Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
PLoS Genet ; 12(6): e1006116, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27362352

RESUMEN

Biological membranes have been proposed to contain microdomains of a specific lipid composition, in which distinct groups of proteins are clustered. Flotillin-like proteins are conserved between pro-and eukaryotes, play an important function in several eukaryotic and bacterial cells, and define in vertebrates a type of so-called detergent-resistant microdomains. Using STED microscopy, we show that two bacterial flotillins, FloA and FloT, form defined assemblies with an average diameter of 85 to 110 nm in the model bacterium Bacillus subtilis. Interestingly, flotillin microdomains are of similar size in eukaryotic cells. The soluble domains of FloA form higher order oligomers of up to several hundred kDa in vitro, showing that like eukaryotic flotillins, bacterial assemblies are based in part on their ability to self-oligomerize. However, B. subtilis paralogs show significantly different diffusion rates, and consequently do not colocalize into a common microdomain. Dual colour time lapse experiments of flotillins together with other detergent-resistant proteins in bacteria show that proteins colocalize for no longer than a few hundred milliseconds, and do not move together. Our data reveal that the bacterial membrane contains defined-sized protein domains rather than functional microdomains dependent on flotillins. Based on their distinct dynamics, FloA and FloT confer spatially distinguishable activities, but do not serve as molecular scaffolds.


Asunto(s)
Membrana Celular/metabolismo , Detergentes/metabolismo , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Bacillus subtilis/metabolismo , Microscopía Fluorescente/métodos , Transporte de Proteínas/fisiología
2.
Dev Dyn ; 246(1): 41-49, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27666728

RESUMEN

BACKGROUND: The conditional Cre/lox system has recently emerged as a valuable tool for studies on both embryonic and adult Zebrafish. Temporal control and site-specific recombination are achieved by using the ligand-inducible CreERT2 and administration of the drug tamoxifen (TAM) or its active metabolite, 4-Hydroxytamoxifen (4-OHT). RESULTS: Here we report the generation of a transgenic Zebrafish line, which expresses an mCherry-tagged variant of CreERT2 under the control of the myelin basic protein a (mbpa) promoter. Our analysis shows that larval and adult expression of the transgene recapitulates the endogenous mbpa expression pattern in oligodendrocytes. Furthermore, combination with a Cre-dependent EGFP reporter results in EGFP-expressing oligodendrocytes in the spinal cord, brain, and optic nerve in TAM- or 4-OHT-treated larvae and 4-month-old fish, but not in untreated controls. CONCLUSIONS: The transgenic Zebrafish line Tg(mbpa:mCherry-T2A-CreERT2 ) elicits CreERT2 expression specifically in myelinating glia cells. Cre-inducible targeted recombination of genes in oligodendrocytes will be useful to elucidate cellular and molecular mechanisms of myelination in vivo during development (myelination) and regeneration (remyelination) after injury to the central nervous system (CNS). It will also allow targeted expression and overexpression of genes of interest (transgenes) in oligodendrocytes at defined developmental and adult stages. Developmental Dynamics 246:41-49, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Integrasas/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Enfermedades Desmielinizantes , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteína Básica de Mielina/genética , Oligodendroglía/ultraestructura , Regiones Promotoras Genéticas , Recombinación Genética , Transgenes , Pez Cebra/metabolismo
3.
Eukaryot Cell ; 12(4): 529-44, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23376944

RESUMEN

The SPFH protein superfamily is assumed to occur universally in eukaryotes, but information from protozoa is scarce. In the Paramecium genome, we found only Stomatins, 20 paralogs grouped in 8 families, STO1 to STO8. According to cDNA analysis, all are expressed, and molecular modeling shows the typical SPFH domain structure for all subgroups. For further analysis we used family-specific sequences for fluorescence and immunogold labeling, gene silencing, and functional tests. With all family members tested, we found a patchy localization at/near the cell surface and on vesicles. The Sto1p and Sto4p families are also associated with the contractile vacuole complex. Sto4p also makes puncta on some food vacuoles and is abundant on vesicles recycling from the release site of spent food vacuoles to the site of nascent food vacuole formation. Silencing of the STO1 family reduces mechanosensitivity (ciliary reversal upon touching an obstacle), thus suggesting relevance for positioning of mechanosensitive channels in the plasmalemma. Silencing of STO4 members increases pulsation frequency of the contractile vacuole complex and reduces phagocytotic activity of Paramecium cells. In summary, Sto1p and Sto4p members seem to be involved in positioning specific superficial and intracellular microdomain-based membrane components whose functions may depend on mechanosensation (extracellular stimuli and internal osmotic pressure).


Asunto(s)
Membrana Celular/fisiología , Genoma de Protozoos , Microdominios de Membrana/fisiología , Proteínas de la Membrana/metabolismo , Paramecium tetraurelia/fisiología , Vesículas Transportadoras/fisiología , Membrana Celular/química , Regulación de la Expresión Génica , Silenciador del Gen , Mecanotransducción Celular/fisiología , Microdominios de Membrana/química , Proteínas de la Membrana/genética , Familia de Multigenes , Paramecium tetraurelia/química , Fagocitosis/fisiología , Fagosomas/química , Fagosomas/fisiología , Estructura Terciaria de Proteína , Vesículas Transportadoras/química , Vacuolas/química , Vacuolas/fisiología
4.
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
5.
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
6.
Biochim Biophys Acta ; 1812(3): 415-22, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21147218

RESUMEN

The two proteins reggie-1 and reggie-2 (flotillins) were identified in axon-regenerating neurons in the central nervous system and shown to be essential for neurite growth and regeneration in fish and mammals. Reggies/flotillins are microdomain scaffolding proteins sharing biochemical properties with lipid raft molecules, form clusters at the cytoplasmic face of the plasma membrane and interact with signaling molecules in a cell type specific manner. In this review, reggie microdomains, lipid rafts, related scaffolding proteins and caveolin-which, however, are responsible for their own microdomains and functions-are introduced. Moreover, the function of the reggies in axon growth is demonstrated: neurons fail to extend axons after reggie knockdown. Furthermore, our current concept of the molecular mechanism underlying reggie function is presented: the association of glycosyl-phophatidyl inositol (GPJ)-anchored surface proteins with reggie microdomains elicits signals which activate src tyrosine and mitogen-activated protein kinases, as well as small guanosine 5'-triphosphate-hydrolyzing enzymes. This leads to the mobilization of intracellular vesicles and to the recruitment of bulk membrane and specific cargo proteins, such as cadherin, to specific sites of the plasma membrane such as the growth cone of elongating axons. Thus, reggies regulate the targeted delivery of cargo-a process which is required for process extension and growth. This article is part of a Special Issue entitled Zebrafish Models of Neurological Diseases.


Asunto(s)
Axones/fisiología , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Regeneración Nerviosa , Pez Cebra/metabolismo , Animales , Diferenciación Celular , Pez Cebra/embriología
7.
Mol Biol Evol ; 28(4): 1363-70, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21098000

RESUMEN

Unlike mammals, fish are able to regenerate axons in their central nervous system. This difference has been partly attributed to the loss/acquisition of inhibitory proteins during evolution. Nogo-A--the longest isoform of the reticulon4 (rtn4) gene product--is commonly found in mammalian myelin where it acts as a potent inhibitor of axonal regeneration. Interestingly, fish RTN4 isoforms were previously reported to lack the most inhibitory Nogo-A-specific region (NSR). Nevertheless, fish axons collapse on contact with mammalian NSR, suggesting that fish possess a functional Nogo-A receptor but not its ligand. To reconcile these findings, we revisited the early evolution of rtn4. Mining of current genome databases established the unequivocal presence of NSR-coding sequences in fish rtn4 paralogues. Further comparative analyses indicate that the common ancestor of fish and tetrapods had an NSR-coding rtn4 gene, which underwent duplication and divergent evolution in bony fish. Our genomic survey also revealed that the cephalochordate Branchiostoma floridae contains a single rtn gene lacking the NSR. Hence, Nogo-A most probably arose independently in the rtn4 gene of a gnathostome ancestor before the split of the fish and tetrapod lineages. Close examination of the NSR uncovered clusters of structural and sequential similarities with neurocan (NCAN), an inhibitory proteoglycan of the glial scar. Notably, the shared presence of transposable elements in ncan and rtn4 genes suggests that Nogo-A originated via insertion of an ncan-like sequence into the rtn4 gene of an early jawed vertebrate with myelinated axons.


Asunto(s)
Evolución Biológica , Maxilares , Proteínas de la Mielina/genética , Isoformas de Proteínas/genética , Vertebrados/genética , Secuencia de Aminoácidos , Animales , Axones/fisiología , Peces/genética , Humanos , Datos de Secuencia Molecular , Proteínas Nogo , Filogenia , Alineación de Secuencia , Vertebrados/clasificación
8.
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
9.
Cell Tissue Res ; 349(1): 71-7, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22350847

RESUMEN

The microdomain-forming proteins reggie-1 and reggie-2 (alias flotillins) were found to be upregulated in axon-regenerating fish retinal ganglion cells (RGCs). They were subsequently shown to be indispensible for axon regeneration and neurite extension in fish and mammals. Our current concept proposes that reggies--often together with the cellular Prion protein (PrP)--regulate the turnover of membrane and specific membrane proteins at the growth cone, which is the prerequisite for neurite elongation and guidance.


Asunto(s)
Axones/fisiología , Proteínas de la Membrana/metabolismo , Regeneración Nerviosa/fisiología , Animales , Humanos , Lisosomas/metabolismo , Células Ganglionares de la Retina/metabolismo , Vesículas Transportadoras/metabolismo
10.
PLoS Biol ; 7(3): e55, 2009 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-19278297

RESUMEN

Prion proteins (PrPs) are key players in fatal neurodegenerative disorders, yet their physiological functions remain unclear, as PrP knockout mice develop rather normally. We report a strong PrP loss-of-function phenotype in zebrafish embryos, characterized by the loss of embryonic cell adhesion and arrested gastrulation. Zebrafish and mouse PrP mRNAs can partially rescue this knockdown phenotype, indicating conserved PrP functions. Using zebrafish, mouse, and Drosophila cells, we show that PrP: (1) mediates Ca(+2)-independent homophilic cell adhesion and signaling; and (2) modulates Ca(+2)-dependent cell adhesion by regulating the delivery of E-cadherin to the plasma membrane. In vivo time-lapse analyses reveal that the arrested gastrulation in PrP knockdown embryos is due to deficient morphogenetic cell movements, which rely on E-cadherin-based adhesion. Cell-transplantation experiments indicate that the regulation of embryonic cell adhesion by PrP is cell-autonomous. Moreover, we find that the local accumulation of PrP at cell contact sites is concomitant with the activation of Src-related kinases, the recruitment of reggie/flotillin microdomains, and the reorganization of the actin cytoskeleton, consistent with a role of PrP in the modulation of cell adhesion via signaling. Altogether, our data uncover evolutionarily conserved roles of PrP in cell communication, which ultimately impinge on the stability of adherens cell junctions during embryonic development.


Asunto(s)
Adhesión Celular/fisiología , Agregación Celular/fisiología , Priones/fisiología , Transducción de Señal/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Actinas/fisiología , Animales , Cadherinas/fisiología , Membrana Celular/fisiología , Movimiento Celular/fisiología , Citoesqueleto/fisiología , Drosophila/genética , Gastrulación/fisiología , Expresión Génica , Proteínas de la Membrana/fisiología , Ratones/genética , Priones/genética , Uniones Estrechas/fisiología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Familia-src Quinasas/fisiología
11.
J Neurochem ; 116(5): 708-13, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21214550

RESUMEN

The two proteins reggie-1/flotillin-2 and reggie-2/flotillin-1 form microdomains at the plasma membrane and at intracellular compartments where src tyrosine kinases associate with them. Specific GPI-anchored proteins, in particular prion protein and Thy-1, co-cluster with reggie microdomains at the plasma membrane and elicit signal transduction in association with reggies which regulates the activation of several GTPases involved in the recruitment of specific membrane proteins from intracellular carriers to target sites of the cell membrane in a cell type-specific manner. For example, prion protein and reggie regulate the recruitment and targeted delivery of the T cell receptor complex to the T cell cap, of E-cadherin to cell-cell contact sites in epithelial cells, and of bulk membrane and growth receptors to the growth cone in developing neurons. Evidence is accumulating that reggies are involved in guiding the cell-type-specific membrane proteins from the intracellular compartments to their target sites at the cell membrane, a function required in all cells which explains why reggies are expressed in many or all cells in invertebrates and vertebrates.


Asunto(s)
Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Axones/fisiología , Cadherinas/metabolismo , Humanos , Modelos Biológicos , Neuronas/citología , Priones/fisiología , Transducción de Señal/fisiología , Antígenos Thy-1/fisiología , Familia-src Quinasas/metabolismo
12.
J Neurosci ; 29(49): 15489-98, 2009 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-20007473

RESUMEN

In contrast to mammals, lesioned axons in the zebrafish (ZF) optic nerve regenerate and restore vision. This correlates with the absence of the NogoA-specific N-terminal domains from the ZF nogo/rtn-4 (reticulon-4) gene that inhibits regeneration in mammals. However, mammalian nogo/rtn-4 carries a second inhibitory C-terminal domain, Nogo-66, being 70% identical with ZF-Nogo66. The present study examines, (1) whether ZF-Nogo66 is inhibitory and effecting similar signaling pathways upon Nogo66-binding to the Nogo66 receptor NgR and its coreceptors, and (2) whether Rat-Nogo66 on fish, and ZF-Nogo66 on mouse neurons, cause inhibition via NgR. Our results from "outgrowth, collapse and contact assays" suggest, surprisingly, that ZF-Nogo66 is growth-permissive for ZF and mouse neurons, quite in contrast to its Rat-Nogo66 homolog which inhibits growth. The opposite effects of ZF- and Rat-Nogo66 are, in both fish and mouse, transmitted by GPI (glycosylphosphatidylinositol)-anchored receptors, including NgR. The high degree of sequence homology in the predicted binding site is consistent with the ability of ZF- and mammalian-Nogo66 to bind to NgRs of both species. Yet, Rat-Nogo66 elicits phosphorylation of the downstream effector cofilin whereas ZF-Nogo66 has no influence on cofilin phosphorylation--probably because of significantly different Rat- versus ZF-Nogo66 sequences outside of the receptor-binding region effecting, by speculation, recruitment of a different set of coreceptors or microdomain association of NgR. Thus, not only was the NogoA-specific domain lost in fish, but Nogo66, the second inhibitory domain in mammals, and its signaling upon binding to NgR, was modified so that ZF-Nogo/RTN-4 does not impair axon regeneration.


Asunto(s)
Axones/fisiología , Proteínas de la Mielina/metabolismo , Regeneración Nerviosa/fisiología , Nervio Óptico/fisiología , Receptores de Superficie Celular/metabolismo , Proteínas de Pez Cebra/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Animales , Glicosilfosfatidilinositoles/metabolismo , Conos de Crecimiento/fisiología , Células HeLa , Hipocampo/fisiología , Humanos , Técnicas In Vitro , Ratones , Proteínas de la Mielina/genética , Neuritas/fisiología , Neuronas/fisiología , Proteínas Nogo , Ratas , Retina/fisiología , Células Ganglionares de la Retina/fisiología , Transducción de Señal , Especificidad de la Especie , Pez Cebra
13.
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
14.
BMC Evol Biol ; 9: 10, 2009 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-19138386

RESUMEN

BACKGROUND: The SPFH protein superfamily is a diverse family of proteins whose eukaryotic members are involved in the scaffolding of detergent-resistant microdomains. Recently the origin of the SPFH proteins has been questioned. Instead, convergent evolution has been proposed. However, an independent, convergent evolution of three large prokaryotic and three eukaryotic families is highly unlikely, especially when other mechanisms such as lateral gene transfer which could also explain their distribution pattern have not yet been considered.To gain better insight into this very diverse protein family, we have analyzed the genomes of 497 microorganisms and investigated the pattern of occurrence as well as the genomic vicinity of the prokaryotic SPFH members. RESULTS: According to sequence and operon structure, a clear division into 12 subfamilies was evident. Three subfamilies (SPFH1, SPFH2 and SPFH5) show a conserved operon structure and two additional subfamilies are linked to those three through functional aspects (SPFH1, SPFH3, SPFH4: interaction with FtsH protease). Therefore these subgroups most likely share common ancestry. The complex pattern of occurrence among the different phyla is indicative of lateral gene transfer. Organisms that do not possess a single SPFH protein are almost exclusively endosymbionts or endoparasites. CONCLUSION: The conserved operon structure and functional similarities suggest that at least 5 subfamilies that encompass almost 75% of all prokaryotic SPFH members share a common origin. Their similarity to the different eukaryotic SPFH families, as well as functional similarities, suggests that the eukaryotic SPFH families originated from different prokaryotic SPFH families rather than one. This explains the difficulties in obtaining a consistent phylogenetic tree of the eukaryotic SPFH members. Phylogenetic evidence points towards lateral gene transfer as one source of the very diverse patterns of occurrence in bacterial species.


Asunto(s)
Proteínas Bacterianas/genética , Evolución Molecular , Proteínas de la Membrana/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Secuencia Conservada , Bases de Datos de Proteínas , Transferencia de Gen Horizontal , Genoma Bacteriano , Proteínas de la Membrana/química , Datos de Secuencia Molecular , Operón , Filogenia , Estructura Terciaria de Proteína , Alineación de Secuencia , Análisis de Secuencia de Proteína
15.
Eur J Cell Biol ; 87(4): 211-26, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18237819

RESUMEN

The reggie/flotillin proteins oligomerize and associate into clusters which form scaffolds for membrane microdomains. Besides their localization at the plasma membrane, the reggies/flotillins reside at various intracellular compartments; however, the trafficking pathways used by reggie-1/flotillin-2 remain unclear. Here, we show that trafficking of reggie-1/flotillin-2 is BFA sensitive and that deletion mutants of reggie-1/flotillin-2 accumulate in the Golgi complex in HeLa, Jurkat and PC12 cells, suggesting Golgi-dependent trafficking of reggie-1/flotillin-2. Using total internal reflection fluorescence microscopy, we observed fast cycling of reggie-1/flotillin-2-positive vesicles at the plasma membrane, which engaged in transient interactions with the plasma membrane only. Reggie-1/flotillin-2 cycling was independent of clathrin, but was inhibited by cholesterol depletion and microtubule disruption. Cycling of reggie-1/flotillin-2 was negatively correlated with cell-cell contact formation but was stimulated by serum, epidermal growth factor and by cholesterol loading mediated by low density lipoproteins. However, reggie-1/flotillin-2 was neither involved in endocytosis of the epidermal growth factor itself nor in endocytosis of GPI-GFPs or the GPI-anchored cellular prion protein (PrP(c)). Reggie-2/flotillin-1 and stomatin-1 also exhibited cycling at the plasma membrane similar to reggie-1/flotillin-2, but these vesicles and microdomains only partially co-localized with reggie-2/flotillin-1. Thus, regulated vesicular cycling might be a general feature of SPFH protein-dependent trafficking.


Asunto(s)
Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Colesterol/metabolismo , Endocitosis , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Células HeLa , Humanos , Células Jurkat , Microdominios de Membrana/química , Microdominios de Membrana/ultraestructura , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Microscopía Fluorescente , Microtúbulos/metabolismo , Mutación , Proteínas del Tejido Nervioso , Células PC12 , Priones/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas
16.
Eur J Cell Biol ; 87(12): 921-31, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18722032

RESUMEN

The reggies/flotillins were discovered as proteins upregulated during axon regeneration. Here, we show that expression of a trans-negative reggie-1/flotillin-2 deletion mutant, R1EA, which interferes with oligomerization of the reggies/flotillins, inhibited insulin-like growth factor (IGF)-induced neurite outgrowth in N2a neuroblastoma cells and impaired in vitro differentiation of primary rat hippocampal neurons. Cells expressing R1EA formed only short and broad membrane protrusions often with abnormally large growth cones. R1EA expression strongly perturbed the balanced activation of the Rho-family GTPases Rac1 and cdc42. Furthermore, focal adhesion kinase (FAK) activity was also enhanced by R1EA expression, while other signaling pathways like ERK1/2, PKC or PKB signaling were unaffected. These severe signaling defects were caused by an impaired recruitment of the reggie/flotillin-associated adaptor molecule CAP/ponsin to focal contacts at the plasma membrane. Thus, the reggies/flotillins are crucial for coordinated assembly of signaling complexes regulating cytoskeletal remodeling.


Asunto(s)
Citoesqueleto/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Microfilamentos/metabolismo , Neuronas/citología , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Western Blotting , Diferenciación Celular , Membrana Celular/metabolismo , Células Cultivadas , Técnica del Anticuerpo Fluorescente Indirecta , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Eliminación de Gen , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutación/genética , Neuritas/fisiología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neuronas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Wistar
17.
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
18.
Trends Genet ; 19(2): 72-5, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12547513

RESUMEN

Infectious prion proteins cause neurodegenerative disease in mammals owing to the acquisition of an aberrant conformation. We cloned a Fugu rubripes gene that encodes a structurally conserved prion protein, and found rapid rates of molecular divergence among prions from different vertebrate classes, along with molecular stasis within each class. We propose that a directional trend in the evolution of prion sequence motifs associated with pathogenesis and infectivity could account for the origin of scrapie in mammals.


Asunto(s)
Evolución Molecular , Peces , Priones/química , ARN Mensajero/genética , Scrapie/genética , Takifugu/genética , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Disulfuros , Humanos , Filogenia , Proteínas PrPC/patogenicidad , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
19.
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
20.
FASEB J ; 20(6): 711-3, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16452278

RESUMEN

T cell activation after contact with an antigen-presenting cell depends on the regulated assembly of the T cell receptor signaling complex, which involves the polarized assembly of a stable, raft-like macrodomain surrounding engaged T cell receptors. Here we show that the preformed reggie/flotillin caps present in resting T cells act as priming platforms for macrodomain assembly. Preformed reggie-1/flotillin-2 caps are exceptionally stable, as shown by fluorescence recovery after photobleaching (FRAP). Upon T cell stimulation, signaling molecules are recruited to the stable reggie/flotillin caps. Importantly, a trans-negative reggie-1/flotillin-2 deletion mutant, which interferes with assembly of the preformed reggie/flotillin cap, impairs raft polarization and macrodomain formation after T cell activation. Accordingly, expression of the trans-negative reggie-1 mutant leads to the incorrect positioning of the guanine nucleotide exchange factor Vav, resulting in defects in cytoskeletal reorganization. Thus, the preformed reggie/flotillin caps are stable priming platforms for the assembly of multiprotein complexes controlling actin reorganization during T cell activation.


Asunto(s)
Proteínas de la Membrana/metabolismo , Linfocitos T/metabolismo , Actinas/metabolismo , Animales , Citoesqueleto/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Humanos , Células Jurkat , Activación de Linfocitos , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/genética , Mutación , Células PC12 , Ratas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA