Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cancer Res ; 82(21): 3932-3949, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36054547

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest malignancies and potentially curable only with radical surgical resection at early stages. The tumor microenvironment has been shown to be central to the development and progression of PDAC. A better understanding of how early human PDAC metabolically communicates with its environment and differs from healthy pancreas could help improve PDAC diagnosis and treatment. Here we performed deep proteomic analyses from diagnostic specimens of operable, treatment-naïve PDAC patients (n = 14), isolating four tissue compartments by laser-capture microdissection: PDAC lesions, tumor-adjacent but morphologically benign exocrine glands, and connective tissues neighboring each of these compartments. Protein and pathway levels were compared between compartments and with control pancreatic proteomes. Selected targets were studied immunohistochemically in the 14 patients and in additional tumor microarrays, and lipid deposition was assessed by nonlinear label-free imaging (n = 16). Widespread downregulation of pancreatic secretory functions was observed, which was paralleled by high cholesterol biosynthetic activity without prominent lipid storage in the neoplastic cells. Stromal compartments harbored ample blood apolipoproteins, indicating abundant microvasculature at the time of tumor removal. The features best differentiating the tumor-adjacent exocrine tissue from healthy control pancreas were defined by upregulation of proteins related to lipid transport. Importantly, histologically benign exocrine regions harbored the most significant prognostic pathways, with proteins involved in lipid transport and metabolism, such as neutral cholesteryl ester hydrolase 1, associating with shorter survival. In conclusion, this study reveals prognostic molecular changes in the exocrine tissue neighboring pancreatic cancer and identifies enhanced lipid transport and metabolism as its defining features. SIGNIFICANCE: In clinically operable pancreatic cancer, regions distant from malignant cells already display proteomic changes related to lipid transport and metabolism that affect prognosis and may be pharmacologically targeted.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Proteómica , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/patología , Lípidos , Biomarcadores de Tumor/metabolismo , Microambiente Tumoral , Neoplasias Pancreáticas
2.
J Cell Sci ; 130(5): 892-902, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28096473

RESUMEN

The actin and intermediate filament cytoskeletons contribute to numerous cellular processes, including morphogenesis, cytokinesis and migration. These two cytoskeletal systems associate with each other, but the underlying mechanisms of this interaction are incompletely understood. Here, we show that inactivation of vimentin leads to increased actin stress fiber assembly and contractility, and consequent elevation of myosin light chain phosphorylation and stabilization of tropomyosin-4.2 (see Geeves et al., 2015). The vimentin-knockout phenotypes can be rescued by re-expression of wild-type vimentin, but not by the non-filamentous 'unit length form' vimentin, demonstrating that intact vimentin intermediate filaments are required to facilitate the effects on the actin cytoskeleton. Finally, we provide evidence that the effects of vimentin on stress fibers are mediated by activation of RhoA through its guanine nucleotide exchange factor GEF-H1 (also known as ARHGEF2). Vimentin depletion induces phosphorylation of the microtubule-associated GEF-H1 on Ser886, and thereby promotes RhoA activity and actin stress fiber assembly. Taken together, these data reveal a new mechanism by which intermediate filaments regulate contractile actomyosin bundles, and may explain why elevated vimentin expression levels correlate with increased migration and invasion of cancer cells.


Asunto(s)
Actinas/metabolismo , Filamentos Intermedios/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Fibras de Estrés/metabolismo , Vimentina/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Línea Celular Tumoral , Fibroblastos/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Fosforilación
3.
Neurobiol Dis ; 96: 335-345, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27425888

RESUMEN

In Parkinson's disease midbrain dopaminergic neurons degenerate and die. Oral medications and deep brain stimulation can relieve the initial symptoms, but the disease continues to progress. Growth factors that might support the survival, enhance the activity, or even regenerate degenerating dopamine neurons have been tried with mixed results in patients. As growth factors do not pass the blood-brain barrier, they have to be delivered intracranially. Therefore their efficient diffusion in brain tissue is of crucial importance. To improve the diffusion of the growth factor neurturin (NRTN), we modified its capacity to attach to heparan sulfates in the extracellular matrix. We present four new, biologically fully active variants with reduced heparin binding. Two of these variants are more stable than WT NRTN in vitro and diffuse better in rat brains. We also show that one of the NRTN variants diffuses better than its close homolog GDNF in monkey brains. The variant with the highest stability and widest diffusion regenerates dopamine fibers and improves the conditions of rats in a 6-hydroxydopamine model of Parkinson's disease more potently than GDNF, which previously showed modest efficacy in clinical trials. The new NRTN variants may help solve the major problem of inadequate distribution of NRTN in human brain tissue.


Asunto(s)
Diseño de Fármacos , Variación Genética/genética , Neurturina/química , Neurturina/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Anfetamina/farmacología , Animales , Células CHO , Cricetulus , Modelos Animales de Enfermedad , Humanos , Macaca fascicularis , Masculino , Modelos Moleculares , Neurturina/genética , Oxidopamina/toxicidad , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/etiología , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Ratas , Ratas Wistar , Conducta Estereotipada/efectos de los fármacos , Simpaticolíticos/toxicidad , Tirosina 3-Monooxigenasa/metabolismo
4.
Hum Mol Genet ; 24(8): 2185-200, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25552655

RESUMEN

Proper functioning of cilia, hair-like structures responsible for sensation and locomotion, requires nephrocystin-5 (NPHP5) and a multi-subunit complex called the Bardet-Biedl syndrome (BBS)ome, but their precise relationship is not understood. The BBSome is involved in the trafficking of membrane cargos to cilia. While it is known that a loss of any single subunit prevents ciliary trafficking of the BBSome and its cargos, the mechanisms underlying ciliary entry of this complex are not well characterized. Here, we report that a transition zone protein NPHP5 contains two separate BBS-binding sites and interacts with the BBSome to mediate its integrity. Depletion of NPHP5, or expression of NPHP5 mutant missing one binding site, specifically leads to dissociation of BBS2 and BBS5 from the BBSome and loss of ciliary BBS2 and BBS5 without compromising the ability of the other subunits to traffic into cilia. Depletion of Cep290, another transition zone protein that directly binds to NPHP5, causes additional dissociation of BBS8 and loss of ciliary BBS8. Furthermore, delivery of BBSome cargos, smoothened, VPAC2 and Rab8a, to the ciliary compartment is completely disabled in the absence of single BBS subunits, but is selectively impaired in the absence of NPHP5 or Cep290. These findings define a new role of NPHP5 and Cep290 in controlling integrity and ciliary trafficking of the BBSome, which in turn impinge on the delivery of ciliary cargo.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Síndrome de Bardet-Biedl/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Cilios/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas de Neoplasias/metabolismo , Antígenos de Neoplasias/genética , Síndrome de Bardet-Biedl/genética , Proteínas de Unión a Calmodulina/genética , Proteínas de Ciclo Celular , Cilios/genética , Proteínas del Citoesqueleto , Humanos , Complejos Multiproteicos/genética , Proteínas de Neoplasias/genética , Transporte de Proteínas
5.
Exp Cell Res ; 331(2): 278-91, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25447204

RESUMEN

ORP3 is an R-Ras interacting oxysterol-binding protein homolog that regulates cell adhesion and is overexpressed in several cancers. We investigated here a novel function of ORP3 dependent on its targeting to both the endoplasmic reticulum (ER) and the plasma membrane (PM). Using biochemical and cell imaging techniques we demonstrate the mechanistic requirements for the subcellular targeting and function of ORP3 in control of R-Ras activity. We show that hyperphosphorylated ORP3 (ORP3-P) selectively interacts with the ER membrane protein VAPA, and ORP3-VAPA complexes are targeted to PM sites via the ORP3 pleckstrin homology (PH) domain. A novel FFAT (two phenylalanines in an acidic tract)-like motif was identified in ORP3; only disruption of both the FFAT-like and canonical FFAT motif abolished the phorbol-12-myristate-13-acetate (PMA) stimulated interaction of ORP3-P with VAPA. Co-expression of ORP3 and VAPA induced R-Ras activation, dependent on the interactions of ORP3 with VAPA and the PM. Consistently, downstream AktS473 phosphorylation and ß1-integrin activity were enhanced by ORP3-VAPA. To conclude, phosphorylation of ORP3 controls its association with VAPA. Furthermore, we present evidence that ORP3-VAPA complexes stimulate R-Ras signaling.


Asunto(s)
Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas ras/metabolismo , Secuencias de Aminoácidos , Proteínas Portadoras/biosíntesis , Línea Celular Tumoral , Activación Enzimática , Proteínas de Unión a Ácidos Grasos , Células HEK293 , Humanos , Integrina beta1/metabolismo , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología , Proteínas de Transporte Vesicular/biosíntesis
6.
Proc Natl Acad Sci U S A ; 112(1): 148-53, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25535387

RESUMEN

Exocytosis is tightly regulated in many cellular processes, from neurite expansion to tumor proliferation. Rab8, a member of the Rab family of small GTPases, plays an important role in membrane trafficking from the trans-Golgi network and recycling endosomes to the plasma membrane. Rabin8 is a guanine nucleotide exchange factor (GEF) and major activator of Rab8. Investigating how Rabin8 is activated in cells is thus pivotal to the understanding of the regulation of exocytosis. Here we show that phosphorylation serves as an important mechanism for Rabin8 activation. We identified Rabin8 as a direct phospho-substrate of ERK1/2 in response to EGF signaling. At the molecular level, ERK phosphorylation relieves the autoinhibition of Rabin8, thus promoting its GEF activity. We further demonstrate that blocking ERK1/2-mediated phosphorylation of Rabin8 inhibits transferrin recycling to the plasma membrane. Together, our results suggest that ERK1/2 activate Rabin8 to regulate vesicular trafficking to the plasma membrane in response to extracellular signaling.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rab/metabolismo , Secuencia de Aminoácidos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Endocitosis/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Quinasas del Centro Germinal , Células HEK293 , Células HeLa , Humanos , Datos de Secuencia Molecular , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Conformación Proteica , Proteínas Serina-Treonina Quinasas/química , Estructura Terciaria de Proteína , Transducción de Señal/efectos de los fármacos , Transferrina/metabolismo
7.
Cell Mol Life Sci ; 72(10): 1967-87, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25420878

RESUMEN

Oxysterol-binding protein/OSBP-related proteins (ORPs) constitute a conserved family of sterol/phospholipid-binding proteins with lipid transporter or sensor functions. We investigated the spatial occurrence and regulation of the interactions of human OSBP/ORPs or the S. cerevisiae orthologs, the Osh (OSBP homolog) proteins, with their endoplasmic reticulum (ER) anchors, the VAMP-associated proteins (VAPs), by employing bimolecular fluorescence complementation and pull-down set-ups. The ORP-VAP interactions localize frequently at distinct subcellular sites, shown in several cases to represent membrane contact sites (MCSs). Using established ORP ligand-binding domain mutants and pull-down assays with recombinant proteins, we show that ORP liganding regulates the ORP-VAP association, alters the subcellular targeting of ORP-VAP complexes, or modifies organelle morphology. There is distinct protein specificity in the effects of the mutants on subcellular targeting of ORP-VAP complexes. We provide evidence that complexes of human ORP2 and VAPs at ER-lipid droplet interfaces regulate the hydrolysis of triglycerides and lipid droplet turnover. The data suggest evolutionarily conserved, complex ligand-dependent functions of ORP-VAP complexes at MCSs, with implications for cellular lipid homeostasis and signaling.


Asunto(s)
Amina Oxidasa (conteniendo Cobre)/metabolismo , Moléculas de Adhesión Celular/metabolismo , Retículo Endoplásmico/metabolismo , Gotas Lipídicas/metabolismo , Complejos Multiproteicos/metabolismo , Receptores de Esteroides/metabolismo , Proteínas Recombinantes/metabolismo , Triglicéridos/metabolismo , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Prueba de Complementación Genética , Humanos , Hidrólisis , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Interferencia de ARN , Levaduras
8.
PLoS Genet ; 10(3): e1004193, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24603431

RESUMEN

Although the growth factor (GF) signaling guiding renal branching is well characterized, the intracellular cascades mediating GF functions are poorly understood. We studied mitogen-activated protein kinase (MAPK) pathway specifically in the branching epithelia of developing kidney by genetically abrogating the pathway activity in mice lacking simultaneously dual-specificity protein kinases Mek1 and Mek2. Our data show that MAPK pathway is heterogeneously activated in the subset of G1- and S-phase epithelial cells, and its tissue-specific deletion results in severe renal hypodysplasia. Consequently to the deletion of Mek1/2, the activation of ERK1/2 in the epithelium is lost and normal branching pattern in mutant kidneys is substituted with elongation-only phenotype, in which the epithelium is largely unable to form novel branches and complex three-dimensional patterns, but able to grow without primary defects in mitosis. Cellular characterization of double mutant epithelium showed increased E-cadherin at the cell surfaces with its particular accumulation at baso-lateral locations. This indicates changes in cellular adhesion, which were revealed by electron microscopic analysis demonstrating intercellular gaps and increased extracellular space in double mutant epithelium. When challenged to form monolayer cultures, the mutant epithelial cells were impaired in spreading and displayed strong focal adhesions in addition to spiky E-cadherin. Inhibition of MAPK activity reduced paxillin phosphorylation on serine 83 while remnants of phospho-paxillin, together with another focal adhesion (FA) protein vinculin, were augmented at cell surface contacts. We show that MAPK activity is required for branching morphogenesis, and propose that it promotes cell cycle progression and higher cellular motility through remodeling of cellular adhesions.


Asunto(s)
Adhesiones Focales/genética , Riñón/crecimiento & desarrollo , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 2/genética , Animales , Células Epiteliales/metabolismo , Riñón/metabolismo , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Ratones , Proteínas Quinasas Activadas por Mitógenos/genética , Morfogénesis/genética , Fosforilación , Transducción de Señal/genética , Vinculina/metabolismo
9.
Dev Cell ; 27(3): 249-62, 2013 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-24209575

RESUMEN

Mammalian cells acquire cholesterol, a major membrane constituent, via low-density lipoprotein (LDL) uptake. However, the mechanisms by which LDL cholesterol reaches the plasma membrane (PM) have remained obscure. Here, we applied LDL labeled with BODIPY cholesteryl linoleate to identify this pathway in living cells. The egress of BODIPY cholesterol (BC) from late endosomal (LE) organelles was dependent on acid lipase and Niemann-Pick C1 (NPC1) protein, as for natural cholesterol. We show that NPC1 was needed to recruit Rab8a to BC-containing LEs, and Rab8a enhanced the motility and segregation of BC- and CD63-positive organelles from lysosomes. The BC carriers docked to the cortical actin by a Rab8a- and Myosin5b (Myo5b)-dependent mechanism, typically in the proximity of focal adhesions (FAs). LDL increased the number and dynamics of FAs and stimulated cell migration in an acid lipase, NPC1, and Rab8a-dependent fashion, providing evidence that this cholesterol delivery route to the PM is important for cell movement.


Asunto(s)
Actinas/metabolismo , Membrana Celular/metabolismo , LDL-Colesterol/metabolismo , Miosinas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Transporte Biológico , Proteínas Portadoras/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Técnica del Anticuerpo Fluorescente , Adhesiones Focales/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/metabolismo , Microscopía Inmunoelectrónica , Proteína Niemann-Pick C1 , Porfobilinógeno/análogos & derivados , Porfobilinógeno/farmacología , Tetraspanina 30/metabolismo , Cicatrización de Heridas/efectos de los fármacos
10.
Brain Behav ; 3(2): 75-88, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23532969

RESUMEN

Cerebral dopamine neurotrophic factor (CDNF) protein has been shown to protect the nigrostriatal dopaminergic pathway when given as intrastriatal infusions in rat and mouse models of Parkinson's disease (PD). In this study, we assessed the neuroprotective effect of CDNF delivered with a recombinant adeno-associated viral (AAV) serotype 2 vector in a rat 6-hydroxydopamine (6-OHDA) model of PD. AAV2 vectors encoding CDNF, glial cell line-derived neurotrophic factor (GDNF), or green fluorescent protein were injected into the rat striatum. Protein expression analysis showed that our AAV2 vector efficiently delivered the neurotrophic factor genes into the brain and gave rise to a long-lasting expression of the proteins. Two weeks after AAV2 vector injection, 6-OHDA was injected into the rat striatum, creating a progressive degeneration of the nigrostriatal dopaminergic system. Treatment with AAV2-CDNF resulted in a marked decrease in amphetamine-induced ipsilateral rotations while it provided only partial protection of tyrosine hydroxylase (TH)-immunoreactive cells in the rat substantia nigra pars compacta and TH-reactive fibers in the striatum. Results from this study provide additional evidence that CDNF can be considered a potential treatment of Parkinson's disease.

11.
Mol Biol Cell ; 24(7): 1007-19, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23389633

RESUMEN

Multiple Rabs are associated with secretory granules/vesicles, but how these GTPases are coordinated to promote regulated exocytosis is not well understood. In bladder umbrella cells a subapical pool of discoidal/fusiform-shaped vesicles (DFVs) undergoes Rab11a-dependent regulated exocytosis in response to bladder filling. We show that Rab11a-associated vesicles are enmeshed in an apical cytokeratin meshwork and that Rab11a likely acts upstream of Rab8a to promote exocytosis. Surprisingly, expression of Rabin8, a previously described Rab11a effector and guanine nucleotide exchange factor for Rab8, stimulates stretch-induced exocytosis in a manner that is independent of its catalytic activity. Additional studies demonstrate that the unconventional motor protein myosin5B motor (Myo5B) works in association with the Rab8a-Rab11a module to promote exocytosis, possibly by ensuring transit of DFVs through a subapical, cortical actin cytoskeleton before fusion. Our results indicate that Rab11a, Rab8a, and Myo5B function as part of a network to promote stretch-induced exocytosis, and we predict that similarly organized Rab networks will be common to other regulated secretory pathways.


Asunto(s)
Exocitosis , Miosinas/metabolismo , Vejiga Urinaria/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hormona de Crecimiento Humana/genética , Hormona de Crecimiento Humana/metabolismo , Humanos , Microscopía Confocal , Microscopía Electrónica , Miosinas/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Estrés Mecánico , Vejiga Urinaria/citología , Vejiga Urinaria/ultraestructura , Proteínas de Unión al GTP rab/genética
12.
Mol Biol Cell ; 24(3): 331-41, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23197474

RESUMEN

Sec1/Munc18 family proteins are important components of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex-mediated membrane fusion processes. However, the molecular interactions and the mechanisms involved in Sec1p/Munc18 control and SNARE complex assembly are not well understood. We provide evidence that Mso1p, a Sec1p- and Sec4p-binding protein, interacts with membranes to regulate membrane fusion. We identify two membrane-binding sites on Mso1p. The N-terminal region inserts into the lipid bilayer and appears to interact with the plasma membrane, whereas the C-terminal region of the protein binds phospholipids mainly through electrostatic interactions and may associate with secretory vesicles. The Mso1p membrane interactions are essential for correct subcellular localization of Mso1p-Sec1p complexes and for membrane fusion in Saccharomyces cerevisiae. These characteristics are conserved in the phosphotyrosine-binding (PTB) domain of ß-amyloid precursor protein-binding Mint1, the mammalian homologue of Mso1p. Both Mint1 PTB domain and Mso1p induce vesicle aggregation/clustering in vitro, supporting a role in a membrane-associated process. The results identify Mso1p as a novel lipid-interacting protein in the SNARE complex assembly machinery. Furthermore, our data suggest that a general mode of interaction, consisting of a lipid-binding protein, a Rab family GTPase, and a Sec1/Munc18 family protein, is important in all SNARE-mediated membrane fusion events.


Asunto(s)
Membrana Celular/metabolismo , Exocitosis , Fusión de Membrana , Proteínas de la Membrana/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Secuencia Conservada , Humanos , Proteínas de la Membrana/química , Datos de Secuencia Molecular , Proteínas Munc18/metabolismo , Proteínas del Tejido Nervioso/química , Células PC12 , Fosfatidilinositol 4,5-Difosfato/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Saccharomyces cerevisiae/citología , Proteínas de Saccharomyces cerevisiae/química , Vesículas Secretoras/metabolismo , Proteínas de Unión al GTP rab/metabolismo
13.
J Biol Chem ; 286(32): 28276-86, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21685394

RESUMEN

Primary cilia regulate polarized protein trafficking in photoreceptors, which are dynamic and highly compartmentalized sensory neurons of retina. The ciliary protein Cep290 modulates cilia formation and is frequently mutated in syndromic and non-syndromic photoreceptor degeneration. However, the underlying mechanism of associated retinopathy is unclear. Using the Cep290 mutant mouse rd16 (retinal degeneration 16), we show that Cep290-mediated photoreceptor degeneration is associated with aberrant accumulation of its novel interacting partner Rkip (Raf-1 kinase inhibitory protein). This effect is phenocopied by morpholino-mediated depletion of cep290 in zebrafish. We further demonstrate that ectopic accumulation of Rkip leads to defective cilia formation in zebrafish and cultured cells, an effect mediated by its interaction with the ciliary GTPase Rab8A. Our data suggest that Rkip prevents cilia formation and is associated with Cep290-mediated photoreceptor degeneration. Furthermore, our results indicate that preventing accumulation of Rkip could potentially ameliorate such degeneration.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Trastornos de la Motilidad Ciliar/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Degeneración Retiniana/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Antígenos de Neoplasias/genética , Proteínas de Ciclo Celular , Chlorocebus aethiops , Cilios/genética , Cilios/metabolismo , Cilios/patología , Trastornos de la Motilidad Ciliar/genética , Trastornos de la Motilidad Ciliar/patología , Proteínas del Citoesqueleto , Células HEK293 , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas de Unión a Fosfatidiletanolamina/genética , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
14.
PLoS One ; 5(11): e13886, 2010 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-21079737

RESUMEN

The current view of peroxisome inheritance provides for the formation of new peroxisomes by both budding from the endoplasmic reticulum and autonomous division. Here we investigate peroxisome-cytoskeleton interactions and show by proteomics, biochemical and immunofluorescence analyses that actin, non-muscle myosin IIA (NMM IIA), RhoA, Rho kinase II (ROCKII) and Rab8 associate with peroxisomes. Our data provide evidence that (i) RhoA in its inactive state, maintained for example by C. botulinum toxin exoenzyme C3, dissociates from peroxisomes enabling microtubule-based peroxisomal movements and (ii) dominant-active RhoA targets to peroxisomes, uncouples the organelles from microtubules and favors Rho kinase recruitment to peroxisomes. We suggest that ROCKII activates NMM IIA mediating local peroxisomal constrictions. Although our understanding of peroxisome-cytoskeleton interactions is still incomplete, a picture is emerging demonstrating alternate RhoA-dependent association of peroxisomes to the microtubular and actin cytoskeleton. Whereas association of peroxisomes to microtubules clearly serves bidirectional, long-range saltatory movements, peroxisome-acto-myosin interactions may support biogenetic functions balancing peroxisome size, shape, number, and clustering.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Peroxisomas/metabolismo , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Técnica del Anticuerpo Fluorescente , Proteínas Fluorescentes Verdes , Guanosina Trifosfato/metabolismo , Guanosina Trifosfato/farmacología , Humanos , Immunoblotting , Microscopía Confocal , Microscopía Electrónica de Transmisión , Miosina Tipo IIA no Muscular/metabolismo , Peroxisomas/ultraestructura , Unión Proteica/efectos de los fármacos , Proteómica/métodos , Ratas , Espectrometría de Masa por Ionización de Electrospray , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
15.
Nat Cell Biol ; 12(11): 1035-45, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20890297

RESUMEN

To form epithelial organs cells must polarize and generate de novo an apical domain and lumen. Epithelial polarization is regulated by polarity complexes that are hypothesized to direct downstream events, such as polarized membrane traffic, although this interconnection is not well understood. We have found that Rab11a regulates apical traffic and lumen formation through the Rab guanine nucleotide exchange factor (GEF), Rabin8, and its target, Rab8a. Rab8a and Rab11a function through the exocyst to target Par3 to the apical surface, and control apical Cdc42 activation through the Cdc42 GEF, Tuba. These components assemble at a transient apical membrane initiation site to form the lumen. This Rab11a-directed network directs Cdc42-dependent apical exocytosis during lumen formation, revealing an interaction between the machineries of vesicular transport and polarization.


Asunto(s)
Polaridad Celular/fisiología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Morfogénesis , Animales , Anexina A2/metabolismo , Membrana Celular/metabolismo , Perros , Quinasas del Centro Germinal , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rab/metabolismo
16.
J Neurosci ; 30(34): 11403-13, 2010 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-20739562

RESUMEN

Endocrine and neuronal cells have highly developed secretion mechanisms, and the secretion can be either constitutive or regulated by physiological stimuli. In the constitutive pathway, intracellular transport vesicles undergo immediate fusion reactions after arrival at the target. In regulated secretion, vesicles accumulate near the target membrane until triggered to fuse, typically by a local rise in free Ca(2+). In the present study, we characterize the processing and secretion mechanisms of the glial cell line-derived neurotrophic factor (GDNF). Although the function of GDNF has been extensively studied, very little is known about the basic cell biology of GDNF and its precursor forms (alpha)pro-GDNF and (beta)pro-GDNF that have different pro-regions. Our results show that both (alpha)pro-GDNF and (beta)pro-GDNF are secreted. We demonstrate that KCl-induced depolarization increases the secretion of (beta)pro-GDNF and corresponding mature GDNF, but not (alpha)pro-GDNF and corresponding mature GDNF, to the cell medium in a Ca(2+)-dependent manner. In parallel with this, immunofluorescence analysis of cells show that (alpha)pro-GDNF/GDNF is localized mostly in the Golgi complex, whereas (beta)pro-GDNF/GDNF is localized primarily in secretogranin II and Rab3A-positive vesicles of the regulated secretory pathway. In addition, we find that matrix metalloproteinases and plasmin that cleave pro-BDNF and pro-NGF are not responsible for the cleavage of pro-GDNF, whereas furin endoproteinase, PACE4, and proprotein convertases PC5A, PC5B, and PC7 can cleave pro-GDNF into mature GDNF. Thus, the processing and secretion mechanisms of GDNF are different from those of BDNF and NGF.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/análisis , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Líquido Intracelular/metabolismo , Procesamiento Postranscripcional del ARN , Empalme Alternativo/genética , Secuencia de Aminoácidos , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Aparato de Golgi/química , Aparato de Golgi/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Neuritas/metabolismo , Células PC12 , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Precursores de Proteínas/análisis , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Procesamiento Postranscripcional del ARN/genética , Ratas , Vesículas Secretoras/química , Vesículas Secretoras/genética , Vesículas Secretoras/metabolismo
17.
Exp Neurol ; 225(1): 104-13, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20685313

RESUMEN

Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a secreted protein which reduces endoplasmic reticulum (ER) stress and has neurotrophic effects on dopaminergic neurons. Intracortical delivery of recombinant MANF protein protects tissue from ischemic brain injury in vivo. In this study, we examined the protective effect of adeno-associated virus serotype 7 encoding MANF in a rodent model of stroke. An AAV vector containing human MANF cDNA (AAV-MANF) was constructed and verified for expression of MANF protein. AAV-MANF or an AAV control vector was administered into three sites in the cerebral cortex of adult rats. One week after the vector injections, the right middle cerebral artery (MCA) was ligated for 60min. Behavioral monitoring was conducted using body asymmetry analysis, neurological testing, and locomotor activity. Standard immunohistochemical and western blotting procedures were conducted to study MANF expression. Our data showed that AAV-induced MANF expression is redistributed in neurons and glia in cerebral cortex after ischemia. Pretreatment with AAV-MANF reduced the volume of cerebral infarction and facilitated behavioral recovery in stroke rats. In conclusion, our data suggest that intracortical delivery of AAV-MANF increases MANF protein production and reduces ischemic brain injury. Ischemia also caused redistribution of AAV-mediated MANF protein suggesting an injury-induced release.


Asunto(s)
Isquemia Encefálica/genética , Isquemia Encefálica/terapia , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Factores de Crecimiento Nervioso/genética , Proteínas/genética , Animales , Isquemia Encefálica/fisiopatología , Células Cultivadas , Dependovirus/fisiología , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Humanos , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/fisiopatología , Infarto de la Arteria Cerebral Media/terapia , Masculino , Actividad Motora/genética , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/uso terapéutico , Proteínas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/genética
18.
PLoS One ; 4(4): e5093, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19352496

RESUMEN

The prototype baculovirus, Autographa californica multiple nucleopolyhedrovirus, an insect pathogen, holds great potential as a gene therapy vector. To develop transductional targeting and gene delivery by baculovirus, we focused on characterizing the nature and regulation of its uptake in human cancer cells. Baculovirus entered the cells along fluid-phase markers from the raft areas into smooth-surfaced vesicles devoid of clathrin. Notably, regulators associated with macropinocytosis, namely EIPA, Pak1, Rab34, and Rac1, had no significant effect on viral transduction, and the virus did not induce fluid-phase uptake. The internalization and nuclear uptake was, however, affected by mutants of RhoA, and of Arf6, a regulator of clathrin-independent entry. Furthermore, the entry of baculovirus induced ruffle formation and triggered the uptake of fluorescent E. coli bioparticles. To conclude, baculovirus enters human cells via a clathrin-independent pathway, which is able to trigger bacterial uptake. This study increases our understanding of virus entry strategies and gives new insight into baculovirus-mediated gene delivery in human cells.


Asunto(s)
Clatrina/fisiología , Endocitosis , Escherichia coli/fisiología , Nucleopoliedrovirus/fisiología , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/fisiología , Adenosina Trifosfatasas/fisiología , Secuencia de Bases , Línea Celular , Humanos , Lípidos de la Membrana/metabolismo , Fagocitosis , Interferencia de ARN
19.
J Comp Neurol ; 515(1): 116-24, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19399876

RESUMEN

Mesencephalic astrocyte-derived neurotrophic factor (MANF), also known as arginine-rich, mutated in early stage of tumors (ARMET), is a secreted protein that reduces endoplasmic reticulum (ER) stress. Previous studies have shown that MANF mRNA expression and protein levels are increased in the cerebral cortex after brain ischemia, a condition that induces ER stress. The function of MANF during brain ischemia is still not known. The purpose of this study was to examine the protective effect of MANF after ischemic brain injury. Recombinant human MANF was administrated locally to the cerebral cortex before a 60-min middle cerebral artery occlusion (MCAo) in adult rats. Triphenyltetrazolium chloride (TTC) staining indicated that pretreatment with MANF significantly reduced the volume of infarction at 2 days after MCAo. MANF also attenuated TUNEL labeling, a marker of cell necrosis/apoptosis, in the ischemic cortex. Animals receiving MANF pretreatment demonstrated a decrease in body asymmetry and neurological score as well as an increase in locomotor activity after MCAo. Taken together, these data suggest that MANF has neuroprotective effects against cerebral ischemia, possibly through the inhibition of cell necrosis/apoptosis in cerebral cortex.


Asunto(s)
Infarto Encefálico/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Degeneración Nerviosa/tratamiento farmacológico , Proteínas del Tejido Nervioso/uso terapéutico , Recuperación de la Función/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Infarto Encefálico/metabolismo , Infarto Encefálico/fisiopatología , Citoprotección/efectos de los fármacos , Citoprotección/fisiología , Modelos Animales de Enfermedad , Humanos , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/fisiopatología , Indicadores y Reactivos , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Necrosis/tratamiento farmacológico , Necrosis/metabolismo , Necrosis/fisiopatología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Factores de Crecimiento Nervioso , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Recuperación de la Función/fisiología , Coloración y Etiquetado , Sales de Tetrazolio , Resultado del Tratamiento
20.
Arterioscler Thromb Vasc Biol ; 29(6): 883-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19304576

RESUMEN

OBJECTIVE: ATP-binding cassette transporter A1 (ABCA1) is thought to lipidate apolipoprotein A-I (apoA-I) at the plasma membrane, with endosomal cholesterol contributing as substrate. The mechanisms of ABCA1 surface delivery are not well understood. We have shown that Rab8 regulates endosomal cholesterol removal to apoA-I in human fibroblasts. Here, we investigated whether Rab8 plays a role in ABCA1 plasma membrane expression and cholesterol removal in primary human macrophages. METHODS AND RESULTS: We found that Rab8 was abundantly expressed in human atherosclerotic lesional macrophages and upregulated on lipid loading of macrophages in vitro. Adenoviral overexpression of Rab8 increased ABCA1 protein levels and reduced cholesterol deposition in macrophage foam cells incubated with apoA-I. Depletion of Rab8 decreased the fraction of ABCA1 at the plasma membrane and inhibited the efflux of lipoprotein-derived endosomal cholesterol to apoA-I. In Rab8-depleted cells, ABCA1-GFP localized in beta1 integrin and transferrin receptor containing recycling organelles. CONCLUSIONS: Rab8 reduces foam cell formation by facilitating ABCA1 surface expression and stimulating endosomal cholesterol efflux to apoA-I in primary human macrophages.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Células Espumosas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Transportador 1 de Casete de Unión a ATP , Apolipoproteína A-I/metabolismo , Transporte Biológico , Células Cultivadas , Endosomas/metabolismo , Humanos , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transfección , Proteínas de Unión al GTP rab/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA