Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nat Commun ; 13(1): 674, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35115535

RESUMEN

Conductin/axin2 is a scaffold protein negatively regulating the pro-proliferative Wnt/ß-catenin signaling pathway. Accumulation of scaffold proteins in condensates frequently increases their activity, but whether condensation contributes to Wnt pathway inhibition by conductin remains unclear. Here, we show that the Gαi2 subunit of trimeric G-proteins induces conductin condensation by targeting a polymerization-inhibiting aggregon in its RGS domain, thereby promoting conductin-mediated ß-catenin degradation. Consistently, transient Gαi2 expression inhibited, whereas knockdown activated Wnt signaling via conductin. Colorectal cancers appear to evade Gαi2-induced Wnt pathway suppression by decreased Gαi2 expression and inactivating mutations, associated with shorter patient survival. Notably, the Gαi2-activating drug guanabenz inhibited Wnt signaling via conductin, consequently reducing colorectal cancer growth in vitro and in mouse models. In summary, we demonstrate Wnt pathway inhibition via Gαi2-triggered conductin condensation, suggesting a tumor suppressor function for Gαi2 in colorectal cancer, and pointing to the FDA-approved drug guanabenz for targeted cancer therapy.


Asunto(s)
Proteína Axina/genética , Neoplasias Colorrectales/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Vía de Señalización Wnt/genética , beta Catenina/genética , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Proteína Axina/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Regulación Neoplásica de la Expresión Génica , Guanabenzo/farmacología , Células HEK293 , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Mutación , Vía de Señalización Wnt/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , beta Catenina/metabolismo
2.
Sci Rep ; 10(1): 21013, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33273532

RESUMEN

Drosophila melanogaster has been a model for multiple human disease conditions, including cancer. Among Drosophila tissues, the eye development is particularly sensitive to perturbations of the embryonic signaling pathways, whose improper activation in humans underlies various forms of cancer. We have launched the HumanaFly project, whereas human genes expressed in breast cancer patients are screened for their ability to aberrate development of the Drosophila eye, hoping to thus identify novel oncogenes. Here we report identification of a breast cancer transgene, which upon expression in Drosophila produces eye malformation similar to the famous Glazed phenotype discovered by Thomas Morgan and decades later dissected to originate from mis-expression of Wingless (Wg). Wg is the ortholog of human Wnt proteins serving as ligands to initiate the developmental/oncogenic Wnt signaling pathway. Through genetic experiments we identified that this transgene interacted with the Wg production machinery, rather than with Wg signal transduction. In Drosophila imaginal discs, we directly show that the transgene promoted long-range diffusion of Wg, affecting expression of the Wg target genes. The transgene emerged to encode RPS12-a protein of the small ribosomal subunit overexpressed in several cancer types and known to also possess extra-ribosomal functions. Our work identifies RPS12 as an unexpected regulator of secretion and activity of Wnts. As Wnt signaling is particularly important in the context of breast cancer initiation and progression, RPS12 might be implicated in tumorigenesis in this and other Wnt-dependent cancers. Continuation of our HumanaFly project may bring further discoveries on oncogenic mechanisms.


Asunto(s)
Neoplasias de la Mama/genética , Ojo Compuesto de los Artrópodos/metabolismo , Proteínas Mitocondriales/genética , Proteínas Ribosómicas/genética , Transgenes , Vía de Señalización Wnt , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Femenino , Humanos , Discos Imaginales/metabolismo , Proteínas Mitocondriales/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Ribosómicas/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
3.
Development ; 141(17): 3399-409, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25139856

RESUMEN

Drosophila neuromuscular junctions (NMJs) represent a powerful model system with which to study glutamatergic synapse formation and remodeling. Several proteins have been implicated in these processes, including components of canonical Wingless (Drosophila Wnt1) signaling and the giant isoforms of the membrane-cytoskeleton linker Ankyrin 2, but possible interconnections and cooperation between these proteins were unknown. Here, we demonstrate that the heterotrimeric G protein Go functions as a transducer of Wingless-Frizzled 2 signaling in the synapse. We identify Ankyrin 2 as a target of Go signaling required for NMJ formation. Moreover, the Go-ankyrin interaction is conserved in the mammalian neurite outgrowth pathway. Without ankyrins, a major switch in the Go-induced neuronal cytoskeleton program is observed, from microtubule-dependent neurite outgrowth to actin-dependent lamellopodial induction. These findings describe a novel mechanism regulating the microtubule cytoskeleton in the nervous system. Our work in Drosophila and mammalian cells suggests that this mechanism might be generally applicable in nervous system development and function.


Asunto(s)
Ancirinas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Receptores Frizzled/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Microtúbulos/metabolismo , Neuronas/citología , Proteína Wnt1/metabolismo , Animales , Línea Celular , Secuencia Conservada , Drosophila melanogaster/citología , Mamíferos , Ratones , Neuritas/metabolismo , Unión Neuromuscular/metabolismo , Neuronas/metabolismo , Unión Proteica , Transducción de Señal , Sinapsis/metabolismo
4.
Mol Cell ; 53(4): 663-71, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24560274

RESUMEN

Regulator of G protein signaling (RGS) proteins accelerate GTP hydrolysis on G protein α subunits, restricting their activity downstream from G protein-coupled receptors. Here we identify Drosophila Double hit (Dhit) as a dual RGS regulator of Gαo. In addition to the conventional GTPase-activating action, Dhit possesses the guanine nucleotide dissociation inhibitor (GDI) activity, slowing the rate of GTP uptake by Gαo; both activities are mediated by the same RGS domain. These findings are recapitulated using homologous mammalian Gαo/i proteins and RGS19. Crystal structure and mutagenesis studies provide clues into the molecular mechanism for this unprecedented GDI activity. Physiologically, we confirm this activity in Drosophila asymmetric cell divisions and HEK293T cells. We show that the oncogenic Gαo mutant found in breast cancer escapes this GDI regulation. Our studies identify Dhit and its homologs as double-action regulators, inhibiting Gαo/i proteins both through suppression of their activation and acceleration of their inactivation through the single RGS domain.


Asunto(s)
Proteínas de Drosophila/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas RGS/metabolismo , Regiones no Traducidas 5' , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/metabolismo , Cristalografía por Rayos X , Drosophila melanogaster , Femenino , Guanosina Trifosfato/química , Células HEK293 , Humanos , Hidrólisis , Datos de Secuencia Molecular , Mutación , Sistemas de Lectura Abierta , Estructura Terciaria de Proteína , Transducción de Señal , Factores de Tiempo
5.
PLoS One ; 8(7): e70327, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936187

RESUMEN

Analyses of cultured cells and transgenic mice expressing prion protein (PrP) deletion mutants have revealed that some properties of PrP -such as its ability to misfold, aggregate and trigger neurotoxicity- are controlled by discrete molecular determinants within its protein domains. Although the contributions of these determinants to PrP biosynthesis and turnover are relatively well characterized, it is still unclear how they modulate cellular functions of PrP. To address this question, we used two defined activities of PrP as functional readouts: 1) the recruitment of PrP to cell-cell contacts in Drosophila S2 and human MCF-7 epithelial cells, and 2) the induction of PrP embryonic loss- and gain-of-function phenotypes in zebrafish. Our results show that homologous mutations in mouse and zebrafish PrPs similarly affect their subcellular localization patterns as well as their in vitro and in vivo activities. Among PrP's essential features, the N-terminal leader peptide was sufficient to drive targeting of our constructs to cell contact sites, whereas lack of GPI-anchoring and N-glycosylation rendered them inactive by blocking their cell surface expression. Importantly, our data suggest that the ability of PrP to homophilically trans-interact and elicit intracellular signaling is primarily encoded in its globular domain, and modulated by its repetitive domain. Thus, while the latter induces the local accumulation of PrPs at discrete punctae along cell contacts, the former counteracts this effect by promoting the continuous distribution of PrP. In early zebrafish embryos, deletion of either domain significantly impaired PrP's ability to modulate E-cadherin cell adhesion. Altogether, these experiments relate structural features of PrP to its subcellular distribution and in vivo activity. Furthermore, they show that despite their large evolutionary history, the roles of PrP domains and posttranslational modifications are conserved between mouse and zebrafish.


Asunto(s)
Espacio Intracelular/metabolismo , Priones/química , Priones/metabolismo , Estructura Terciaria de Proteína , Citoesqueleto de Actina/metabolismo , Animales , Animales Modificados Genéticamente , Cadherinas/metabolismo , Adhesión Celular/genética , Comunicación Celular/genética , Línea Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Glicosilación , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células MCF-7 , Ratones , Ratones Transgénicos , Microscopía Confocal , Mutación , Priones/genética , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
6.
Mol Biol Cell ; 24(17): 2689-702, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23825023

RESUMEN

The lipid raft proteins reggie-1 and -2 (flotillins) are implicated in membrane protein trafficking but exactly how has been elusive. We find that reggie-1 and -2 associate with the Rab11a, SNX4, and EHD1-decorated tubulovesicular recycling compartment in HeLa cells and that reggie-1 directly interacts with Rab11a and SNX4. Short hairpin RNA-mediated down-regulation of reggie-1 (and -2) in HeLa cells reduces association of Rab11a with tubular structures and impairs recycling of the transferrin-transferrin receptor (TfR) complex to the plasma membrane. Overexpression of constitutively active Rab11a rescues TfR recycling in reggie-deficient HeLa cells. Similarly, in a Ca(2+) switch assay in reggie-depleted A431 cells, internalized E-cadherin is not efficiently recycled to the plasma membrane upon Ca(2+) repletion. E-cadherin recycling is rescued, however, by overexpression of constitutively active Rab11a or SNX4 in reggie-deficient A431 cells. This suggests that the function of reggie-1 in sorting and recycling occurs in association with Rab11a and SNX4. Of interest, impaired recycling in reggie-deficient cells leads to de novo E-cadherin biosynthesis and cell contact reformation, showing that cells have ways to compensate the loss of reggies. Together our results identify reggie-1 as a regulator of the Rab11a/SNX4-controlled sorting and recycling pathway, which is, like reggies, evolutionarily conserved.


Asunto(s)
Cadherinas/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Transferrina/metabolismo , Nexinas de Clasificación/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Cadherinas/genética , Línea Celular Tumoral , Movimiento Celular/fisiología , Regulación hacia Abajo , Células HeLa , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Filogenia , Transporte de Proteínas
7.
Int J Mol Sci ; 14(3): 5130-45, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23455472

RESUMEN

Concentration gradients formed by the lipid-modified morphogens of the Wnt family are known for their pivotal roles during embryogenesis and adult tissue homeostasis. Wnt morphogens are also implicated in a variety of human diseases, especially cancer. Therefore, the signaling cascades triggered by Wnts have received considerable attention during recent decades. However, how Wnts are secreted and how concentration gradients are formed remains poorly understood. The use of model organisms such as Drosophila melanogaster has provided important advances in this area. For instance, we have previously shown that the lipid raft-associated reggie/flotillin proteins influence Wnt secretion and spreading in Drosophila. Our work supports the notion that producing cells secrete Wnt molecules in at least two pools: a poorly diffusible one and a reggie/flotillin-dependent highly diffusible pool which allows morphogen spreading over long distances away from its source of production. Here we revise the current views of Wnt secretion and spreading, and propose two models for the role of the reggie/flotillin proteins in these processes: (i) reggies/flotillins regulate the basolateral endocytosis of the poorly diffusible, membrane-bound Wnt pool, which is then sorted and secreted to apical compartments for long-range diffusion, and (ii) lipid rafts organized by reggies/flotillins serve as "dating points" where extracellular Wnt transiently interacts with lipoprotein receptors to allow its capture and further spreading via lipoprotein particles. We further discuss these processes in the context of human breast cancer. A better understanding of these phenomena may be relevant for identification of novel drug targets and therapeutic strategies.

8.
Neurobiol Dis ; 51: 168-76, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23174179

RESUMEN

The ability of fish retinal ganglion cells (RGCs) to regenerate their axons was shown to require the re-expression and function of the two proteins reggie-1 and -2. RGCs in mammals fail to upregulate reggie expression and to regenerate axons after lesion suggesting the possibility that induced upregulation might promote regeneration. In the present study, RGCs in adult rats were induced to express reggie-1 by intravitreal injection of adeno-associated viral vectors (AAV2/1) expressing reggie-1 (AAV.R1-EGFP) 14d prior to optic nerve crush. Four weeks later, GAP-43-positive regenerating axons had crossed the lesion and grown into the nerve at significantly higher numbers and length (up to 5mm) than the control transduced with AAV.EGFP. Consistently, after transduction with AAV.R1-EGFP as opposed to AAV.EGFP, primary RGCs in vitro grew long axons on chondroitin sulfate proteoglycan (CSPG) and Nogo-A, both glial cell-derived inhibitors of neurite growth, suggesting that reggie-1 can provide neurons with the ability to override inhibitors of neurite growth. This reggie-1-mediated enhancement of growth was reproduced in mouse hippocampal and N2a neurons which generated axons 40-60% longer than their control counterparts. This correlates with the reggie-1-dependent activation of Src and PI3 kinase (PI3K), of the Rho family GTPase Rac1 and downstream effectors such as cofilin. This increased growth also depends on TC10, the GTPase involved in cargo delivery to the growth cone. Thus, the upregulation of reggie-1 in mammalian neurons provides nerve cells with neuron-intrinsic properties required for axon growth and successful regeneration in the adult mammalian CNS.


Asunto(s)
Axones/metabolismo , Proteínas de la Membrana/biosíntesis , Regeneración Nerviosa/fisiología , Neuritas/metabolismo , Nervio Óptico/metabolismo , Animales , Western Blotting , Ratones , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Transducción Genética , Regulación hacia Arriba
9.
Mol Biol Cell ; 23(10): 1812-25, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22438585

RESUMEN

The reggie/flotillin proteins are implicated in membrane trafficking and, together with the cellular prion protein (PrP), in the recruitment of E-cadherin to cell contact sites. Here, we demonstrate that reggies, as well as PrP down-regulation, in epithelial A431 cells cause overlapping processes and abnormal formation of adherens junctions (AJs). This defect in cell adhesion results from reggie effects on Src tyrosine kinases and epidermal growth factor receptor (EGFR): loss of reggies reduces Src activation and EGFR phosphorylation at residues targeted by Src and c-cbl and leads to increased surface exposure of EGFR by blocking its internalization. The prolonged EGFR signaling at the plasma membrane enhances cell motility and macropinocytosis, by which junction-associated E-cadherin is internalized and recycled back to AJs. Accordingly, blockage of EGFR signaling or macropinocytosis in reggie-deficient cells restores normal AJ formation. Thus, by promoting EGFR internalization, reggies restrict the EGFR signaling involved in E-cadherin macropinocytosis and recycling and regulate AJ formation and dynamics and thereby cell adhesion.


Asunto(s)
Uniones Adherentes/metabolismo , Cadherinas/metabolismo , Receptores ErbB/metabolismo , Proteínas de la Membrana/metabolismo , Uniones Adherentes/ultraestructura , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Endocitosis , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/genética , Fosfoproteínas/metabolismo , Fosforilación , Priones/genética , Priones/metabolismo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Interferencia de ARN , Transducción de Señal , beta Catenina/metabolismo
10.
J Neurosci ; 31(49): 18013-25, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22159115

RESUMEN

The role of prion protein (PrP) is insufficiently understood partially because PrP-deficient (-/-) neurons from C57BL/6J mice seem to differentiate normally and are functionally mildly impaired. Here, we reassessed this notion and, unexpectedly, discovered that PrP(-/-) hippocampal growth cones were abnormally small and poor in filopodia and cargo-containing vesicles. Based on our findings that PrP-PrP trans-interaction recruits E-cadherin to cell contact sites and reggie microdomains, and that reggies are essential for growth by regulating membrane trafficking, we reasoned that PrP and reggie might promote cargo (N-cadherin) delivery via PrP-reggie-connected signaling upon PrP activation (by PrP-Fc-induced trans-interaction). In wild-type but not PrP(-/-) neurons, PrP activation led to (1) enhanced PrP-reggie cocluster formation, (2) reggie-associated fyn and MAP kinase activation, (3) Exo70 and N-cadherin (cargo) recruitment to reggie, (4) the preference of the growth cone for PrP-Fc as substrate, and (5) longer neurites. Conversely, PrP-reggie-induced N-cadherin recruitment was blocked by mutant TC10, the GTPase downstream of reggie, triggering exocyst-assisted cargo delivery. This implies that PrP functions in reggie-mediated signaling and cargo trafficking, thus promoting growth cone complexity and vitality and thereby growth cone elongation.


Asunto(s)
Cadherinas/metabolismo , Conos de Crecimiento/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Neuronas/citología , Priones/farmacología , Análisis de Varianza , Animales , Animales Recién Nacidos , Células Cultivadas , Exocitosis/efectos de los fármacos , Exocitosis/genética , Hipocampo/citología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuritas/efectos de los fármacos , Neuritas/fisiología , Péptidos/farmacología , Priones/genética , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transfección/métodos , Proteínas de Transporte Vesicular/metabolismo
11.
J Neurosci ; 29(20): 6607-15, 2009 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-19458231

RESUMEN

The reggies/flotillins--proteins upregulated during axon regeneration in retinal ganglion cells (RGCs)--are scaffolding proteins of microdomains and involved in neuronal differentiation. Here, we show that reggies regulate axon regeneration in zebrafish (ZF) after optic nerve section (ONS) in vivo as well as axon/neurite extension in hippocampal and N2a neurons in vitro through signal transduction molecules modulating actin dynamics. ZF reggie-1a, -2a, and -2b downregulation by reggie-specific morpholino (Mo) antisense oligonucleotides directly after ONS significantly reduced ZF RGC axon regeneration: RGC axons from reggie Mo retinas were markedly reduced. Moreover, the number of axon-regenerating RGCs, identified by insertion of A488-coupled dextran, decreased by 69% in retinas 7 d after Mo application. At 10 and 14 d, RGCs decreased by 53 and 33%, respectively, in correlation with the gradual inactivation of the Mos. siRNA-mediated knockdown of reggie-1 and -2 inhibited the differentiation and axon/neurite extension in hippocampal and N2a neurons. N2a cells had significantly shorter filopodia, more cells had lamellipodia and fewer neurites, defects which were rescued by a reggie-1 construct without siRNA-binding sites. Furthermore, reggie knockdown strongly perturbed the balanced activation of the Rho family GTPases Rac1, RhoA, and cdc42, influenced the phosphorylation of cortactin and cofilin, the formation of the N-WASP, cortactin and Arp3 complex, and affected p38, Ras, ERK1/2 (extracellular signal-regulated kinases 1 and 2), and focal adhesion kinase activation. Thus, as suggested by their prominent re-expression after lesion, the reggies represent neuron-intrinsic factors for axon outgrowth and regeneration, being crucial for the coordinated assembly of signaling complexes regulating cytoskeletal remodeling.


Asunto(s)
Diferenciación Celular/fisiología , Hipocampo/citología , Proteínas de la Membrana/metabolismo , Regeneración Nerviosa/fisiología , Neuronas/fisiología , Traumatismos del Nervio Óptico/fisiopatología , Retina/patología , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Dextranos , Regulación hacia Abajo/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Inmunoprecipitación , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas de la Membrana/genética , Ratones , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/genética , Neuroblastoma , Neuronas/efectos de los fármacos , Oligodesoxirribonucleótidos Antisentido/farmacología , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/patología , Soluciones Preservantes de Órganos , ARN Interferente Pequeño/metabolismo , Retina/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transfección/métodos , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Pez Cebra , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo
12.
EMBO J ; 27(3): 509-21, 2008 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-18219274

RESUMEN

The lipid-modified morphogens Wnt and Hedgehog diffuse poorly in isolation yet can spread over long distances in vivo, predicting existence of two distinct forms of these morphogens. The first is poorly mobile and activates short-range target genes. The second is specifically packed for efficient spreading to induce long-range targets. Subcellular mechanisms involved in the discriminative secretion of these two forms remain elusive. Wnt and Hedgehog can associate with membrane microdomains, but the function of this association was unknown. Here we show that a major protein component of membrane microdomains, reggie-1/flotillin-2, plays important roles in secretion and spreading of Wnt and Hedgehog in Drosophila. Reggie-1 loss-of-function results in reduced spreading of the morphogens, while its overexpression stimulates secretion of Wnt and Hedgehog and expands their diffusion. The resulting changes in the morphogen gradients differently affect the short- and long-range targets. In its action reggie-1 appears specific for Wnt and Hedgehog. These data suggest that reggie-1 is an important component of the Wnt and Hedgehog secretion pathway dedicated to formation of the mobile pool of these morphogens.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Proteínas Hedgehog/metabolismo , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular , Microdominios de Membrana/química , Microdominios de Membrana/genética , Microdominios de Membrana/metabolismo , Microdominios de Membrana/fisiología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Transducción de Señal/genética , Alas de Animales/fisiología , Proteína Wnt1
13.
FEBS Lett ; 581(24): 4697-703, 2007 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-17854803

RESUMEN

The reggies/flotillins are oligomeric scaffolding proteins for membrane microdomains. We show here that reggie-1/flotillin-2 microdomains are organized along cortical F-actin in several cell types. Interaction with F-actin is mediated by the SPFH domain as shown by in vivo co-localization and in vitro binding experiments. Reggie-1/flotillin-2 microdomains form independent of actin, but disruption or stabilization of the actin cytoskeleton modulate the lateral mobility of reggie-1/flotillin-2 as shown by FRAP. Furthermore, reggie/flotillin microdomains can efficiently be immobilized by actin polymerisation, while exchange of reggie-1/flotillin-2 molecules between microdomains is enhanced by actin disruption as shown by tracking of individual microdomains using TIRF microscopy.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular Tumoral , Humanos , Proteínas de la Membrana/genética , Ratones , Unión Proteica , Transporte de Proteínas
14.
Biochem J ; 403(2): 313-22, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17206938

RESUMEN

Reggie-1 and -2 proteins (flotillin-2 and -1 respectively) form their own type of non-caveolar membrane microdomains, which are involved in important cellular processes such as T-cell activation, phagocytosis and signalling mediated by the cellular prion protein and insulin; this is consistent with the notion that reggie microdomains promote protein assemblies and signalling. While it is generally known that membrane microdomains contain large multiprotein assemblies, the exact organization of reggie microdomains remains elusive. Using chemical cross-linking approaches, we have demonstrated that reggie complexes are composed of homo- and hetero-tetramers of reggie-1 and -2. Moreover, native reggie oligomers are indeed quite stable, since non-cross-linked tetramers are resistant to 8 M urea treatment. We also show that oligomerization requires the C-terminal but not the N-terminal halves of reggie-1 and -2. Using deletion constructs, we analysed the functional relevance of the three predicted coiled-coil stretches present in the C-terminus of reggie-1. We confirmed experimentally that reggie-1 tetramerization is dependent on the presence of coiled-coil 2 and, partially, of coiled-coil 1. Furthermore, since depletion of reggie-1 by siRNA (small interfering RNA) silencing induces proteasomal degradation of reggie-2, we conclude that the protein stability of reggie-2 depends on the presence of reggie-1. Our data indicate that the basic structural units of reggie microdomains are reggie homo- and hetero-tetramers, which are dependent on the presence of reggie-1.


Asunto(s)
Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Línea Celular Tumoral , Reactivos de Enlaces Cruzados , Eliminación de Gen , Genes Reporteros/genética , Proteínas de la Membrana/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , ARN Interferente Pequeño/genética , Ratas , Succinimidas
15.
Exp Cell Res ; 307(1): 100-8, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15922730

RESUMEN

The giant protein hFat1, a member of the cadherin superfamily, has been proposed to play roles in cerebral development, glomerular slit formation, and also to act as a tumor suppressor, but its mechanisms of action have not been elucidated. To examine functions of the transmembrane and cytoplasmic domains, they were expressed in HEK293 and HeLa cells as chimeric proteins in fusion with EGFP and extracellular domains derived from E-cadherin. Proteins comprising the transmembrane domain localized to the membrane fraction. Deletion of this domain resulted in a predominantly nuclear localization of the cytoplasmic segment of hFat1. Nuclear localization was largely reduced by deletion of a presumed juxta-membrane NLS. Fusion proteins located in the plasma membrane underwent proteolytic processing. In a first proteolytic step, only the extracellular domain was cleaved off. In another step, the cleavage product was released to the cytosol and was also found in a low speed pellet fraction, in accordance with the nuclear localization of the cytoplasmic domain of hFat1.


Asunto(s)
Cadherinas/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Procesamiento Proteico-Postraduccional , Western Blotting , Cadherinas/química , Cadherinas/genética , Fraccionamiento Celular , Línea Celular , Citosol/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Hidrólisis , Microscopía Confocal , Señales de Localización Nuclear , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Fracciones Subcelulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA