Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Nurses Prof Dev ; 36(5): 271-276, 2020.
Article in English | MEDLINE | ID: mdl-32890181

ABSTRACT

In the financial flux of the healthcare industry, resources for education and onboarding are ever dwindling. Nursing professional development specialists are frequently tasked with validating knowledge and skill application in a creative way. The article discusses the use of an escape room format to rapidly validate new graduate nurses' knowledge and skills, as well as decrease the number of education days for the organization.


Subject(s)
Clinical Competence/standards , Inservice Training , Nurses/statistics & numerical data , Problem-Based Learning , Thinking , Education, Nursing, Baccalaureate , Humans , Social Theory
2.
Neural Dev ; 15(1): 4, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32183907

ABSTRACT

BACKGROUND: Recent studies of synapse form and function highlight the importance of the actin cytoskeleton in regulating multiple aspects of morphogenesis, neurotransmission, and neural plasticity. The conserved actin-associated protein Enabled (Ena) is known to regulate development of the Drosophila larval neuromuscular junction through a postsynaptic mechanism. However, the functions and regulation of Ena within the presynaptic terminal has not been determined. METHODS: Here, we use a conditional genetic approach to address a presynaptic role for Ena on presynaptic morphology and ultrastructure, and also examine the pathway in which Ena functions through epistasis experiments. RESULTS: We find that Ena is required to promote the morphogenesis of presynaptic boutons and branches, in contrast to its inhibitory role in muscle. Moreover, while postsynaptic Ena is regulated by microRNA-mediated mechanisms, presynaptic Ena relays the output of the highly conserved receptor protein tyrosine phosphatase Dlar and associated proteins including the heparan sulfate proteoglycan Syndecan, and the non-receptor Abelson tyrosine kinase to regulate addition of presynaptic varicosities. Interestingly, Ena also influences active zones, where it restricts active zone size, regulates the recruitment of synaptic vesicles, and controls the amplitude and frequency of spontaneous glutamate release. CONCLUSION: We thus show that Ena, under control of the Dlar pathway, is required for presynaptic terminal morphogenesis and bouton addition and that Ena has active zone and neurotransmission phenotypes. Notably, in contrast to Dlar, Ena appears to integrate multiple pathways that regulate synapse form and function.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins/metabolism , Epistasis, Genetic/physiology , Morphogenesis/physiology , Receptor-Like Protein Tyrosine Phosphatases/metabolism , Signal Transduction/physiology , Synapses/physiology , Animals , DNA-Binding Proteins/genetics , Drosophila , Epistasis, Genetic/genetics , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Signal Transduction/genetics , Synapses/ultrastructure
3.
Fly (Austin) ; 6(4): 213-27, 2012.
Article in English | MEDLINE | ID: mdl-22940759

ABSTRACT

As tissues and organs are formed, they acquire a specific shape that plays an integral role in their ability to function properly. A relatively simple system that has been used to examine how tissues and organs are shaped is the formation of an elongated Drosophila egg. While it has been known for some time that Drosophila egg elongation requires interactions between a polarized intracellular basal actin network and a polarized extracellular network of basal lamina proteins, how these interactions contribute to egg elongation remained unclear. Recent studies using live imaging have revealed two novel processes, global tissue rotation and oscillating basal actomyosin contractions, which have provided significant insight into how the two polarized protein networks cooperate to produce an elongated egg. This review summarizes the proteins involved in Drosophila egg elongation and how this recent work has contributed to our current understanding of how egg elongation is achieved.


Subject(s)
Drosophila/cytology , Oogenesis , Actins/metabolism , Actins/physiology , Actomyosin/metabolism , Actomyosin/physiology , Animals , Cell Movement , Cell Shape , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/metabolism , Drosophila Proteins/physiology
4.
Dev Biol ; 333(1): 90-107, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19576200

ABSTRACT

During development, cells craft an impressive array of actin-based structures, mediating events as diverse as cytokinesis, apical constriction, and cell migration. One challenge is to determine how cells regulate actin assembly and disassembly to carry out these cell behaviors. During Drosophila oogenesis diverse cell behaviors are seen in the soma and germline. We used oogenesis to explore developmental roles of two important actin regulators: Enabled/VASP proteins and Capping protein. We found that Enabled plays an important role in cortical integrity of nurse cells, formation of robust bundled actin filaments in late nurse cells that facilitate nurse cell dumping, and migration of somatic border cells. During nurse cell dumping, Enabled localizes to barbed ends of the nurse cell actin filaments, suggesting its mechanism of action. We further pursued this mechanism using mutant Enabled proteins, each affecting one of its protein domains. These data suggest critical roles for the EVH2 domain and its tetramerization subdomain, while the EVH1 domain appears less critical. Enabled appears to be negatively regulated during oogenesis by Abelson kinase. We also explored the function of Capping protein. This revealed important roles in oocyte determination, nurse cell cortical integrity and nurse cell dumping, and support the idea that Capping protein and Enabled act antagonistically during dumping. Together these data reveal places that these actin regulators shape oogenesis.


Subject(s)
Actin Capping Proteins/physiology , Actin Cytoskeleton/physiology , DNA-Binding Proteins/physiology , Animals , Cell Movement/physiology , Cell Shape/physiology , Drosophila , Female , Oogenesis/physiology
5.
Development ; 134(11): 2027-39, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17507404

ABSTRACT

Studies in cultured cells and in vitro have identified many actin regulators and begun to define their mechanisms of action. Among these are Enabled (Ena)/VASP proteins, anti-Capping proteins that influence fibroblast migration, growth cone motility, and keratinocyte cell adhesion in vitro. However, partially redundant family members in mammals and maternal Ena contribution in Drosophila previously prevented assessment of the roles of Ena/VASP proteins in embryonic morphogenesis in flies or mammals. We used several approaches to remove maternal and zygotic Ena function, allowing us to address this question. We found that inactivating Ena does not disrupt cell adhesion or epithelial organization, suggesting its role in these processes is cell type-specific. However, Ena plays an important role in many morphogenetic events, including germband retraction, segmental groove retraction and head involution, whereas it is dispensable for other morphogenetic movements. We focused on dorsal closure, analyzing mechanisms by which Ena acts. Ena modulates filopodial number and length, thus influencing the speed of epithelial zippering and the ability of cells to match with correct neighbors. We also explored filopodial regulation in cultured Drosophila cells and embryos. These data provide new insights into developmental and mechanistic roles of this important actin regulator.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila/embryology , Epithelium/embryology , Morphogenesis/physiology , Actins/metabolism , Animals , Immunohistochemistry , Morphogenesis/genetics , Pseudopodia/physiology
6.
Cell ; 123(5): 769-72, 2005 Dec 02.
Article in English | MEDLINE | ID: mdl-16325573

ABSTRACT

Coupling between cell adhesion and the actin cytoskeleton is thought to require a stable link between the cadherin-catenin complex and actin that is mediated by alpha-catenin. In this issue of Cell, the Weis and Nelson groups call this static model into question, showing that alpha-catenin can directly regulate actin dynamics (Drees et al., 2005 and Yamada et al., 2005).


Subject(s)
Actins/metabolism , Adherens Junctions/metabolism , Models, Biological , alpha Catenin/metabolism , Animals , Cell Adhesion/physiology , Multiprotein Complexes
7.
Development ; 131(1): 25-36, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14645129

ABSTRACT

Pulses of the steroid hormone ecdysone trigger the major developmental transitions in Drosophila, including molting and puparium formation. The ecdysone signal is transduced by the EcR/USP nuclear receptor heterodimer that binds to specific response elements in the genome and directly regulates target gene transcription. We describe a novel nuclear receptor interacting protein encoded by rigor mortis (rig) that is required for ecdysone responses during larval development. rig mutants display defects in molting, delayed larval development, larval lethality, duplicated mouth parts, and defects in puparium formation--phenotypes that resemble those seen in EcR, usp, E75A and betaFTZ-F1 mutants. Although the expression of these nuclear receptor genes is essentially normal in rig mutant larvae, the ecdysone-triggered switch in E74 isoform expression is defective. rig encodes a protein with multiple WD-40 repeats and an LXXLL motif, sequences that act as specific protein-protein interaction domains. Consistent with the presence of these elements and the lethal phenotypes of rig mutants, Rig protein interacts with several Drosophila nuclear receptors in GST pull-down experiments, including EcR, USP, DHR3, SVP and betaFTZ-F1. The ligand binding domain of betaFTZ-F1 is sufficient for this interaction, which can occur in an AF-2-independent manner. Antibody stains reveal that Rig protein is present in the brain and imaginal discs of second and third instar larvae, where it is restricted to the cytoplasm. In larval salivary gland and midgut cells, however, Rig shuttles between the cytoplasm and nucleus in a spatially and temporally regulated manner, at times that correlate with the major lethal phase of rig mutants and major switches in ecdysone-regulated gene expression. Taken together, these data indicate that rig exerts essential functions during larval development through gene-specific effects on ecdysone-regulated transcription, most likely as a cofactor for one or more nuclear receptors. Furthermore, the dynamic intracellular redistribution of Rig protein suggests that it may act to refine spatial and temporal responses to ecdysone during development.


Subject(s)
Carrier Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Ecdysone/physiology , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction/physiology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Brain/growth & development , Carrier Proteins/genetics , Cell Nucleus/physiology , DNA Primers , Dimerization , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Larva , Molecular Sequence Data , Polymerase Chain Reaction , Protein Isoforms/genetics , Protein Isoforms/physiology , Pupa , Receptors, Cytoplasmic and Nuclear/genetics
8.
J Cell Biol ; 163(6): 1267-79, 2003 Dec 22.
Article in English | MEDLINE | ID: mdl-14676307

ABSTRACT

The proto-oncogenic kinase Abelson (Abl) regulates actin in response to cell signaling. Drosophila Abl is required in the nervous system, and also in epithelial cells, where it regulates adherens junction stability and actin organization. Abl acts at least in part via the actin regulator Enabled (Ena), but the mechanism by which Abl regulates Ena is unknown. We describe a novel role for Abl in early Drosophila development, where it regulates the site and type of actin structures produced. In Abl's absence, excess actin is polymerized in apical microvilli, whereas too little actin is assembled into pseudocleavage and cellularization furrows. These effects involve Ena misregulation. In abl mutants, Ena accumulates ectopically at the apical cortex where excess actin is observed, suggesting that Abl regulates Ena's subcellular localization. We also examined other actin regulators. Loss of Abl leads to changes in the localization of the Arp2/3 complex and the formin Diaphanous, and mutations in diaphanous or capping protein beta enhance abl phenotypes.


Subject(s)
Actins/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins , Drosophila melanogaster/embryology , Embryo, Nonmammalian/embryology , Proto-Oncogene Proteins c-abl/metabolism , Actin Depolymerizing Factors , Actin-Related Protein 2 , Actins/genetics , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Destrin , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Formins , Gene Expression Regulation, Developmental/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microvilli/metabolism , Microvilli/ultrastructure , Mutation/genetics , Phenotype , Protein Transport/physiology , Proto-Oncogene Proteins c-abl/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...