Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Biol ; 34(7): 1390-1402.e4, 2024 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-38428416

RESUMO

Collective cell migration is integral to many developmental and disease processes. Previously, we discovered that protein phosphatase 1 (Pp1) promotes border cell collective migration in the Drosophila ovary. We now report that the Pp1 phosphatase regulatory subunit dPPP1R15 is a critical regulator of border cell migration. dPPP1R15 is an ortholog of mammalian PPP1R15 proteins that attenuate the endoplasmic reticulum (ER) stress response. We show that, in collectively migrating border cells, dPPP1R15 phosphatase restrains an active physiological protein kinase R-like ER kinase- (PERK)-eIF2α-activating transcription factor 4 (ATF4) stress pathway. RNAi knockdown of dPPP1R15 blocks border cell delamination from the epithelium and subsequent migration, increases eIF2α phosphorylation, reduces translation, and drives expression of the stress response transcription factor ATF4. We observe similar defects upon overexpression of ATF4 or the eIF2α kinase PERK. Furthermore, we show that normal border cells express markers of the PERK-dependent ER stress response and require PERK and ATF4 for efficient migration. In many other cell types, unresolved ER stress induces initiation of apoptosis. In contrast, border cells with chronic RNAi knockdown of dPPP1R15 survive. Together, our results demonstrate that the PERK-eIF2α-ATF4 pathway, regulated by dPPP1R15 activity, counteracts the physiological ER stress that occurs during collective border cell migration. We propose that in vivo collective cell migration is intrinsically "stressful," requiring tight homeostatic control of the ER stress response for collective cell cohesion, dynamics, and movement.


Assuntos
Transdução de Sinais , eIF-2 Quinase , Animais , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Apoptose , Movimento Celular , Monoéster Fosfórico Hidrolases/metabolismo , Fator de Iniciação 2 em Eucariotos/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Mamíferos
2.
BMC Genomics ; 24(1): 728, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38041052

RESUMO

BACKGROUND: Collective cell migration underlies many essential processes, including sculpting organs during embryogenesis, wound healing in the adult, and metastasis of cancer cells. At mid-oogenesis, Drosophila border cells undergo collective migration. Border cells round up into a small group at the pre-migration stage, detach from the epithelium and undergo a dynamic and highly regulated migration at the mid-migration stage, and stop at the oocyte, their final destination, at the post-migration stage. While specific genes that promote cell signaling, polarization of the cluster, formation of protrusions, and cell-cell adhesion are known to regulate border cell migration, there may be additional genes that promote these distinct active phases of border cell migration. Therefore, we sought to identify genes whose expression patterns changed during border cell migration. RESULTS: We performed RNA-sequencing on border cells isolated at pre-, mid-, and post-migration stages. We report that 1,729 transcripts, in nine co-expression gene clusters, are temporally and differentially expressed across the three migration stages. Gene ontology analyses and constructed protein-protein interaction networks identified genes expected to function in collective migration, such as regulators of the cytoskeleton, adhesion, and tissue morphogenesis, but also uncovered a notable enrichment of genes involved in immune signaling, ribosome biogenesis, and stress responses. Finally, we validated the in vivo expression and function of a subset of identified genes in border cells. CONCLUSIONS: Overall, our results identified differentially and temporally expressed genetic networks that may facilitate the efficient development and migration of border cells. The genes identified here represent a wealth of new candidates to investigate the molecular nature of dynamic collective cell migrations in developing tissues.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Redes Reguladoras de Genes , Oogênese/genética , Movimento Celular/genética , Perfilação da Expressão Gênica , Drosophila melanogaster/genética
3.
Dev Biol ; 501: 1-19, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37269969

RESUMO

Having intact epithelial tissues is critical for embryonic development and adult homeostasis. How epithelia respond to damaging insults or tissue growth while still maintaining intercellular connections and barrier integrity during development is poorly understood. The conserved small GTPase Rap1 is critical for establishing cell polarity and regulating cadherin-catenin cell junctions. Here, we identified a new role for Rap1 in maintaining epithelial integrity and tissue shape during Drosophila oogenesis. Loss of Rap1 activity disrupted the follicle cell epithelium and the shape of egg chambers during a period of major growth. Rap1 was required for proper E-Cadherin localization in the anterior epithelium and for epithelial cell survival. Both Myo-II and the adherens junction-cytoskeletal linker protein α-Catenin were required for normal egg chamber shape but did not strongly affect cell viability. Blocking the apoptotic cascade failed to rescue the cell shape defects caused by Rap1 inhibition. One consequence of increased cell death caused by Rap1 inhibition was the loss of polar cells and other follicle cells, which later in development led to fewer cells forming a migrating border cell cluster. Our results thus indicate dual roles for Rap1 in maintaining epithelia and cell survival in a growing tissue during development.


Assuntos
Proteínas de Drosophila , Animais , Caderinas/metabolismo , Sobrevivência Celular , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Epitélio/metabolismo
4.
Biochem Soc Trans ; 51(4): 1495-1504, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37387360

RESUMO

Migrating cell collectives navigate complex tissue environments both during normal development and in pathological contexts such as tumor invasion and metastasis. To do this, cells in collectives must stay together but also communicate information across the group. The cadherin superfamily of proteins mediates junctional adhesions between cells, but also serve many essential functions in collective cell migration. Besides keeping migrating cell collectives cohesive, cadherins help follower cells maintain their attachment to leader cells, transfer information about front-rear polarity among the cohort, sense and respond to changes in the tissue environment, and promote intracellular signaling, in addition to other cellular behaviors. In this review, we highlight recent studies that reveal diverse but critical roles for both classical and atypical cadherins in collective cell migration, specifically focusing on four in vivo model systems in development: the Drosophila border cells, zebrafish mesendodermal cells, Drosophila follicle rotation, and Xenopus neural crest cells.


Assuntos
Caderinas , Peixe-Zebra , Animais , Caderinas/metabolismo , Peixe-Zebra/metabolismo , Transdução de Sinais , Movimento Celular/fisiologia , Drosophila/metabolismo , Adesão Celular
5.
Curr Biol ; 33(13): 2587-2601.e5, 2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37244252

RESUMO

Collective cell movements contribute to tissue development and repair and spread metastatic disease. In epithelia, cohesive cell movements require reorganization of adherens junctions and the actomyosin cytoskeleton. However, the mechanisms that coordinate cell-cell adhesion and cytoskeletal remodeling during collective cell migration in vivo are unclear. We investigated the mechanisms of collective cell migration during epidermal wound healing in Drosophila embryos. Upon wounding, the cells adjacent to the wound internalize cell-cell adhesion molecules and polarize actin and the motor protein non-muscle myosin II to form a supracellular cable around the wound that coordinates cell movements. The cable anchors at former tricellular junctions (TCJs) along the wound edge, and TCJs are reinforced during wound closure. We found that the small GTPase Rap1 was necessary and sufficient for rapid wound repair. Rap1 promoted myosin polarization to the wound edge and E-cadherin accumulation at TCJs. Using embryos expressing a mutant form of the Rap1 effector Canoe/Afadin that cannot bind Rap1, we found that Rap1 signals through Canoe for adherens junction remodeling, but not for actomyosin cable assembly. Instead, Rap1 was necessary and sufficient for RhoA/Rho1 activation at the wound edge. The RhoGEF Ephexin localized to the wound edge in a Rap1-dependent manner, and Ephexin was necessary for myosin polarization and rapid wound repair, but not for E-cadherin redistribution. Together, our data show that Rap1 coordinates the molecular rearrangements that drive embryonic wound healing, promoting actomyosin cable assembly through Ephexin-Rho1, and E-cadherin redistribution through Canoe, thus enabling rapid collective cell migration in vivo.


Assuntos
Actomiosina , Proteínas de Drosophila , Animais , Actomiosina/metabolismo , Adesão Celular , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Movimento Celular/fisiologia , Miosinas/metabolismo , Junções Aderentes/metabolismo , Caderinas/genética , Caderinas/metabolismo
6.
Methods Mol Biol ; 2626: 193-217, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36715906

RESUMO

Drosophila border cells have emerged as a genetically tractable model to investigate dynamic collective cell migration within the context of a developing organ. Studies of live border cell cluster migration have revealed similarities with other migrating collectives, including formation and restriction of cellular protrusions to the front of the cluster, supracellular actomyosin contractility of the entire collective, and intra-collective cell motility. Here, we describe protocols to prepare ex vivo cultures of stage 9 egg chambers followed by live time-lapse imaging of fluorescently labeled border cells to image dynamic cell behaviors. We provide options to perform live imaging using either a widefield epifluorescent microscope or a confocal microscope. We further outline steps to quantify various cellular behaviors and protein dynamics of live migrating border cells using the Fiji image processing package of ImageJ. These methods can be adapted to other migrating cell collectives in cultured tissues and organs.


Assuntos
Proteínas de Drosophila , Oogênese , Animais , Movimento Celular , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Actomiosina/metabolismo
7.
G3 (Bethesda) ; 12(1)2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34849760

RESUMO

Migrating cell collectives are key to embryonic development but also contribute to invasion and metastasis of a variety of cancers. Cell collectives can invade deep into tissues, leading to tumor progression and resistance to therapies. Collective cell invasion is also observed in the lethal brain tumor glioblastoma (GBM), which infiltrates the surrounding brain parenchyma leading to tumor growth and poor patient outcomes. Drosophila border cells, which migrate as a small cell cluster in the developing ovary, are a well-studied and genetically accessible model used to identify general mechanisms that control collective cell migration within native tissue environments. Most cell collectives remain cohesive through a variety of cell-cell adhesion proteins during their migration through tissues and organs. In this study, we first identified cell adhesion, cell matrix, cell junction, and associated regulatory genes that are expressed in human brain tumors. We performed RNAi knockdown of the Drosophila orthologs in border cells to evaluate if migration and/or cohesion of the cluster was impaired. From this screen, we identified eight adhesion-related genes that disrupted border cell collective migration upon RNAi knockdown. Bioinformatics analyses further demonstrated that subsets of the orthologous genes were elevated in the margin and invasive edge of human GBM patient tumors. These data together show that conserved cell adhesion and adhesion regulatory proteins with potential roles in tumor invasion also modulate collective cell migration. This dual screening approach for adhesion genes linked to GBM and border cell migration thus may reveal conserved mechanisms that drive collective tumor cell invasion.


Assuntos
Proteínas de Drosophila , Glioblastoma , Animais , Adesão Celular/genética , Movimento Celular/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Feminino , Glioblastoma/metabolismo , Humanos , Interferência de RNA
8.
Elife ; 92020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32369438

RESUMO

Collective cell migration is central to many developmental and pathological processes. However, the mechanisms that keep cell collectives together and coordinate movement of multiple cells are poorly understood. Using the Drosophila border cell migration model, we find that Protein phosphatase 1 (Pp1) activity controls collective cell cohesion and migration. Inhibition of Pp1 causes border cells to round up, dissociate, and move as single cells with altered motility. We present evidence that Pp1 promotes proper levels of cadherin-catenin complex proteins at cell-cell junctions within the cluster to keep border cells together. Pp1 further restricts actomyosin contractility to the cluster periphery rather than at individual internal border cell contacts. We show that the myosin phosphatase Pp1 complex, which inhibits non-muscle myosin-II (Myo-II) activity, coordinates border cell shape and cluster cohesion. Given the high conservation of Pp1 complexes, this study identifies Pp1 as a major regulator of collective versus single cell migration.


Assuntos
Movimento Celular/fisiologia , Proteínas de Drosophila/fisiologia , Proteína Fosfatase 1/fisiologia , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Feminino , Genes/genética , Genes/fisiologia , Masculino , Proteína Fosfatase 1/genética , Proteína Fosfatase 1/metabolismo
9.
Elife ; 92020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32463359

RESUMO

The origins of the posterior lobe, a recently evolved structure in some species of Drosophila, have become clearer.


Assuntos
Evolução Biológica , Drosophila melanogaster , Animais , Drosophila , Matriz Extracelular
10.
Cell Death Dis ; 11(2): 152, 2020 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-32102991

RESUMO

Glioblastoma (GBM) is the most prevalent primary malignant brain tumor and is associated with extensive tumor cell infiltration into the adjacent brain parenchyma. However, there are limited targeted therapies that address this disease hallmark. While the invasive capacity of self-renewing cancer stem cells (CSCs) and their non-CSC progeny has been investigated, the mode(s) of migration used by CSCs during invasion is currently unknown. Here we used time-lapse microscopy to evaluate the migratory behavior of CSCs, with a focus on identifying key regulators of migration. A head-to-head migration assay demonstrated that CSCs are more invasive than non-CSCs. Time-lapse live cell imaging further revealed that GBM patient-derived CSC models either migrate in a collective manner or in a single cell fashion. To uncover conserved molecular regulators responsible for collective cell invasion, we utilized the genetically tractable Drosophila border cell collective migration model. Candidates for functional studies were generated using results from a targeted Drosophila genetic screen followed by gene expression analysis of the human homologs in GBM tumors and associated GBM patient prognosis. This strategy identified the highly conserved small GTPase, Rap1a, as a potential regulator of cell invasion. Alteration of Rap1a activity impaired the forward progress of Drosophila border cells during development. Rap1a expression was elevated in GBM and associated with higher tumor grade. Functionally, the levels of activated Rap1a impacted CSC migration speed out of spheres onto extracellular matrix. The data presented here demonstrate that CSCs are more invasive than non-CSCs, are capable of both collective and single cell migration, and express conserved genes that are required for migration and invasion. Using this integrated approach, we identified a new role for Rap1a in the migration of GBM CSCs.


Assuntos
Neoplasias Encefálicas/metabolismo , Movimento Celular/fisiologia , Glioblastoma/patologia , Células-Tronco Neoplásicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/fisiologia , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/diagnóstico , Glioblastoma/metabolismo , Humanos , Células-Tronco Neoplásicas/patologia , Prognóstico
11.
Mol Biol Cell ; 29(22): 2656-2673, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30156466

RESUMO

During development and in cancer, cells often move together in small to large collectives. To move as a unit, cells within collectives need to stay coupled together and coordinate their motility. How cell collectives remain interconnected and migratory, especially when moving through in vivo environments, is not well understood. The genetically tractable border cell group undergoes a highly polarized and cohesive cluster-type migration in the Drosophila ovary. Here we report that the small GTPase Rap1, through activation by PDZ-GEF, regulates border cell collective migration. We find that Rap1 maintains cell contacts within the cluster, at least in part by promoting the organized distribution of E-cadherin at specific cell-cell junctions. Rap1 also restricts migratory protrusions to the front of the border cell cluster and promotes the extension of protrusions with normal dynamics. Further, Rap1 is required in the outer migratory border cells but not in the central nonmigratory polar cells. Such cell specificity correlates well with the spatial distribution of the inhibitory Rapgap1 protein, which is higher in polar cells than in border cells. We propose that precisely regulated Rap1 activity reinforces connections between cells and polarizes the cluster, thus facilitating the coordinated collective migration of border cells.


Assuntos
Movimento Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/enzimologia , Proteínas de Ligação a Telômeros/metabolismo , Animais , Caderinas/metabolismo , Extensões da Superfície Celular/metabolismo , Feminino , Proteínas Ativadoras de GTPase , Complexo Shelterina
12.
Nat Commun ; 9(1): 1210, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29572440

RESUMO

The actomyosin cytoskeleton, a key stress-producing unit in epithelial cells, oscillates spontaneously in a wide variety of systems. Although much of the signal cascade regulating myosin activity has been characterized, the origin of such oscillatory behavior is still unclear. Here, we show that basal myosin II oscillation in Drosophila ovarian epithelium is not controlled by actomyosin cortical tension, but instead relies on a biochemical oscillator involving ROCK and myosin phosphatase. Key to this oscillation is a diffusive ROCK flow, linking junctional Rho1 to medial actomyosin cortex, and dynamically maintained by a self-activation loop reliant on ROCK kinase activity. In response to the resulting myosin II recruitment, myosin phosphatase is locally enriched and shuts off ROCK and myosin II signals. Coupling Drosophila genetics, live imaging, modeling, and optogenetics, we uncover an intrinsic biochemical oscillator at the core of myosin II regulatory network, shedding light on the spatio-temporal dynamics of force generation.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Miosina Tipo II/química , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Actomiosina/química , Animais , Animais Geneticamente Modificados , Drosophila/genética , Feminino , Transferência Ressonante de Energia de Fluorescência , Luz , Masculino , Microscopia Confocal , Optogenética , Oscilometria , Transdução de Sinais , Quinases Associadas a rho
13.
F1000Res ; 52016.
Artigo em Inglês | MEDLINE | ID: mdl-27303647

RESUMO

Tissues in developing embryos exhibit complex and dynamic rearrangements that shape forming organs, limbs, and body axes. Directed migration, mediolateral intercalation, lumen formation, and other rearrangements influence the topology and topography of developing tissues. These collective cell behaviors are distinct phenomena but all involve the fine-grained control of cell polarity. Here we review recent findings in the dynamics of polarized cell behavior in both the Drosophila ovarian border cells and the Ciona notochord. These studies reveal the remarkable reorganization of cell polarity during organ formation and underscore conserved mechanisms of developmental cell polarity including the Par/atypical protein kinase C (aPKC) and planar cell polarity pathways. These two very different model systems demonstrate important commonalities but also key differences in how cell polarity is controlled in tissue morphogenesis. Together, these systems raise important, broader questions on how the developmental control of cell polarity contributes to morphogenesis of diverse tissues across the metazoa.

14.
Development ; 143(15): 2791-802, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27317808

RESUMO

The pattern of the Drosophila melanogaster adult wing is heavily influenced by the expression of proteins that dictate cell fate decisions between intervein and vein during development. dSRF (Blistered) expression in specific regions of the larval wing disc promotes intervein cell fate, whereas EGFR activity promotes vein cell fate. Here, we report that the chromatin-organizing protein CAP-D3 acts to dampen dSRF levels at the anterior/posterior boundary in the larval wing disc, promoting differentiation of cells into the anterior crossvein. CAP-D3 represses KNOT expression in cells immediately adjacent to the anterior/posterior boundary, thus blocking KNOT-mediated repression of EGFR activity and preventing cell death. Maintenance of EGFR activity in these cells depresses dSRF levels in the neighboring anterior crossvein progenitor cells, allowing them to differentiate into vein cells. These findings uncover a novel transcriptional regulatory network influencing Drosophila wing vein development, and are the first to identify a Condensin II subunit as an important regulator of EGFR activity and cell fate determination in vivo.


Assuntos
Cromossomos/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Proteínas de Ciclo Celular , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Imunoprecipitação da Cromatina , Cromossomos/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Drosophila , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hibridização In Situ , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
15.
Mol Biol Cell ; 27(12): 1898-910, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27122602

RESUMO

Migrating cells need to overcome physical constraints from the local microenvironment to navigate their way through tissues. Cells that move collectively have the additional challenge of negotiating complex environments in vivo while maintaining cohesion of the group as a whole. The mechanisms by which collectives maintain a migratory morphology while resisting physical constraints from the surrounding tissue are poorly understood. Drosophila border cells represent a genetic model of collective migration within a cell-dense tissue. Border cells move as a cohesive group of 6-10 cells, traversing a network of large germ line-derived nurse cells within the ovary. Here we show that the border cell cluster is compact and round throughout their entire migration, a shape that is maintained despite the mechanical pressure imposed by the surrounding nurse cells. Nonmuscle myosin II (Myo-II) activity at the cluster periphery becomes elevated in response to increased constriction by nurse cells. Furthermore, the distinctive border cell collective morphology requires highly dynamic and localized enrichment of Myo-II. Thus, activated Myo-II promotes cortical tension at the outer edge of the migrating border cell cluster to resist compressive forces from nurse cells. We propose that dynamic actomyosin tension at the periphery of collectives facilitates their movement through restrictive tissues.


Assuntos
Miosina Tipo II/metabolismo , Miosinas/fisiologia , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Animais , Movimento Celular/fisiologia , Forma Celular/fisiologia , Microambiente Celular , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Oogênese , Transdução de Sinais/genética
16.
Int Rev Cell Mol Biol ; 312: 169-99, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25262242

RESUMO

The partitioning defective gene 1 (Par-1)/microtubule affinity-regulating kinase (MARK) family of serine-threonine kinases have diverse cellular roles. Primary among these roles are the establishment and maintenance of cell polarity and the promotion of microtubule dynamics. Par-1/MARK kinases also regulate a growing number of cellular functions via noncanonical protein targets. Recent studies have demonstrated that Par-1/MARK proteins are required for the migration of multiple cell types. This review outlines the current evidence for regulation of cell migration by Par-1/MARK through both canonical and noncanonical roles. Par-1/MARK canonical control of microtubules during nonneuronal and neuronal migration is described. Next, regulation of cell polarity by Par-1/MARK and its dynamic effect on the movement of migrating cells are discussed. As examples of recent research that have expanded, the roles of the Par-1/MARK in cell migration, noncanonical functions of Par-1/MARK in Wnt signaling and actomyosin dynamics are described. This review also highlights questions and current challenges to further understanding how the versatile Par-1/MARK proteins function in cell migration during development, homeostatic processes, and cancer.


Assuntos
Movimento Celular/fisiologia , Polaridade Celular/fisiologia , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Humanos , Microtúbulos/genética , Microtúbulos/metabolismo , Neurônios/citologia , Proteínas Serina-Treonina Quinases/genética
17.
Dev Dyn ; 242(5): 414-31, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23335293

RESUMO

BACKGROUND: Cell motility is essential for embryonic development and physiological processes such as the immune response, but also contributes to pathological conditions such as tumor progression and inflammation. However, our understanding of the mechanisms underlying migratory processes is incomplete. Drosophila border cells provide a powerful genetic model to identify the roles of genes that contribute to cell migration. RESULTS: Members of the Hedgehog signaling pathway were uncovered in two independent screens for interactions with the small GTPase Rac and the polarity protein Par-1 in border cell migration. Consistent with a role in migration, multiple Hh signaling components were enriched in the migratory border cells. Interference with Hh signaling by several different methods resulted in incomplete cell migration. Moreover, the polarized distribution of E-Cadherin and a marker of tyrosine kinase activity were altered when Hh signaling was disrupted. Conservation of Hh-Rac and Hh-Par-1 signaling was illustrated in the wing, in which Hh-dependent phenotypes were enhanced by loss of Rac or par-1. CONCLUSIONS: We identified a pathway by which Hh signaling connects to Rac and Par-1 in cell migration. These results further highlight the importance of modifier screens in the identification of new genes that function in developmental pathways.


Assuntos
Movimento Celular/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/embriologia , Perfilação da Expressão Gênica , Proteínas Hedgehog/fisiologia , Ovário/citologia , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiologia , Epistasia Genética/fisiologia , Feminino , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Masculino , Morfogênese/genética , Morfogênese/fisiologia , Oogênese/genética , Oogênese/fisiologia , Ovário/embriologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
18.
F1000Res ; 2: 220, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24715958

RESUMO

The dsRNA binding protein (dsRBP) PACT was first described as an activator of the dsRNA dependent protein kinase PKR in response to stress signals.  Additionally, it has been identified as a component of the small RNA processing pathway.  A role for PACT in this pathway represents an important interplay between two modes of post-transcriptional gene regulation.  The function of PACT in this context is poorly understood.  Thus, additional approaches are required to clarify the mechanism by which PACT functions.  In this study, the genetic utility of  Drosophila melanogaster was employed to identify dsRNA-binding proteins that are functionally orthologous to PACT.  Transgenic  Drosophila expressing human PACT were generated to determine whether PACT is capable of functionally substituting for the  Drosophila dsRBP R2D2, which has a well-defined role in small RNA biogenesis.  Results presented here indicate that PACT is unable to substitute for R2D2 at the whole organism level.

19.
G3 (Bethesda) ; 2(11): 1379-91, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23173089

RESUMO

Cells often move as collective groups during normal embryonic development and wound healing, although the mechanisms governing this type of migration are poorly understood. The Drosophila melanogaster border cells migrate as a cluster during late oogenesis and serve as a powerful in vivo genetic model for collective cell migration. To discover new genes that participate in border cell migration, 64 out of 66 genes that encode PDZ domain-containing proteins were systematically targeted by in vivo RNAi knockdown. The PDZ domain is one of the largest families of protein-protein interaction domains found in eukaryotes. Proteins that contain PDZ domains participate in a variety of biological processes, including signal transduction and establishment of epithelial apical-basal polarity. Targeting PDZ proteins effectively assesses a larger number of genes via the protein complexes and pathways through which these proteins function. par-6, a known regulator of border cell migration, was a positive hit and thus validated the approach. Knockdown of 14 PDZ domain genes disrupted migration with multiple RNAi lines. The candidate genes have diverse predicted cellular functions and are anticipated to provide new insights into the mechanisms that control border cell movement. As a test of this concept, two genes that disrupted migration were characterized in more detail: big bang and the Dlg5 homolog CG6509. We present evidence that Big bang regulates JAK/STAT signaling, whereas Dlg5/CG6509 maintains cluster cohesion. Moreover, these results demonstrate that targeting a selected class of genes by RNAi can uncover novel regulators of collective cell migration.


Assuntos
Movimento Celular/genética , Proteínas de Drosophila/genética , Drosophila/genética , Genes de Insetos , Domínios PDZ , Animais , Drosophila/embriologia , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Janus Quinases/metabolismo , RNA Interferente Pequeno , Transdução de Sinais
20.
Curr Biol ; 22(5): 363-72, 2012 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-22326025

RESUMO

BACKGROUND: Localized actomyosin contraction couples with actin polymerization and cell-matrix adhesion to regulate cell protrusions and retract trailing edges of migrating cells. Although many cells migrate in collective groups during tissue morphogenesis, mechanisms that coordinate actomyosin dynamics in collective cell migration are poorly understood. Migration of Drosophila border cells, a genetically tractable model for collective cell migration, requires nonmuscle myosin-II (Myo-II). How Myo-II specifically controls border cell migration and how Myo-II is itself regulated is largely unknown. RESULTS: We show that Myo-II regulates two essential features of border cell migration: (1) initial detachment of the border cell cluster from the follicular epithelium and (2) the dynamics of cellular protrusions. We further demonstrate that the cell polarity protein Par-1 (MARK), a serine-threonine kinase, regulates the localization and activation of Myo-II in border cells. Par-1 binds to myosin phosphatase and phosphorylates it at a known inactivating site. Par-1 thus promotes phosphorylated myosin regulatory light chain, thereby increasing Myo-II activity. Furthermore, Par-1 localizes to and increases active Myo-II at the cluster rear to promote detachment; in the absence of Par-1, spatially distinct active Myo-II is lost. CONCLUSIONS: We identify a critical new role for Par-1 kinase: spatiotemporal regulation of Myo-II activity within the border cell cluster through localized inhibition of myosin phosphatase. Polarity proteins such as Par-1, which intrinsically localize, can thus directly modulate the actomyosin dynamics required for border cell detachment and migration. Such a link between polarity proteins and cytoskeletal dynamics may also occur in other collective cell migrations.


Assuntos
Movimento Celular/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Miosina Tipo II/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Actomiosina/metabolismo , Animais , Polaridade Celular/fisiologia , Citoesqueleto/metabolismo , Proteínas de Drosophila/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Cadeias Leves de Miosina/metabolismo , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...