Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cell Sci ; 134(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34704593

RESUMEN

In response to environmental stress, human cells have been shown to form reversible amyloid aggregates within the nucleus, termed amyloid bodies (A-bodies). These protective physiological structures share many of the biophysical characteristics associated with the pathological amyloids found in Alzheimer's and Parkinson's disease. Here, we show that A-bodies are evolutionarily conserved across the eukaryotic domain, with their detection in Drosophila melanogaster and Saccharomyces cerevisiae marking the first examples of these functional amyloids being induced outside of a cultured cell setting. The conditions triggering amyloidogenesis varied significantly among the species tested, with results indicating that A-body formation is a severe, but sublethal, stress response pathway that is tailored to the environmental norms of an organism. RNA-sequencing analyses demonstrate that the regulatory low-complexity long non-coding RNAs that drive A-body aggregation are both conserved and essential in human, mouse and chicken cells. Thus, the identification of these natural and reversible functional amyloids in a variety of evolutionarily diverse species highlights the physiological significance of this protein conformation, and will be informative in advancing our understanding of both functional and pathological amyloid aggregation events. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Drosophila melanogaster , Animales , Biofisica , Drosophila melanogaster/genética , Ratones
2.
Biol Open ; 10(8)2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34296248

RESUMEN

Steroid hormones influence diverse biological processes throughout the animal life cycle, including metabolism, stress resistance, reproduction, and lifespan. In insects, the steroid hormone, 20-hydroxyecdysone (20E), is the central hormone regulator of molting and metamorphosis, and plays roles in tissue morphogenesis. For example, amnioserosa contraction, which is a major driving force in Drosophila dorsal closure (DC), is defective in embryos mutant for 20E biosynthesis. Here, we show that 20E signaling modulates the transcription of several DC participants in the amnioserosa and other dorsal tissues during late embryonic development, including zipper, which encodes for non-muscle myosin. Canonical ecdysone signaling typically involves the binding of Ecdysone receptor (EcR) and Ultraspiracle heterodimers to ecdysone-response elements (EcREs) within the promoters of responsive genes to drive expression. During DC, however, we provide evidence that 20E signaling instead acts in parallel to the JNK cascade via a direct interaction between EcR and the AP-1 transcription factor subunit, Jun, which together binds to genomic regions containing AP-1 binding sites but no EcREs to control gene expression. Our work demonstrates a novel mode of action for 20E signaling in Drosophila that likely functions beyond DC, and may provide further insights into mammalian steroid hormone receptor interactions with AP-1.


Asunto(s)
Drosophila/embriología , Ecdisterona/metabolismo , Morfogénesis , Transducción de Señal , Animales , Epidermis/metabolismo , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Metamorfosis Biológica , Subunidades de Proteína , Factor de Transcripción AP-1/metabolismo
3.
J Cell Sci ; 133(23)2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33199523

RESUMEN

Both functional and dysfunctional mitochondria are known to underlie tumor progression. Here, we establish use of the proto-oncogene Drosophila Homeodomain-interacting protein kinase (Hipk) as a new tool to address this paradox. We find that, in Hipk-overexpressing tumor-like cells, mitochondria accumulate and switch from fragmented to highly fused interconnected morphologies. Moreover, elevated Hipk promotes mitochondrial membrane hyperpolarization. These mitochondrial changes are at least in part driven by the upregulation of Myc. Furthermore, we show that the altered mitochondrial energetics, but not morphology, is required for Hipk-induced tumor-like growth, because knockdown of pdsw (also known as nd-pdsw; NDUFB10 in mammals; a Complex I subunit) abrogates the growth. Knockdown of ATPsynß (a Complex V subunit), which produces higher levels of reactive oxygen species (ROS) than pdsw knockdown, instead synergizes with Hipk to potentiate JNK activation and the downstream induction of matrix metalloproteinases. Accordingly, ATPsynß knockdown suppresses Hipk-induced tumor-like growth only when ROS scavengers are co-expressed. Together, our work presents an in vivo tumor model featuring the accumulation of hyperfused and hyperpolarized mitochondria, and reveals respiratory complex subunit-dependent opposing effects on tumorigenic outcomes.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Drosophila melanogaster , Neoplasias , Animales , Carcinogénesis , Drosophila melanogaster/genética , Mitocondrias/genética , Proteínas Quinasas , Especies Reactivas de Oxígeno
4.
PLoS One ; 15(3): e0221006, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32187190

RESUMEN

Homeodomain-interacting protein kinases (Hipks) have been previously associated with cell proliferation and cancer, however, their effects in the nervous system are less well understood. We have used Drosophila melanogaster to evaluate the effects of altered Hipk expression on the nervous system and muscle. Using genetic manipulation of Hipk expression we demonstrate that knockdown and over-expression of Hipk produces early adult lethality, possibly due to the effects on the nervous system and muscle involvement. We find that optimal levels of Hipk are critical for the function of dopaminergic neurons and glial cells in the nervous system, as well as muscle. Furthermore, manipulation of Hipk affects the structure of the larval neuromuscular junction (NMJ) by promoting its growth. Hipk regulates the phosphorylation of the synapse-associated cytoskeletal protein Hu-li tai shao (Hts; adducin in mammals) and modulates the expression of two important protein kinases, Calcium-calmodulin protein kinase II (CaMKII) and Partitioning-defective 1 (PAR-1), all of which may alter neuromuscular structure/function and influence lethality. Hipk also modifies the levels of an important nuclear protein, TBPH, the fly orthologue of TAR DNA-binding protein 43 (TDP-43), which may have relevance for understanding motor neuron diseases.


Asunto(s)
Proteínas de Drosophila/aislamiento & purificación , Drosophila melanogaster/enzimología , Drosophila melanogaster/fisiología , Músculos/anatomía & histología , Músculos/metabolismo , Sistema Nervioso/anatomía & histología , Sistema Nervioso/metabolismo , Proteínas Quinasas/aislamiento & purificación , Animales , Tipificación del Cuerpo , Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/anatomía & histología , Ojo/embriología , Larva/metabolismo , Masculino , Músculos/citología , Sistema Nervioso/citología , Unión Neuromuscular/metabolismo , Tamaño de los Órganos , Fosforilación , Sinapsis/metabolismo
5.
Methods Mol Biol ; 1652: 37-42, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28791632

RESUMEN

Genetic analysis of Egfr signaling in Drosophila has a long-standing track record of making important contributions to our understanding of the Egfr pathway. While the central Ras/MAPK pathway has long been well defined, there is much to learn with regard to its cross talk with other pathways and how it is regulated. A better understanding of the regulation of Egfr signaling is of particular interest with regard to the participation of misregulated Egfr signaling in tumorigenesis. Recent studies in the fly have led to some surprising results, identifying regulators of Egfr acting in unexpected ways.


Asunto(s)
Drosophila/metabolismo , Receptores ErbB/metabolismo , Transducción de Señal , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Receptores ErbB/genética , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Transducción de Señal/genética , Proteínas ras/genética , Proteínas ras/metabolismo
6.
Methods Mol Biol ; 1652: 229-256, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28791646

RESUMEN

For several decades, genetic analysis in Drosophila has made important contributions to the understanding of signaling by Egfr. Egfr has been well characterized with regard to its oncogenic potential but is also being studied for its roles in organismal development. We have recently developed dorsal closure of the Drosophila embryo as a system for characterizing Egfr regulation of events that do not involve proliferation, as no cell divisions occur during this process. Dorsal closure is essentially a developmental wound healing event with parallels to vertebrate developmental epithelial fusions such as neural tube closure and palate fusion. We describe here a set of materials and protocols for studying Egfr signaling during dorsal closure, including assessing effects of altering Egfr signaling on other pathways, gene expression and, using live imaging, morphogenesis and programmed cell death. Although this "tool kit" is designed for looking at Egfr, it can be readily adapted to look at the participation of any signaling molecule in dorsal closure.


Asunto(s)
Drosophila/embriología , Drosophila/metabolismo , Embrión no Mamífero , Desarrollo Embrionario , Receptores ErbB/metabolismo , Morfogénesis , Transducción de Señal , Animales , Drosophila/genética , Desarrollo Embrionario/genética , Receptores ErbB/genética , Homocigoto , Inmunohistoquímica , Hibridación Fluorescente in Situ , Microscopía , Imagen Molecular , Morfogénesis/genética , Mutación , Imagen de Lapso de Tiempo
7.
J Cell Sci ; 129(16): 3067-76, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27528207

RESUMEN

The pleated septate junction (pSJ), an ancient structure for cell-cell contact in invertebrate epithelia, has protein components that are found in three more-recent junctional structures, the neuronal synapse, the paranodal region of the myelinated axon and the vertebrate epithelial tight junction. These more-recent structures appear to have evolved through alterations of the ancestral septate junction. During its formation in the developing animal, the pSJ exhibits plasticity, although the final structure is extremely robust. Similar to the immature pSJ, the synapse and tight junctions both exhibit plasticity, and we consider evidence that this plasticity comes at least in part from the interaction of members of the immunoglobulin cell adhesion molecule superfamily with highly regulated membrane-associated guanylate kinases. This plasticity regulation probably arose in order to modulate the ancestral pSJ and is maintained in the derived structures; we suggest that it would be beneficial when studying plasticity of one of these structures to consider the literature on the others. Finally, looking beyond the junctions, we highlight parallels between epithelial and synaptic membranes, which both show a polarized distribution of many of the same proteins - evidence that determinants of apicobasal polarity in epithelia also participate in patterning of the synapse.


Asunto(s)
Evolución Biológica , Uniones Intercelulares/metabolismo , Sinapsis/metabolismo , Animales , Epitelio/metabolismo , Humanos , Membranas/metabolismo , Modelos Biológicos
9.
Front Cell Neurosci ; 10: 11, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26858605

RESUMEN

The neurological dysfunction in amyotrophic lateral sclerosis (ALS)/motor neurone disease (MND) is associated with defective nerve-muscle contacts early in the disease suggesting that perturbations of cell adhesion molecules (CAMs) linking the pre- and post-synaptic components of the neuromuscular junction (NMJ) are involved. To search for candidate proteins implicated in this degenerative process, researchers have studied the Drosophila larval NMJ and find that the cytoskeleton-associated protein, adducin, is ideally placed to regulate synaptic contacts. By controlling the levels of synaptic proteins, adducin can de-stabilize synaptic contacts. Interestingly, elevated levels of phosphorylated adducin have been reported in ALS patients and in a mouse model of the disease. Adducin is regulated by phosphorylation through protein kinase C (PKC), some isoforms of which exhibit Ca(2+)-dependence, raising the possibility that changes in intracellular Ca(2+) might alter PKC activation and secondarily influence adducin phosphorylation. Furthermore, adducin has interactions with the alpha subunit of the Na(+)/K(+)-ATPase. Thus, the phosphorylation of adducin may secondarily influence synaptic stability at the NMJ and so influence pre- and post-synaptic integrity at the NMJ in ALS.

10.
J Vis Exp ; (95): 52139, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25650626

RESUMEN

Discs large (Dlg) is a conserved member of the membrane-associated guanylate kinase family, and serves as a major scaffolding protein at the larval neuromuscular junction (NMJ) in Drosophila. Previous studies have shown that the postsynaptic distribution of Dlg at the larval NMJ overlaps with that of Hu-li tai shao (Hts), a homologue to the mammalian adducins. In addition, Dlg and Hts are observed to form a complex with each other based on co-immunoprecipitation experiments involving whole adult fly lysates. Due to the nature of these experiments, however, it was unknown whether this complex exists specifically at the NMJ during larval development. Proximity Ligation Assay (PLA) is a recently developed technique used mostly in cell and tissue culture that can detect protein-protein interactions in situ. In this assay, samples are incubated with primary antibodies against the two proteins of interest using standard immunohistochemical procedures. The primary antibodies are then detected with a specially designed pair of oligonucleotide-conjugated secondary antibodies, termed PLA probes, which can be used to generate a signal only when the two probes have bound in close proximity to each other. Thus, proteins that are in a complex can be visualized. Here, it is demonstrated how PLA can be used to detect in situ protein-protein interactions at the Drosophila larval NMJ. The technique is performed on larval body wall muscle preparations to show that a complex between Dlg and Hts does indeed exist at the postsynaptic region of NMJs.


Asunto(s)
Proteínas de Unión a Calmodulina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Unión Neuromuscular/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas de Unión a Calmodulina/análisis , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/análisis , Guanilato-Quinasas/metabolismo , Inmunohistoquímica , Larva/química , Larva/metabolismo , Unión Neuromuscular/química , Mapeo de Interacción de Proteínas/métodos , Proteínas Supresoras de Tumor/análisis
11.
Development ; 142(1): 82-91, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25516970

RESUMEN

In the germarium of the Drosophila ovary, germline cysts are encapsulated one at a time by a follicular epithelium derived from two follicle stem cells (FSCs). Ovaries in flies mutant for the serine/threonine kinase Pak exhibit a novel phenotype, in which two side-by-side cysts are encapsulated at a time, generating paired egg chambers. This striking phenotype originates in the pupal ovary, where the developing germarium is shaped by the basal stalk, a stack of cells formed by cell intercalation. The process of basal stalk formation is not well understood, and we provide evidence that the cell intercalation is driven by actomyosin contractility of DE-Cadherin-adhered cells, leading to a column of disk-shaped cells exhibiting a novel radial cell polarity. Cell intercalation fails in Pak mutant ovaries, leading to abnormally wide basal stalks and consequently wide germaria with side-by-side cysts. We present evidence that Pak mutant germaria have extra FSCs, and we propose that contact of a germline cyst with the basal stalk in the pupal ovary contributes to FSC niche formation. The wide basal stalk in Pak mutants enables the formation of extra FSC niches which are mispositioned and yet functional, indicating that the FSC niche can be established in diverse locations.


Asunto(s)
Polaridad Celular , Drosophila melanogaster/citología , Drosophila melanogaster/enzimología , Folículo Ovárico/citología , Folículo Ovárico/enzimología , Nicho de Células Madre , Animales , Proteínas de Drosophila , Drosophila melanogaster/anatomía & histología , Femenino , Modelos Biológicos , Mutación/genética , Óvulo/citología , Óvulo/metabolismo , Fenotipo , Quinasas p21 Activadas
12.
Biol Open ; 3(12): 1196-206, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25416060

RESUMEN

Adducin is a ubiquitously expressed actin- and spectrin-binding protein involved in cytoskeleton organization, and is regulated through phosphorylation of the myristoylated alanine-rich C-terminal kinase (MARCKS)-homology domain by protein kinase C (PKC). We have previously shown that the Drosophila adducin, Hu-li tai shao (Hts), plays a role in larval neuromuscular junction (NMJ) growth. Here, we find that the predominant isoforms of Hts at the NMJ contain the MARCKS-homology domain, which is important for interactions with Discs large (Dlg) and phosphatidylinositol 4,5-bisphosphate (PIP2). Through the use of Proximity Ligation Assay (PLA), we show that the adducin-like Hts isoforms are in complexes with Dlg and PIP2 at the NMJ. We provide evidence that Hts promotes the phosphorylation and delocalization of Dlg at the NMJ through regulation of the transcript distribution of the PAR-1 and CaMKII kinases in the muscle. We also show that Hts interactions with Dlg and PIP2 are impeded through phosphorylation of the MARCKS-homology domain. These results are further evidence that Hts is a signaling-responsive regulator of synaptic plasticity in Drosophila.

13.
PLoS One ; 8(4): e60180, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23579691

RESUMEN

During Drosophila embryogenesis the process of dorsal closure (DC) results in continuity of the embryonic epidermis, and DC is well recognized as a model system for the analysis of epithelial morphogenesis as well as wound healing. During DC the flanking lateral epidermal sheets stretch, align, and fuse along the dorsal midline, thereby sealing a hole in the epidermis occupied by an extra-embryonic tissue known as the amnioserosa (AS). Successful DC requires the regulation of cell shape change via actomyosin contractility in both the epidermis and the AS, and this involves bidirectional communication between these two tissues. We previously demonstrated that transcriptional regulation of myosin from the zipper (zip) locus in both the epidermis and the AS involves the expression of Ack family tyrosine kinases in the AS in conjunction with Dpp secreted from the epidermis. A major function of Ack in other species, however, involves the negative regulation of Egfr. We have, therefore, asked what role Egfr might play in the regulation of DC. Our studies demonstrate that Egfr is required to negatively regulate epidermal expression of dpp during DC. Interestingly, we also find that Egfr signaling in the AS is required to repress zip expression in both the AS and the epidermis, and this may be generally restrictive to the progression of morphogenesis in these tissues. Consistent with this theme of restricting morphogenesis, it has previously been shown that programmed cell death of the AS is essential for proper DC, and we show that Egfr signaling also functions to inhibit or delay AS programmed cell death. Finally, we present evidence that Ack regulates zip expression by promoting the endocytosis of Egfr in the AS. We propose that the general role of Egfr signaling during DC is that of a braking mechanism on the overall progression of DC.


Asunto(s)
Drosophila/embriología , Drosophila/genética , Receptores ErbB/genética , Morfogénesis/genética , Alelos , Animales , Apoptosis/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Embrión no Mamífero/metabolismo , Endocitosis/genética , Células Epidérmicas , Epidermis/embriología , Epidermis/metabolismo , Receptores ErbB/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/genética , Modelos Biológicos , Cadenas Pesadas de Miosina/genética , Proteínas Tirosina Quinasas/genética , Transducción de Señal
14.
Dev Biol ; 357(2): 392-403, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21791202

RESUMEN

Adducin is a cytoskeletal protein having regulatory roles that involve actin filaments, functions that are inhibited by phosphorylation of adducin by protein kinase C. Adducin is hyperphosphorylated in nervous system tissue in patients with the neurodegenerative disease amyotrophic lateral sclerosis, and mice lacking ß-adducin have impaired synaptic plasticity and learning. We have found that Drosophila adducin, encoded by hu-li tai shao (hts), is localized to the post-synaptic larval neuromuscular junction (NMJ) in a complex with the scaffolding protein Discs large (Dlg), a regulator of synaptic plasticity during growth of the NMJ. hts mutant NMJs are underdeveloped, whereas over-expression of Hts promotes Dlg phosphorylation, delocalizes Dlg away from the NMJ, and causes NMJ overgrowth. Dlg is a component of septate junctions at the lateral membrane of epithelial cells, and we show that Hts regulates Dlg localization in the amnioserosa, an embryonic epithelium, and that embryos doubly mutant for hts and dlg exhibit defects in epithelial morphogenesis. The phosphorylation of Dlg by the kinases PAR-1 and CaMKII has been shown to disrupt Dlg targeting to the NMJ and we present evidence that Hts regulates Dlg targeting to the NMJ in muscle and the lateral membrane of epithelial cells by controlling the protein levels of PAR-1 and CaMKII, and consequently the extent of Dlg phosphorylation.


Asunto(s)
Proteínas de Unión a Calmodulina/metabolismo , Membrana Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Epiteliales/metabolismo , Epitelio/metabolismo , Sinapsis/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Células Epiteliales/citología , Epitelio/embriología , Larva/citología , Larva/metabolismo , Modelos Biológicos , Músculos/metabolismo , Mutagénesis Insercional/genética , Unión Neuromuscular/citología , Unión Neuromuscular/embriología , Unión Neuromuscular/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas
15.
Genetics ; 187(2): 501-12, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21098722

RESUMEN

During Drosophila oogenesis, basally localized F-actin bundles in the follicle cells covering the egg chamber drive its elongation along the anterior-posterior axis. The basal F-actin of the follicle cell is an attractive system for the genetic analysis of the regulation of the actin cytoskeleton, and results obtained in this system are likely to be broadly applicable in understanding tissue remodeling. Mutations in a number of genes, including that encoding the p21-activated kinase Pak, have been shown to disrupt organization of the basal F-actin and in turn affect egg chamber elongation. pak mutant egg chambers have disorganized F-actin distribution and remain spherical due to a failure to elongate. In a genetic screen to identify modifiers of the pak rounded egg chamber phenotype several second chromosome deficiencies were identified as suppressors. One suppressing deficiency removes the rho1 locus, and we determined using several rho1 alleles that removal of a single copy of rho1 can suppress the pak phenotype. Reduction of any component of the Rho1-activated actomyosin contractility pathway suppresses pak oogenesis defects, suggesting that Pak counteracts Rho1 signaling. There is ectopic myosin light chain phosphorylation in pak mutant follicle cell clones in elongating egg chambers, probably due at least in part to mislocalization of RhoGEF2, an activator of the Rho1 pathway. In early egg chambers, pak mutant follicle cells have reduced levels of myosin phosphorylation and we conclude that Pak both promotes and restricts myosin light chain phosphorylation in a temporally distinct manner during oogenesis.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila , Oogénesis/genética , Transducción de Señal , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Actomiosina/metabolismo , Animales , Drosophila/enzimología , Drosophila/genética , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Femenino , Genotipo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Modelos Biológicos , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Folículo Ovárico/metabolismo , Fenotipo , Fosforilación , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/genética , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/genética
16.
Development ; 137(12): 2023-32, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20501591

RESUMEN

Dorsal closure (DC) of the Drosophila embryo is a model for the study of wound healing and developmental epithelial fusions, and involves the sealing of a hole in the epidermis through the migration of the epidermal flanks over the tissue occupying the hole, the amnioserosa. During DC, the cells at the edge of the migrating epidermis extend Rac- and Cdc42-dependent actin-based lamellipodia and filopodia from their leading edge (LE), which exhibits a breakdown in apicobasal polarity as adhesions are severed with the neighbouring amnioserosa cells. Studies using mammalian cells have demonstrated that Scribble (Scrib), an important determinant of apicobasal polarity that functions in a protein complex, controls polarized cell migration through recruitment of Rac, Cdc42 and the serine/threonine kinase Pak, an effector for Rac and Cdc42, to the LE. We have used DC and the follicular epithelium to study the relationship between Pak and the Scrib complex at epithelial membranes undergoing changes in apicobasal polarity and adhesion during development. We propose that, during DC, the LE membrane undergoes an epithelial-to-mesenchymal-like transition to initiate epithelial sheet migration, followed by a mesenchymal-to-epithelial-like transition as the epithelial sheets meet up and restore cell-cell adhesion. This latter event requires integrin-localized Pak, which recruits the Scrib complex in septate junction formation. We conclude that there are bidirectional interactions between Pak and the Scrib complex modulating epithelial plasticity. Scrib can recruit Pak to the LE for polarized cell migration but, as migratory cells meet up, Pak can recruit the Scrib complex to restore apicobasal polarity and cell-cell adhesion.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Drosophila/metabolismo , Epitelio/metabolismo , Quinasas p21 Activadas/metabolismo , Actinas/metabolismo , Animales , Adhesión Celular , Membrana Celular/metabolismo , Polaridad Celular , Drosophila/metabolismo , Epidermis/metabolismo , Integrinas/metabolismo , Seudópodos/metabolismo , Uniones Estrechas/metabolismo
17.
Curr Biol ; 19(17): 1429-37, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19716703

RESUMEN

BACKGROUND: Developmental processes are intimately tied to signaling events that integrate the dynamic reorganization of the actin cytoskeleton and membrane dynamics. The F-BAR-domain-containing proteins are prime candidates to couple actin dynamics and membrane trafficking in different morphogenetic processes. RESULTS: Here, we present the functional analysis of the Drosophila F-BAR protein Cip4/Toca1 (Cdc42-interacting protein 4/transducer of Cdc42-dependent actin assembly 1). Cip4 is able to form a complex with WASP and SCAR/WAVE and recruits both actin-nucleation-promoting factors to invaginating membranes and endocytic vesicles. Actin-comet-tail-based movement of these vesicles depends not only on WASP but largely on WAVE function. In vivo, loss of cip4 function causes multiple wing hairs. A similar phenotype is observed when vesicle scission is affected after Dynamin suppression. Gene dosage experiments show that Cip4 and WAVE functionally interact to restrict wing hair formation. Further rescue experiments confirm that Cip4 is able to act through WAVE and WASP in vivo. Biochemical and functional data support a model in which Cdc42 acts upstream of Cip4 and recruits not only WASP but also SCAR/WAVE via Abi to control Dynamin-dependent cell polarization in the wing. CONCLUSION: Cip4 integrates membrane trafficking and actin dynamics through WASP and WAVE. First, Cip4 promotes membrane invaginations and triggers the vesicle scission by recruiting Dynamin to the neck of nascent vesicles. Second, Cip4 recruits WASP and WAVE proteins to induce actin polymerization, supporting vesicle scission and providing the force for vesicle movement.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Drosophila/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Actinas/metabolismo , Actinas/fisiología , Animales , Transporte Biológico , Tipificación del Cuerpo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Drosophila/citología , Proteínas de Drosophila/genética , Dinaminas/metabolismo , Dinaminas/fisiología , Endocitosis , Endosomas/metabolismo , Endosomas/fisiología , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/fisiología , Dosificación de Gen , Modelos Biológicos , Alas de Animales/anatomía & histología , Alas de Animales/crecimiento & desarrollo
18.
Dev Dyn ; 237(10): 2936-46, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18816840

RESUMEN

Dorsal closure of the Drosophila embryo is an epithelial fusion in which the epidermal flanks migrate to close a hole in the epidermis occupied by the amnioserosa, a process driven in part by myosin-dependent cell shape change. Dpp signaling is required for the morphogenesis of both tissues, where it promotes transcription of myosin from the zipper (zip) gene. Drosophila has two members of the activated Cdc42-associated kinase (ACK) family: DACK and PR2. Overexpression of DACK in embryos deficient in Dpp signaling can restore zip expression and suppress dorsal closure defects, while reducing the levels of DACK and PR2 simultaneously using mutations or amnioserosa-specific knock down by RNAi results in loss of zip expression. ACK function in the amnioserosa may generate a signal cooperating with Dpp secreted from the epidermis in driving zip expression in these two tissues, ensuring that cell shape changes in dorsal closure occur in a coordinated manner.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Miosinas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación/genética , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Proteínas Tirosina Quinasas/genética
19.
Dev Biol ; 305(2): 470-82, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17383630

RESUMEN

During epithelial development cells become polarized along their apical-basal axis and some epithelia also exhibit polarity in the plane of the tissue. Mutations in the gene encoding a Drosophila Pak family serine/threonine kinase, dPak, disrupt the follicular epithelium that covers developing egg chambers during oogenesis. The follicular epithelium normally exhibits planar polarized organization of basal F-actin bundles such that they lie perpendicular to the anterior-posterior axis of the egg chamber, and requires contact with the basement membrane for apical-basal polarization. During oogenesis, dPak becomes localized to the basal end of follicle cells and is required for polarized organization of the basal actin cytoskeleton and for epithelial integrity and apical-basal polarity. The receptor protein tyrosine phosphatase Dlar and integrins, all receptors for extracellular matrix proteins, are required for polarization of the basal F-actin bundles, and for correct dPak localization in follicle cells. dpak mutant follicle cells show increased beta(Heavy)-spectrin levels, and we speculate that dPak regulation of beta(Heavy)-spectrin, a known participant in the maintenance of membrane domains, is required for correct apical-basal polarization of the membrane. We propose that dPak mediates communication between the basement membrane and intracellular proteins required for polarization of the basal F-actin and for apical-basal polarity.


Asunto(s)
Actinas/metabolismo , Polaridad Celular/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/enzimología , Folículo Ovárico/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Membrana Basal/enzimología , Membrana Basal/metabolismo , Polaridad Celular/genética , Células Clonales , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Epitelio/enzimología , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Femenino , Humanos , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Quinasas p21 Activadas
20.
Dev Genes Evol ; 216(9): 543-50, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16609869

RESUMEN

The Rho family small GTPases Rho, Rac, and Cdc42 regulate cell shape and motility through the actin cytoskeleton. These proteins cycle between a GTP-bound "on" state and a GDP-bound "off" state and are negatively regulated by GTPase-activating proteins (GAPs), which accelerate the small GTPase's intrinsic hydrolysis of bound GTP to GDP. Drosophila RhoGAP68F is similar to the mammalian protein p50RhoGAP/Cdc42GAP, which exhibits strong GAP activity toward Cdc42. We find that, despite the strong similarities between RhoGAP68F and p50RhoGAP/Cdc42GAP, RhoGAP68F is most effective as a GAP for RhoA. These in vitro data are supported by the in vivo analysis of mutants in RhoGAP68F. We demonstrate through the characterization of two alleles of the RhoGAP68F gene that RhoGAP68F participates in gastrulation of the embryo, a morphogenetic event driven by cell constriction that involves RhoA signaling. We propose that RhoGAP68F functions as a regulator of RhoA signaling during gastrulation.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Drosophila/embriología , Proteínas Activadoras de GTPasa/fisiología , Gástrula/enzimología , Morfogénesis , Proteínas de Unión al GTP rho/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Drosophila/anatomía & histología , Drosophila/enzimología , Proteínas de Drosophila/genética , Proteínas Activadoras de GTPasa/genética , Datos de Secuencia Molecular , Morfogénesis/genética , Mutación , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA