challenge in developmental biology and regenerative medicine. Drosophila follicle stem cells (FSCs) represent an excellent model system to study adult stem cell behavior; however, the earliest stages of follicle cell differentiation remain largely mysterious. Here we identify Castor (Cas) as a nuclear protein that is expressed in FSCs and early follicle cell precursors and then is restricted to differentiated polar and stalk cells once egg chambers form. Cas is required for FSC maintenance and polar and stalk cell fate specification. Eyes absent (Eya) is excluded from polar and stalk cells and represses their fate by inhibiting Cas expression. Hedgehog signaling is essential to repress Eya to allow Cas expression in polar and stalk cells. Finally, we show that the complementary patterns of Cas and Eya reveal the gradual differentiation of polar and stalk precursor cells at the earliest stages of their development. Our studies provide a marker for cell fates in this model and insight into the molecular and cellular mechanisms by which FSC progeny diverge into distinct fates.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Oogénesis/fisiología , Células Madre/citología , Animales , Linaje de la Célula , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Epistasis Genética , Proteínas del Ojo/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Prueba de Complementación Genética , Inmunohistoquímica , Folículo Ovárico/citología
7.
Nat Cell Biol ; 11(5): 569-79, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19350016

RESUMEN

During development, elaborate patterns of cell differentiation and movement must occur in the correct locations and at the proper times. Developmental timing has been studied less than spatial pattern formation, and the mechanisms integrating the two are poorly understood. Border-cell migration in the Drosophila ovary occurs specifically at stage 9. Timing of the migration is regulated by the steroid hormone ecdysone, whereas spatial patterning of the migratory population requires localized activity of the JAK-STAT pathway. Ecdysone signalling is patterned spatially as well as temporally, although the mechanisms are not well understood. In stage 9 egg chambers, ecdysone signalling is highest in anterior follicle cells including the border cells. We identify the gene abrupt as a repressor of ecdysone signalling and border-cell migration. Abrupt protein is normally lost from border-cell nuclei during stage 9, in response to JAK-STAT activity. This contributes to the spatial pattern of the ecdysone response. Abrupt attenuates ecdysone signalling by means of a direct interaction with the basic helix-loop-helix (bHLH) domain of the P160 ecdysone receptor coactivator Taiman (Tai). Taken together, these findings provide a molecular mechanism by which spatial and temporal cues are integrated.


Asunto(s)
Movimiento Celular/fisiología , Proteínas de Drosophila/fisiología , Proteínas Nucleares/fisiología , Oogénesis/fisiología , Folículo Ovárico/citología , Transducción de Señal/fisiología , Animales , Sitios de Unión/fisiología , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Ecdisona/metabolismo , Femenino , Regulación de la Expresión Génica/fisiología , Quinasas Janus/metabolismo , Modelos Biológicos , Folículo Ovárico/fisiología , Unión Proteica/fisiología , Isoformas de Proteínas/metabolismo , Receptores de Esteroides/metabolismo , Factores de Transcripción STAT/metabolismo , Factores de Transcripción/metabolismo
8.
Nat Protoc ; 2(10): 2467-73, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17947988

RESUMEN

This protocol describes a method for the dissection of egg chambers from intact Drosophila females and culture conditions that permit live imaging of them, with a particular emphasis on stage 9. This stage of development is characterized by oocyte growth and patterning, outer follicle cell rearrangement and migration of border cells. Although in vitro culture of egg chambers of later developmental stages has long been possible, until recently stage 9 egg chambers could only be kept alive for short periods, did not develop normally, and border cell migration failed entirely. We have established culture conditions that support overall egg chamber development including border cell migration in vitro. This protocol makes possible direct observation of molecular and cellular dynamics in both wild-type and mutant egg chambers, and opens the door to testing of pharmacological inhibitors and the use of biosensors. The entire protocol takes approximately 24 h while the preparation of egg chambers for live imaging requires only 15-20 min.


Asunto(s)
Técnicas de Cultivo de Célula , Drosophila melanogaster/citología , Animales , Drosophila melanogaster/crecimiento & desarrollo , Femenino , Oocitos/citología , Oocitos/crecimiento & desarrollo , Folículo Ovárico/citología , Ovario/citología
9.
J Mammary Gland Biol Neoplasia ; 12(2-3): 103-14, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17549611

RESUMEN

Cell motility makes essential contributions to normal embryonic development and homeostasis. It is also thought to contribute in important ways to tumor metastasis. Because of this dual importance, cell migration has been extensively studied. The fruit fly Drosophila melanogaster has served as an important model organism for genetic analysis of many aspects of developmental biology, including cell migration. Here we describe the various types of cell movements that have been studied in detail, which represent models for epithelial-to-mesenchymal transition, transepithelial migration, inflammation, wound healing and invasion. We summarize what has been learned about the molecular control of cell migration from genetic studies in the fly. In addition, we describe recent efforts to model tumor metastasis directly in Drosophila by expressing oncogenes and/or mutating tumor suppressor genes. Together these studies suggest that Drosophila has much to offer as a model for varied aspects of tumor metastasis.


Asunto(s)
Movimiento Celular , Drosophila melanogaster/citología , Modelos Biológicos , Metástasis de la Neoplasia/patología , Animales , Cadherinas/metabolismo , Drosophila melanogaster/inmunología , Drosophila melanogaster/metabolismo , Humanos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA