Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 298(11): 102531, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36162505

RESUMEN

α-synucleinopathy is driven by an imbalance of synthesis and degradation of α-synuclein (αSyn), causing a build up of αSyn aggregates and post-translationally modified species, which not only interfere with normal cellular metabolism but also by their secretion propagates the disease. Therefore, a better understanding of αSyn degradation pathways is needed to address α-synucleinopathy. Here, we used the nerve growth factor-differentiated catecholaminergic PC12 neuronal cell line, which was conferred α-synucleinopathy by inducible expression of αSyn and tubulin polymerization-promoting protein p25α. p25α aggregates αSyn, and imposes a partial autophagosome-lysosome block to mimic aspects of lysosomal deficiency common in neurodegenerative disease. Under basal conditions, αSyn was degraded by multiple pathways but most prominently by macroautophagy and Nedd4/Ndfip1-mediated degradation. We found that expression of p25α induced strong p38MAPK activity. Remarkably, when opposed by inhibitor SB203580 or p38MAPK shRNA knockdown, endolysosomal localization and degradation of αSyn increased, and αSyn secretion and cytotoxicity decreased. This effect was specifically dependent on Hsc70 and the endosomal sorting complex required for transport machinery, but different from classical microautophagy, as the αSyn Hsc70 binding motif was unnecessary. Furthermore, in a primary neuronal (h)-αSyn seeding model, p38MAPK inhibition decreased pathological accumulation of phosphorylated serine-129-αSyn and cytotoxicity. In conclusion, p38MAPK inhibition shifts αSyn degradation from various forms of autophagy to an endosomal sorting complex required for transport-dependent uptake mechanism, resulting in increased αSyn turnover and cell viability in p25α-expressing cells. More generally, our results suggest that under conditions of autophagolysosomal malfunction, the uninterrupted endosomal pathway offers a possibility to achieve disease-associated protein degradation.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte , Proteínas del Tejido Nervioso , alfa-Sinucleína , Proteínas Quinasas p38 Activadas por Mitógenos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Sinucleinopatías , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células PC12 , Animales , Ratas
2.
J Neuroinflammation ; 13(1): 59, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26957005

RESUMEN

BACKGROUND: Secretion of proteopathic α-synuclein (α-SNC) species from neurons is a suspected driving force in the propagation of Parkinson's disease (PD). We have previously implicated exophagy, the exocytosis of autophagosomes, as a dominant mechanism of α-SNC secretion in differentiated PC12 or SH-SY5Y nerve cells. Here we have examined the regulation of exophagy associated with different forms of nerve cell stress relevant to PD. RESULTS: We identify cJUN-N-terminal kinase (JNK) activity as pivotal in the secretory fate of autophagosomes containing α-SNC. Pharmacological inhibition or genetic (shRNA) knockdown of JNK2 or JNK3 decreases α-SNC secretion in differentiated PC12 and SH-SY5Y cells, respectively. Conversely, expression of constitutively active mitogen-activated protein kinase kinase 7 (MKK7)-JNK2 and -JNK3 constructs augment secretion. The transcriptional activity of cJUN was not required for the observed effects. We establish a causal relationship between increased α-SNC release by exophagy and JNK activation subsequent to lysosomal fusion deficiency (overexpression of Lewy body-localized protein p25α or bafilomycin A1). JNK activation following neuronal ER or oxidative stress was not correlated with exophagy, but of note, we demonstrate that reciprocal signaling between microglia and neurons modulates α-SNC secretion. NADPH oxidase activity of microglia cell lines was upregulated by direct co-culture with α-SNC-expressing PC12 neurons or by passive transfer of nerve cell-conditioned medium. Conversely, inflammatory factors secreted from activated microglia increased JNK activation and α-SNC secretion several-fold in PC12 cells. While we do not identify these factors, we extend our observations by showing that exposure of neurons in monoculture to TNFα, a classical pro-inflammatory mediator of activated microglia, is sufficient to increase α-SNC secretion in a mechanism dependent on JNK2 or JNK3. In continuation hereof, we show that also IFNß and TGFß increase the release of α-SNC from PC12 neurons. CONCLUSIONS: We implicate stress kinases of the JNK family in the regulation of exophagy and release of α-SNC following endogenous or exogenous stimulation. In a wider scope, our results imply that microglia not only inflict bystander damage to neurons in late phases of inflammatory brain disease but may also be active mediators of disease propagation.


Asunto(s)
MAP Quinasa Quinasa 4/fisiología , Microglía/fisiología , Neuronas/fisiología , alfa-Sinucleína/metabolismo , Animales , Autofagia/fisiología , Comunicación Celular/fisiología , Estrés del Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/fisiología , Activación Enzimática , Técnicas de Silenciamiento del Gen , Humanos , Microglía/enzimología , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Neuronas/enzimología , Células PC12 , Ratas , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/farmacología
3.
J Virol ; 89(3): 1851-66, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25428868

RESUMEN

UNLABELLED: The entry mechanism of murine amphotropic retrovirus (A-MLV) has not been unambiguously determined. We show here that A-MLV is internalized not by caveolae or other pinocytic mechanisms but by macropinocytosis. Thus, A-MLV infection of mouse embryonic fibroblasts deficient for caveolin or dynamin, and NIH 3T3 cells knocked down for caveolin expression, was unaffected. Conversely, A-MLV infection of NIH 3T3 and HeLa cells was sensitive to amiloride analogues and actin-depolymerizing drugs that interfere with macropinocytosis. Further manipulation of the actin cytoskeleton through conditional expression of dominant positive or negative mutants of Rac1, PAK1, and RhoG, to increase or decrease macropinocytosis, similarly correlated with an augmented or inhibited infection with A-MLV, respectively. The same experimental perturbations affected the infection of viruses that use clathrin-coated-pit endocytosis or other pathways for entry only mildly or not at all. These data agree with immunofluorescence studies and cryo-immunogold labeling for electron microscopy, which demonstrate the presence of A-MLV in protrusion-rich areas of the cell surface and in cortical fluid phase (dextran)-filled macropinosomes, which also account for up to a half of the cellular uptake of the cell surface-binding lectin concanavalin A. We conclude that A-MLV use macropinocytosis as the predominant entry portal into cells. IMPORTANCE: Binding and entry of virus particles into mammalian cells are the first steps of infection. Understanding how pathogens and toxins exploit or divert endocytosis pathways has advanced our understanding of membrane trafficking pathways, which benefits development of new therapeutic schemes and methods of drug delivery. We show here that amphotropic murine leukemia virus (A-MLV) pseudotyped with the amphotropic envelope protein (which expands the host range to many mammalian cells) gains entry into host cells by macropinocytosis. Macropinosomes form as large, fluid-filled vacuoles (up to 10 µm) following the collapse of cell surface protrusions and membrane scission. We used drugs or the introduction of mutant proteins that affect the actin cytoskeleton and cell surface dynamics to show that macropinocytosis and A-MLV infection are correlated, and we provide both light- and electron-microscopic evidence to show the localization of A-MLV in macropinosomes. Finally, we specifically exclude some other potential entry portals, including caveolae, previously suggested to internalize A-MLV.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Leucemia Murina/fisiología , Pinocitosis , Internalización del Virus , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Noqueados , Microscopía Fluorescente , Microscopía Inmunoelectrónica
4.
J Biol Chem ; 288(24): 17313-35, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23629650

RESUMEN

Aggregation of α-synuclein can be promoted by the tubulin polymerization-promoting protein/p25α, which we have used here as a tool to study the role of autophagy in the clearance of α-synuclein. In NGF-differentiated PC12 catecholaminergic nerve cells, we show that de novo expressed p25α co-localizes with α-synuclein and causes its aggregation and distribution into autophagosomes. However, p25α also lowered the mobility of autophagosomes and hindered the final maturation of autophagosomes by preventing their fusion with lysosomes for the final degradation of α-synuclein. Instead, p25α caused a 4-fold increase in the basal level of α-synuclein secreted into the medium. Secretion was strictly dependent on autophagy and could be up-regulated (trehalose and Rab1A) or down-regulated (3-methyladenine and ATG5 shRNA) by enhancers or inhibitors of autophagy or by modulating minus-end-directed (HDAC6 shRNA) or plus-end-directed (Rab8) trafficking of autophagosomes along microtubules. Finally, we show in the absence of tubulin polymerization-promoting protein/p25α that α-synuclein release was modulated by dominant mutants of Rab27A, known to regulate exocytosis of late endosomal (and amphisomal) elements, and that both lysosomal fusion block and secretion of α-synuclein could be replicated by knockdown of the p25α target, HDAC6, the predominant cytosolic deacetylase in neurons. Our data indicate that unconventional secretion of α-synuclein can be mediated through exophagy and that factors, which increase the pool of autophagosomes/amphisomes (e.g. lysosomal disturbance) or alter the polarity of vesicular transport of autophagosomes on microtubules, can result in an increased release of α-synuclein monomer and aggregates to the surroundings.


Asunto(s)
Proteínas Portadoras/fisiología , Lisosomas/metabolismo , Fagosomas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Autofagia , Diferenciación Celular , Vesículas Citoplasmáticas/metabolismo , Técnicas de Silenciamiento del Gen , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Cuerpos de Inclusión/metabolismo , Fusión de Membrana , Proteínas Asociadas a Microtúbulos/metabolismo , Factor de Crecimiento Nervioso/fisiología , Neuritas/fisiología , Células PC12 , Transporte de Proteínas , Proteolisis , ARN Interferente Pequeño/genética , Ratas , Vías Secretoras , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Trehalosa/farmacología , Proteínas de Unión al GTP rab/metabolismo
5.
BMC Immunol ; 11: 44, 2010 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-20825680

RESUMEN

BACKGROUND: Most in vivo studies that have addressed the role of actin dynamics in NADPH oxidase function in phagocytes have used toxins to modulate the polymerization state of actin and mostly effects on actin has been evaluated by end point measurements of filamentous actin, which says little about actin dynamics, and without consideration for the subcellular distribution of the perturbed actin cytoskeleton. RESULTS: Here, we in addition to toxins use conditional expression of the major actin regulatory protein LIM kinase-1 (LIMK1), and shRNA knock-down of cofilin to modulate the cellular F/G-actin ratio in the Ra2 microglia cell line, and we use Fluorescence Recovery after Photobleaching (FRAP) in ß-actin-YFP-transduced cells to obtain a dynamic measure of actin recovery rates (actin turn-over rates) in different F/G-actin states of the actin cytoskeleton. Our data demonstrate that stimulated NADPH oxidase function was severely impaired only at extreme actin recovery rates and F/G-actin ratios, and surprisingly, that any moderate changes of these parameters of the actin cytoskeleton invariably resulted in an increased NADPH oxidase activity. CONCLUSION: moderate actin polymerization and depolymerization both increase the FMLP and PMA-stimulated NADPH oxidase activity of microglia, which is directly correlated with neither actin recovery rate nor F/G- actin ratio. Our results indicate that NADPH oxidase functions in an enhanced state of activity in stimulated phagocytes despite widely different states of the actin cytoskeleton.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Quinasas Lim/metabolismo , Microglía/metabolismo , NADPH Oxidasas/metabolismo , Factores Despolimerizantes de la Actina/genética , Actinas/genética , Animales , Línea Celular , Clonación Molecular , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Humanos , Quinasas Lim/genética , Ratones , Microglía/patología , N-Formilmetionina Leucil-Fenilalanina/farmacología , NADPH Oxidasas/genética , Polimerizacion/efectos de los fármacos , ARN Interferente Pequeño/genética , Transgenes/genética
6.
J Biol Chem ; 283(12): 7983-93, 2008 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-18160398

RESUMEN

The p21-activated kinase-1 (PAK1) is best known for its role in the regulation of cytoskeletal and transcriptional signaling pathways. We show here in the microglia cell line Ra2 that PAK1 regulates NADPH oxidase (NOX-2) activity in a stimulus-specific manner. Thus, conditional expression of PAK1 dominant-positive mutants enhanced, whereas dominant-negative mutants inhibited, NADPH oxidase-mediated superoxide generation following formyl-methionyl-leucylphenylalanine or phorbol 12-myristate 13-acetate stimulation. Both Rac1 and the GTP exchange factor VAV1 were required as upstream signaling proteins in the formyl-methionyl-leucyl-phenylalanine-induced activation of endogenous PAK1. In contrast, PAK1 mutants had no effect on superoxide generation downstream of FcgammaR signaling during phagocytosis of IgG-immune complexes. We further present evidence that the effect of PAK1 on the respiratory burst is mediated through phosphorylation of p47(Phox), and we show that expression of a p47(Phox) (S303D/S304D/S320D) mutant, which mimics phosphorylation by PAK1, induced basal superoxide generation in vivo. In contrast PAK1 substrates LIMK-1 or RhoGDI are not likely to contribute to the PAK1 effect on NADPH oxidase activation. Collectively, our findings define a VAV1-Rac1-PAK1 signaling axis in mononuclear phagocytes regulating superoxide production in a stimulus-dependent manner.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Neuropéptidos/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Transducción de Señal/fisiología , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Sustitución de Aminoácidos , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Complejo Antígeno-Anticuerpo/farmacología , Carcinógenos/farmacología , Línea Celular , Inhibidores de Disociación de Guanina Nucleótido/genética , Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Humanos , Quinasas Lim/genética , Quinasas Lim/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Microglía/citología , Microglía/metabolismo , Mutación Missense , N-Formilmetionina Leucil-Fenilalanina/farmacología , NADPH Oxidasa 2 , NADPH Oxidasas/genética , Neuropéptidos/genética , Fagocitos/metabolismo , Proteínas Proto-Oncogénicas c-vav/genética , Receptores de IgG/genética , Receptores de IgG/metabolismo , Estallido Respiratorio , Transducción de Señal/efectos de los fármacos , Superóxidos/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Quinasas p21 Activadas/genética , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/genética , Inhibidor alfa de Disociación del Nucleótido Guanina rho , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...