Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
PLoS Biol ; 21(6): e3002172, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37379333

RESUMEN

The basement membrane (BM) is a specialized extracellular matrix (ECM), which underlies or encases developing tissues. Mechanical properties of encasing BMs have been shown to profoundly influence the shaping of associated tissues. Here, we use the migration of the border cells (BCs) of the Drosophila egg chamber to unravel a new role of encasing BMs in cell migration. BCs move between a group of cells, the nurse cells (NCs), that are enclosed by a monolayer of follicle cells (FCs), which is, in turn, surrounded by a BM, the follicle BM. We show that increasing or reducing the stiffness of the follicle BM, by altering laminins or type IV collagen levels, conversely affects BC migration speed and alters migration mode and dynamics. Follicle BM stiffness also controls pairwise NC and FC cortical tension. We propose that constraints imposed by the follicle BM influence NC and FC cortical tension, which, in turn, regulate BC migration. Encasing BMs emerge as key players in the regulation of collective cell migration during morphogenesis.


Asunto(s)
Colágeno Tipo IV , Drosophila , Animales , Constricción , Membrana Basal/metabolismo , Colágeno Tipo IV/metabolismo , Movimiento Celular , Drosophila/metabolismo
2.
PLoS Genet ; 17(8): e1009738, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34411095

RESUMEN

Activation of Ras signaling occurs in ~30% of human cancers. However, activated Ras alone is insufficient to produce malignancy. Thus, it is imperative to identify those genes cooperating with activated Ras in driving tumoral growth. In this work, we have identified a novel EGFR inhibitor, which we have named EGFRAP, for EGFR adaptor protein. Elimination of EGFRAP potentiates activated Ras-induced overgrowth in the Drosophila wing imaginal disc. We show that EGFRAP interacts physically with the phosphorylated form of EGFR via its SH2 domain. EGFRAP is expressed at high levels in regions of maximal EGFR/Ras pathway activity, such as at the presumptive wing margin. In addition, EGFRAP expression is up-regulated in conditions of oncogenic EGFR/Ras activation. Normal and oncogenic EGFR/Ras-mediated upregulation of EGRAP levels depend on the Notch pathway. We also find that elimination of EGFRAP does not affect overall organogenesis or viability. However, simultaneous downregulation of EGFRAP and its ortholog PVRAP results in defects associated with increased EGFR function. Based on these results, we propose that EGFRAP is a new negative regulator of the EGFR/Ras pathway, which, while being required redundantly for normal morphogenesis, behaves as an important modulator of EGFR/Ras-driven tissue hyperplasia. We suggest that the ability of EGFRAP to functionally inhibit the EGFR pathway in oncogenic cells results from the activation of a feedback loop leading to increase EGFRAP expression. This could act as a surveillance mechanism to prevent excessive EGFR activity and uncontrolled cell growth.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Genes ras/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Ciclo Celular , Proliferación Celular/genética , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Genes ras/fisiología , Discos Imaginales/metabolismo , Morfogénesis , Fosforilación , Receptores de Péptidos de Invertebrados/antagonistas & inhibidores , Receptores de Péptidos de Invertebrados/genética , Receptores de Péptidos de Invertebrados/metabolismo , Transducción de Señal/genética , Proteínas ras/genética
3.
PLoS Genet ; 16(6): e1008717, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32479493

RESUMEN

Forces generated by the actomyosin cytoskeleton are key contributors to many morphogenetic processes. The actomyosin cytoskeleton organises in different types of networks depending on intracellular signals and on cell-cell and cell-extracellular matrix (ECM) interactions. However, actomyosin networks are not static and transitions between them have been proposed to drive morphogenesis. Still, little is known about the mechanisms that regulate the dynamics of actomyosin networks during morphogenesis. This work uses the Drosophila follicular epithelium, real-time imaging, laser ablation and quantitative analysis to study the role of integrins on the regulation of basal actomyosin networks organisation and dynamics and the potential contribution of this role to cell shape. We find that elimination of integrins from follicle cells impairs F-actin recruitment to basal medial actomyosin stress fibers. The available F-actin redistributes to the so-called whip-like structures, present at tricellular junctions, and into a new type of actin-rich protrusions that emanate from the basal cortex and project towards the medial region. These F-actin protrusions are dynamic and changes in total protrusion area correlate with periodic cycles of basal myosin accumulation and constriction pulses of the cell membrane. Finally, we find that follicle cells lacking integrin function show increased membrane tension and reduced basal surface. Furthermore, the actin-rich protrusions are responsible for these phenotypes as their elimination in integrin mutant follicle cells rescues both tension and basal surface defects. We thus propose that the role of integrins as regulators of stress fibers plays a key role on controlling epithelial cell shape, as integrin disruption promotes reorganisation into other types of actomyosin networks, in a manner that interferes with proper expansion of epithelial basal surfaces.


Asunto(s)
Actomiosina/metabolismo , Forma de la Célula , Proteínas de Drosophila/metabolismo , Células Epiteliales/metabolismo , Integrinas/metabolismo , Fibras de Estrés/metabolismo , Animales , Membrana Celular/metabolismo , Drosophila , Células Epiteliales/citología , Fibras de Estrés/ultraestructura
4.
BMC Biol ; 20(1): 90, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35459165

RESUMEN

BACKGROUND: The dynamics of the actomyosin machinery is at the core of many important biological processes. Several relevant cellular responses such as the rhythmic compression of the cell cortex are governed, at a mesoscopic level, by the nonlinear interaction between actin monomers, actin crosslinkers, and myosin motors. Coarse-grained models are an optimal tool to study actomyosin systems, since they can include processes that occur at long time and space scales, while maintaining the most relevant features of the molecular interactions. RESULTS: Here, we present a coarse-grained model of a two-dimensional actomyosin cortex, adjacent to a three-dimensional cytoplasm. Our simplified model incorporates only well-characterized interactions between actin monomers, actin crosslinkers and myosin, and it is able to reproduce many of the most important aspects of actin filament and actomyosin network formation, such as dynamics of polymerization and depolymerization, treadmilling, network formation, and the autonomous oscillatory dynamics of actomyosin. CONCLUSIONS: We believe that the present model can be used to study the in vivo response of actomyosin networks to changes in key parameters of the system, such as alterations in the attachment of actin filaments to the cell cortex.


Asunto(s)
Actinas , Actomiosina , Citoesqueleto de Actina , Modelos Biológicos , Miosinas
5.
PLoS Genet ; 14(9): e1007483, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30260959

RESUMEN

Basement membranes (BMs) are thin sheet-like specialized extracellular matrices found at the basal surface of epithelia and endothelial tissues. They have been conserved across evolution and are required for proper tissue growth, organization, differentiation and maintenance. The major constituents of BMs are two independent networks of Laminin and Type IV Collagen in addition to the proteoglycan Perlecan and the glycoprotein Nidogen/entactin (Ndg). The ability of Ndg to bind in vitro Collagen IV and Laminin, both with key functions during embryogenesis, anticipated an essential role for Ndg in morphogenesis linking the Laminin and Collagen IV networks. This was supported by results from cultured embryonic tissue experiments. However, the fact that elimination of Ndg in C. elegans and mice did not affect survival strongly questioned this proposed linking role. Here, we have isolated mutations in the only Ndg gene present in Drosophila. We find that while, similar to C.elegans and mice, Ndg is not essential for overall organogenesis or viability, it is required for appropriate fertility. We also find, alike in mice, tissue-specific requirements of Ndg for proper assembly and maintenance of certain BMs, namely those of the adipose tissue and flight muscles. In addition, we have performed a thorough functional analysis of the different Ndg domains in vivo. Our results support an essential requirement of the G3 domain for Ndg function and unravel a new key role for the Rod domain in regulating Ndg incorporation into BMs. Furthermore, uncoupling of the Laminin and Collagen IV networks is clearly observed in the larval adipose tissue in the absence of Ndg, indeed supporting a linking role. In light of our findings, we propose that BM assembly and/or maintenance is tissue-specific, which could explain the diverse requirements of a ubiquitous conserved BM component like Nidogen.


Asunto(s)
Membrana Basal/fisiología , Proteínas de Drosophila/fisiología , Drosophila/fisiología , Glicoproteínas de Membrana/fisiología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Animales Modificados Genéticamente , Femenino , Fertilidad/fisiología , Masculino , Músculos/citología , Músculos/metabolismo , Mutación , Especificidad de Órganos/fisiología , Organogénesis/fisiología , Dominios Proteicos/fisiología
6.
Semin Cell Dev Biol ; 70: 58-64, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28860103

RESUMEN

The impact that research has on shaping the future of societies is perhaps as significant as never before. One of the problems for most regions in Africa is poor quality and quantity of research-based education, as well as low level of funding. Hence, African researchers produce only around one percent of the world's research. We believe that research with Drosophila melanogaster can contribute to changing that. As seen before in other places, Drosophila can be used as a powerful and cost-effective model system to scale-up and improve both academia and research output. The DrosAfrica project was founded to train and establish a connected community of researchers using Drosophila as a model system to investigate biomedical problems in Africa. Since founding, the project has trained eighty scientists from numerous African countries, and continues to grow. Here, we describe the DrosAfrica project, its conception and its mission. We also give detailed insights into DrosAfrica's approaches to achieve its aims, as well as future perspectives, and opportunities beyond Africa.


Asunto(s)
Investigación Biomédica/educación , Drosophila melanogaster/genética , Educación/organización & administración , Comunicación Interdisciplinaria , Investigación Biomédica Traslacional/educación , África , Animales , Investigación Biomédica/economía , Investigación Biomédica/métodos , Modelos Animales de Enfermedad , Drosophila melanogaster/metabolismo , Humanos , Red Social , Crecimiento Sostenible , Investigación Biomédica Traslacional/economía , Investigación Biomédica Traslacional/métodos
7.
PLoS Genet ; 12(1): e1005763, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26808525

RESUMEN

The extracellular matrix (ECM) is a pivotal component adult tissues and of many tissue-specific stem cell niches. It provides structural support and regulates niche signaling during tissue maintenance and regeneration. In many tissues, ECM remodeling depends on the regulation of MMP (matrix metalloproteinase) activity by inhibitory TIMP (tissue inhibitors of metalloproteinases) proteins. Here, we report that the only Drosophila timp gene is required for maintaining the normal organization and function of the germline stem cell niche in adult females. timp mutant ovaries show reduced levels of both Drosophila Collagen IV α chains. In addition, tissue stiffness and the cellular organization of the ovarian niche are affected in timp mutants. Finally, loss of timp impairs the ability of the germline stem cell niche to generate new cysts. Our results demonstrating a crucial role for timp in tissue organization and gamete production thus provide a link between the regulation of ECM metabolism and tissue homeostasis.


Asunto(s)
Matriz Extracelular/metabolismo , Ovario/metabolismo , Nicho de Células Madre/genética , Inhibidores Tisulares de Metaloproteinasas/genética , Animales , Colágeno Tipo IV/genética , Drosophila , Matriz Extracelular/genética , Femenino , Células Germinativas , Metaloproteinasas de la Matriz/genética , Ovario/crecimiento & desarrollo
8.
J Cell Sci ; 127(Pt 14): 3162-73, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24794494

RESUMEN

Muscle differentiation requires the assembly of high-order structures called myofibrils, composed of sarcomeres. Even though the molecular organization of sarcomeres is well known, the mechanisms underlying myofibrillogenesis are poorly understood. It has been proposed that integrin-dependent adhesion nucleates myofibrils at the periphery of the muscle cell to sustain sarcomere assembly. Here, we report a role for the gene perdido (perd, also known as kon-tiki, a transmembrane chondroitin proteoglycan) in myofibrillogenesis. Expression of perd RNAi in muscles, prior to adult myogenesis, can induce misorientation and detachment of Drosophila adult abdominal muscles. In comparison to controls, perd-depleted muscles contain fewer myofibrils, which are localized at the cell periphery. These myofibrils are detached from each other and display a defective sarcomeric structure. Our results demonstrate that the extracellular matrix receptor Perd has a specific role in the assembly of myofibrils and in sarcomeric organization. We suggest that Perd acts downstream or in parallel to integrins to enable the connection of nascent myofibrils to the Z-bands. Our work identifies the Drosophila adult abdominal muscles as a model to investigate in vivo the mechanisms behind myofibrillogenesis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/crecimiento & desarrollo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Proteoglicanos/metabolismo , Sarcómeros/fisiología , Animales , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de la Membrana/genética , Desarrollo de Músculos , Sarcómeros/metabolismo
9.
J Cell Sci ; 127(Pt 21): 4667-78, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25179603

RESUMEN

Coordinating exit from the cell cycle with differentiation is crucial for proper development and tissue homeostasis. Failure to do so can lead to aberrant organogenesis and tumorigenesis. However, little is known about the developmental signals that regulate the switch from cell cycle exit to differentiation. Signals downstream of two key developmental pathways, Notch and Salvador-Warts-Hippo (SWH), and signals downstream of myosin activity regulate this switch during the development of the follicle cell epithelium of the Drosophila ovary. Here, we have identified a fourth player, the integrin signaling pathway. Elimination of integrin function blocks the mitosis-to-endocycle switch and differentiation in posterior follicle cells (PFCs), by regulation of the cyclin-dependent kinase inhibitor (CKI) dacapo. In addition, integrin-mutant PFCs show defective Notch signaling and endocytosis. Furthermore, integrins act in PFCs by modulating the activity of the Notch pathway, as reducing the amount of Hairless, the major antagonist of Notch, or misexpressing Notch intracellular domain rescues the cell cycle and differentiation defects. Taken together, our findings reveal a direct involvement of integrin signaling on the spatial and temporal regulation of epithelial cell differentiation during development.


Asunto(s)
Proteínas de Drosophila/metabolismo , Células Epiteliales/metabolismo , Integrinas/metabolismo , Receptores Notch/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Drosophila , Proteínas de Drosophila/genética , Células Epiteliales/citología , Femenino , Inmunohistoquímica , Integrinas/genética , Masculino , Receptores Notch/genética
10.
J Cell Sci ; 126(Pt 10): 2285-93, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23525006

RESUMEN

Guided cell migration is a key mechanism for cell positioning in morphogenesis. The current model suggests that the spatially controlled activation of receptor tyrosine kinases (RTKs) by guidance cues limits Rac activity at the leading edge, which is crucial for establishing and maintaining polarized cell protrusions at the front. However, little is known about the mechanisms by which RTKs control the local activation of Rac. Here, using a multidisciplinary approach, we identify the GTP exchange factor (GEF) Vav as a key regulator of Rac activity downstream of RTKs in a developmentally regulated cell migration event, that of the Drosophila border cells (BCs). We show that elimination of the vav gene impairs BC migration. Live imaging analysis reveals that vav is required for the stabilization and maintenance of protrusions at the front of the BC cluster. In addition, activation of the PDGF/VEGF-related receptor (PVR) by its ligand the PDGF/PVF1 factor brings about activation of Vav protein by direct interaction with the intracellular domain of PVR. Finally, FRET analyses demonstrate that Vav is required in BCs for the asymmetric distribution of Rac activity at the front. Our results unravel an important role for the Vav proteins as signal transducers that couple signalling downstream of RTKs with local Rac activation during morphogenetic movements.


Asunto(s)
Drosophila melanogaster/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Animales , Animales Modificados Genéticamente , Movimiento Celular/genética , Extensiones de la Superficie Celular/genética , Células Cultivadas , Drosophila melanogaster/citología , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Represión Enzimática/genética , Femenino , Morfogénesis/genética , Proteínas Proto-Oncogénicas c-vav/genética , ARN Interferente Pequeño/genética , Eliminación de Secuencia/genética , Técnicas del Sistema de Dos Híbridos
11.
J Cell Sci ; 126(Pt 15): 3475-84, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23704353

RESUMEN

Throughout embryonic development, macrophages not only act as the first line of defence against infection but also help to sculpt organs and tissues of the embryo by removing dead cells and secreting extracellular matrix components. Key to their function is the ability of embryonic macrophages to migrate and disperse throughout the embryo. Despite these important developmental functions, little is known about the molecular mechanisms underlying embryonic macrophage migration in vivo. Integrins are key regulators of many of the adult macrophage responses, but their role in embryonic macrophages remains poorly characterized. Here, we have used Drosophila macrophages (haemocytes) as a model system to address the role of integrins during embryonic macrophage dispersal in vivo. We show that the main ßPS integrin, myospheroid, affects haemocyte migration in two ways; by shaping the three-dimensional environment in which haemocytes migrate and by regulating the migration of haemocytes themselves. Live imaging revealed a requirement for myospheroid within haemocytes to coordinate the microtubule and actin dynamics, and to enable haemocyte developmental dispersal, contact repulsion and inflammatory migration towards wounds.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/citología , Cadenas beta de Integrinas/metabolismo , Animales , Movimiento Celular/fisiología , Drosophila/embriología , Macrófagos/citología
12.
Cancers (Basel) ; 15(22)2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38001693

RESUMEN

Cancer is the second leading cause of death worldwide. Key to cancer initiation and progression is the crosstalk between cancer cells and their microenvironment. The extracellular matrix (ECM) is a major component of the tumour microenvironment and integrins, main cell-ECM adhesion receptors, are involved in every step of cancer progression. However, accumulating evidence has shown that integrins can act as tumour promoters but also as tumour suppressor factors, revealing that the biological roles of integrins in cancer are complex. This incites a better understating of integrin function in cancer progression. To achieve this goal, simple model organisms, such as Drosophila, offer great potential to unravel underlying conceptual principles. Here, we find that in the Drosophila wing disc epithelium the ßPS integrins act as suppressors of tumours induced by a gain of function of the oncogenic form of Ras, RasV12. We show that ßPS integrin depletion enhances the growth, delamination and invasive behaviour of RasV12 tumour cells, as well as their ability to affect the tumour microenvironment. These results strongly suggest that integrin function as tumour suppressors might be evolutionarily conserved. Drosophila can be used to understand the complex tumour modulating activities conferred by integrins, thus facilitating drug development.

13.
Genes (Basel) ; 14(7)2023 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-37510408

RESUMEN

Oncogenic mutations in the small GTPase Ras contribute to ~30% of human cancers. However, tissue growth induced by oncogenic Ras is restrained by the induction of cellular senescence, and additional mutations are required to induce tumor progression. Therefore, identifying cooperating cancer genes is of paramount importance. Recently, the tensin family of focal adhesion proteins, TNS1-4, have emerged as regulators of carcinogenesis, yet their role in cancer appears somewhat controversial. Around 90% of human cancers are of epithelial origin. We have used the Drosophila wing imaginal disc epithelium as a model system to gain insight into the roles of two orthologs of human TNS2 and 4, blistery (by) and PVRAP, in epithelial cancer progression. We have generated null mutations in PVRAP and found that, as is the case for by and mammalian tensins, PVRAP mutants are viable. We have also found that elimination of either PVRAP or by potentiates RasV12-mediated wing disc hyperplasia. Furthermore, our results have unraveled a mechanism by which tensins may limit Ras oncogenic capacity, the regulation of cell shape and growth. These results demonstrate that Drosophila tensins behave as suppressors of Ras-driven tissue hyperplasia, suggesting that the roles of tensins as modulators of cancer progression might be evolutionarily conserved.


Asunto(s)
Drosophila , Neoplasias , Animales , Humanos , Tensinas/genética , Hiperplasia , Neoplasias/patología , Carcinogénesis/genética , Mamíferos
14.
Front Cell Dev Biol ; 11: 1114458, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36926523

RESUMEN

Cell proliferation and differentiation show a remarkable inverse relationship. The temporal coupling between cell cycle withdrawal and differentiation of stem cells (SCs) is crucial for epithelial tissue growth, homeostasis and regeneration. Proliferation vs. differentiation SC decisions are often controlled by the surrounding microenvironment, of which the basement membrane (BM; a specialized form of extracellular matrix surrounding cells and tissues), is one of its main constituents. Years of research have shown that integrin-mediated SC-BM interactions regulate many aspects of SC biology, including the proliferation-to-differentiation switch. However, these studies have also demonstrated that the SC responses to interactions with the BM are extremely diverse and depend on the cell type and state and on the repertoire of BM components and integrins involved. Here, we show that eliminating integrins from the follicle stem cells (FSCs) of the Drosophila ovary and their undifferentiated progeny increases their proliferation capacity. This results in an excess of various differentiated follicle cell types, demonstrating that cell fate determination can occur in the absence of integrins. Because these phenotypes are similar to those found in ovaries with decreased laminin levels, our results point to a role for the integrin-mediated cell-BM interactions in the control of epithelial cell division and subsequent differentiation. Finally, we show that integrins regulate proliferation by restraining the activity of the Notch/Delta pathway during early oogenesis. Our work increases our knowledge of the effects of cell-BM interactions in different SC types and should help improve our understanding of the biology of SCs and exploit their therapeutic potential.

15.
Development ; 136(24): 4165-76, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19906841

RESUMEN

Laminins are heterotrimeric molecules found in all basement membranes. In mammals, they have been involved in diverse developmental processes, from gastrulation to tissue maintenance. The Drosophila genome encodes two laminin alpha chains, one beta and one Gamma, which form two distinct laminin trimers. So far, only mutations affecting one or other trimer have been analysed. In order to study embryonic development in the complete absence of laminins, we mutated the gene encoding the sole laminin beta chain in Drosophila, LanB1, so that no trimers can be made. We show that LanB1 mutant embryos develop until the end of embryogenesis. Electron microscopy analysis of mutant embryos reveals that the basement membranes are absent and the remaining extracellular material appears disorganised and diffuse. Accordingly, abnormal accumulation of major basement membrane components, such as Collagen IV and Perlecan, is observed in mutant tissues. In addition, we show that elimination of LanB1 prevents the normal morphogenesis of most organs and tissues, including the gut, trachea, muscles and nervous system. In spite of the above structural roles for laminins, our results unravel novel functions in cell adhesion, migration and rearrangement. We propose that while an early function of laminins in gastrulation is not conserved in Drosophila and mammals, their function in basement membrane assembly and organogenesis seems to be maintained throughout evolution.


Asunto(s)
Membrana Basal/fisiología , Proteínas de Drosophila/fisiología , Drosophila/embriología , Embrión no Mamífero/fisiología , Laminina/fisiología , Animales , Membrana Basal/embriología , Adhesión Celular , Movimiento Celular , Colágeno Tipo IV/metabolismo , Drosophila/fisiología , Proteoglicanos de Heparán Sulfato/metabolismo , Morfogénesis/genética , Mutación , Especificidad de Órganos
16.
EMBO Rep ; 11(12): 943-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21102643

RESUMEN

Stationary-to-migratory transitions of epithelial cells have a key role in development and tumour progression. Border cell migration is a powerful system in which to investigate this transition in living organisms. Here, we identify the Ste20-like kinase misshapen (msn) as a novel regulator of border-cell migration in Drosophila. Expression of msn in border cells is independent of the transcription factor slow border cells and of inputs from all pathways that are known to control border-cell migration. The msn gene functions to modulate the levels and/or distribution of Drosophila E-cadherin to promote the invasive migratory behaviour of border cells.


Asunto(s)
Movimiento Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/enzimología , Células Epiteliales/citología , Células Epiteliales/enzimología , Ovario/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Cadherinas/genética , Femenino , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Transducción de Señal , Transcripción Genética , Proteínas de Unión al GTP rab/metabolismo
17.
Cell Rep ; 39(4): 110734, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35476979

RESUMEN

Biological tubes are fundamental units of most metazoan organs. Their defective morphogenesis can cause malformations and pathologies. An integral component of biological tubes is the extracellular matrix, present apically (aECM) and basally (BM). Studies using the Drosophila tracheal system established an essential function for the aECM in tubulogenesis. Here, we demonstrate that the BM also plays a critical role in this process. We find that BM components are deposited in a spatial-temporal manner in the trachea. We show that laminins, core BM components, control size and shape of tracheal tubes and their topology within the embryo. At a cellular level, laminins control cell shape changes and distribution of the cortical cytoskeleton component α-spectrin. Finally, we report that the BM and aECM act independently-yet cooperatively-to control tube elongation and together to guarantee tissue integrity. Our results unravel key roles for the BM in shaping, positioning, and maintaining biological tubes.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Membrana Basal , Matriz Extracelular , Laminina , Morfogénesis
18.
Front Cell Dev Biol ; 10: 892691, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35769262

RESUMEN

Adhesion to the extracellular matrix (ECM) is required for normal epithelial cell survival. Disruption of this interaction leads to a specific type of apoptosis known as anoikis. Yet, there are physiological and pathological situations in which cells not connected to the ECM are protected from anoikis, such as during cell migration or metastasis. The main receptors transmitting signals from the ECM are members of the integrin family. However, although integrin-mediated cell-ECM anchorage has been long recognized as crucial for epithelial cell survival, the in vivo significance of this interaction remains to be weighed. In this work, we have used the Drosophila wing imaginal disc epithelium to analyze the importance of integrins as survival factors during epithelia morphogenesis. We show that reducing integrin expression in the wing disc induces caspase-dependent cell death and basal extrusion of the dead cells. In this case, anoikis is mediated by the activation of the JNK pathway, which in turn triggers expression of the proapoptotic protein Hid. In addition, our results strongly suggest that, during wing disc morphogenesis, the EGFR pathway protects cells undergoing cell shape changes upon ECM detachment from anoikis. Furthermore, we show that oncogenic activation of the EGFR/Ras pathway in integrin mutant cells rescues them from apoptosis while promoting their extrusion from the epithelium. Altogether, our results support the idea that integrins promote cell survival during normal tissue morphogenesis and prevent the extrusion of transformed cells.

19.
Curr Biol ; 17(8): 683-8, 2007 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-17363255

RESUMEN

Epithelia act as important physiological barriers and as structural components of tissues and organs. In the Drosophila ovary, follicle cells envelop the germline cysts to form a monolayer epithelium. During division, the orientation of the mitotic spindle in follicle cells is such that both daughter cells remain within the same plane, and the simple structure of the follicular epithelium is thus preserved. Here we show that integrins, heterodimeric transmembrane receptors that connect the extracellular matrix to the cell's cytoskeleton [1, 2], are required for maintaining the ovarian monolayer epithelium in Drosophila. Mosaic egg chambers containing integrin mutant follicle cells develop stratified epithelia at both poles. This stratification is due neither to abnormal cell proliferation nor to defects in the apical-basal polarity of the mutant cells. Instead, integrin function is required for the correct orientation of the mitotic apparatus both in mutant cells and in their immediately adjacent wild-type neighbors. We further demonstrate that integrin-mediated signaling, rather than adhesion, is sufficient for maintaining the integrity of the follicular epithelium. The above data show that integrins are necessary for preserving the simple organization of a specialized epithelium and link integrin-mediated signaling to the correct orientation of the mitotic spindle in this epithelial cell type.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Integrinas/metabolismo , Huso Acromático/metabolismo , Animales , Drosophila/citología , Epitelio/metabolismo , Femenino , Folículo Ovárico/citología , Ovario/citología , Ovario/metabolismo , Transducción de Señal
20.
Int J Dev Biol ; 52(7): 925-32, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18956322

RESUMEN

The generation of epithelial cell polarity is a key process during development. Although the induction and orientation of cell polarity by cell-cell and cell-extracellular matrix (ECM) interactions is well established, the molecular mechanisms by which signals from the ECM control cell polarity in developing epithelial tissues remain poorly understood. Here, we have used the follicular epithelium of the Drosophila ovary to investigate the role that integrins, the main cell-ECM receptors, play in the establishment of apicobasal polarity. Mature follicle cells have an apical side facing the germ line and a basal side in contact with a basement membrane. Our results show that integrins - presumably via interactions with the basement membrane - play a reinforcing role in follicle cell polarization, as they are required to establish and/or maintain follicle cell membrane asymmetry only when contact with the germ line is prevented. We suggest that the primary cue for polarization of the follicular epithelium is contact with the germline cells. In addition, while interfering with apical and lateral polarization cues leads to apoptosis, we show here that inhibition of contact with the basement membrane mediated by integrins does not affect cell survival. Finally, we provide evidence to suggest that integrins are required to orientate epithelial polarity in vivo.


Asunto(s)
Polaridad Celular/fisiología , Drosophila/fisiología , Células Epiteliales/fisiología , Integrinas/fisiología , Folículo Ovárico/fisiología , Animales , Drosophila/genética , Células Epiteliales/metabolismo , Femenino , Inmunohistoquímica , Modelos Biológicos , Folículo Ovárico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA