Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 668: 19-26, 2023 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-37235915

RESUMEN

Nanoplastics (NPs) are potentially toxic and pose a health risk as they can induce an inflammatory response and oxidative stress at cellular and organismal levels. Humans can be exposed to NPs through various routes, including ingestion, inhalation, and skin contact. Notably, uptake into the body via inhalation could result in brain accumulation, which may occur directly across the blood-brain barrier or via other routes. NPs that accumulate in the brain may be endocytosed into neurons, inducing neurotoxicity. Recently, we demonstrated that exposure to polystyrene (PS)-NPs reduces the viability of neurons. We have also reported that inhibiting the retrograde transport of PS-NPs by histone deacetylase 6 (HDAC6) prevents their intracellular accumulation and promotes their export in mouse embryonic fibroblasts. However, whether HDAC6 inhibition can improve neuronal viability by increasing exocytosis of PS-NPs from neurons remains unknown. In this study, mice were intranasally administered fluorescent PS-NPs (PS-YG), which accumulated in the brain and showed potential neurotoxic effects. In cultured neurons, the HDAC6 inhibitor ACY-1215 reduced the fluorescence signal detected from PS-YG, suggesting that the removal of PS-YG from neurons was promoted. Therefore, these results suggest that blocking the retrograde transport of PS-NPs using an HDAC6 inhibitor can alleviate the neurotoxic effects of PS-NPs that enter the brain.


Asunto(s)
Nanopartículas , Contaminantes Químicos del Agua , Humanos , Animales , Ratones , Poliestirenos/toxicidad , Microplásticos , Nanopartículas/toxicidad , Fibroblastos , Neuronas
2.
Int J Mol Sci ; 24(21)2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37958847

RESUMEN

Astrocytes and microglia, the most abundant glial cells in the central nervous system, are involved in maintaining homeostasis in the brain microenvironment and in the progression of various neurological disorders. Lipocalin-2 (LCN2) is a small secretory protein that can be transcriptionally upregulated via nuclear factor kappa B (NF-κB) signaling. It is synthesized and secreted by glial cells, resulting in either the restoration of damaged neural tissues or the induction of neuronal apoptosis in a context-dependent manner. It has recently been reported that when glial cells are under lipopolysaccharide-induced inflammatory stress, either reduced production or accelerated degradation of LCN2 can alleviate neurotoxicity. However, the regulatory mechanisms of LCN2 in glial cells are not yet fully understood. In this study, we used primary astroglial-enriched cells which produce LCN2 and found that the production of LCN2 could be reduced by sodium arsenite treatment. Surprisingly, the reduced LCN2 production was not due to the suppression of NF-κB signaling. Mild oxidative stress induced by sodium arsenite treatment activated antioxidant responses and downregulated Lcn2 expression without reducing the viability of astroglial-enriched cells. Intriguingly, reduced LCN2 production could not be achieved by simple activation of the nuclear factor erythroid-2-related factor 2 (Nrf2)-Kelch-like ECH-associated protein 1 (Keap1) pathway in astroglial-enriched cells. Thus, it appears that mild oxidative stress, occurring in an Nrf2-independent manner, is required for the downregulation of Lcn2 expression. Taken together, our findings provide new insights into the regulatory mechanisms of LCN2 and suggest that mild oxidative stress may alter LCN2 homeostasis, even under neuroinflammatory conditions.


Asunto(s)
Factor 2 Relacionado con NF-E2 , FN-kappa B , Lipocalina 2/genética , Lipocalina 2/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , FN-kappa B/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Neuroglía/metabolismo , Estrés Oxidativo
3.
Int J Mol Sci ; 22(4)2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33672484

RESUMEN

Polystyrene (PS) nanoplastic exposure has been shown to affect the viability of neuronal cells isolated from mouse embryonic brains. However, the viability of mouse embryonic fibroblasts (MEFs) was not affected although PS nanoplastics accumulated in the cytoplasm. It is currently unknown whether MEFs do not respond to PS nanoplastics or their cellular functions are altered without compromising viability. Here, we found that PS nanoplastics entered the cells via endocytosis and were then released into the cytoplasm, probably by endosomal escape, or otherwise remained in the endosome. Oxidative and inflammatory stress caused by intracellular PS nanoplastics induced the antioxidant response pathway and activated the autophagic pathway. However, colocalization of the autophagic marker LC3B and PS nanoplastics suggested that PS nanoplastics in the cytoplasm might interfere with normal autophagic function. Furthermore, autophagic flux could be impaired, probably due to accumulation of PS nanoplastic-containing lysosomes or autolysosomes. Intriguingly, the level of accumulated PS nanoplastics decreased during prolonged culture when MEFs were no longer exposed to PS nanoplastics. These results indicate that accumulated PS nanoplastics are removed or exported out of the cells. Therefore, PS nanoplastics in the cytoplasm affect cellular functions, but it is temporal and MEFs can overcome the stress caused by PS nanoplastic exposure.


Asunto(s)
Embrión de Mamíferos/patología , Fibroblastos/patología , Microplásticos/toxicidad , Nanopartículas/toxicidad , Poliestirenos/toxicidad , Estrés Fisiológico , Animales , Autofagia/efectos de los fármacos , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Endocitosis/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Espacio Intracelular/metabolismo , Ratones , Estrés Fisiológico/efectos de los fármacos
4.
Biochem Biophys Res Commun ; 527(2): 372-378, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32321641

RESUMEN

Ubiquitin (Ub) is a highly conserved eukaryotic protein that plays pivotal roles in cellular signal transduction, differentiation, and proteolysis. Although we have previously reported that disruption of the polyubiquitin gene Ubb is associated with the dysregulated differentiation of neural stem cells (NSCs) into neurons, it is unclear how gene expression patterns are altered in Ubb knockout (KO) NSCs, and whether this altered gene expression contributes to Ubb KO neural phenotypes. To answer these questions, we used RNA-Seq to compare the transcriptomes of Ubb KO NSCs and Ubb heterozygous (HT) controls. We found that the expression levels of most proliferation markers were decreased in Ubb KO NSCs. To determine whether the reduced levels of proliferation markers were due to reduced self-renewal of NSCs, such as radial glia, we measured the levels of the radial glia marker, Pax6, in mouse embryonic brains at 14.5 dpc. We found that Pax6 levels were decreased and the ventricular zone was thinner in the embryonic brains of Ubb KO mice compared to those of wild-type (WT) control mice. To determine whether the decreased self-renewal of Ubb KO NSCs was caused by cell-autonomous defects and not due to their microenvironment, we transplanted NSCs into WT mouse brains using a cannula system. In mouse brain sections, immunoreactivity of the NSC marker, nestin, was much lower in Ubb KO NSCs than in Ubb HT controls. Therefore, our data suggest that cell-autonomous defects, due to the disruption of Ubb, lead to a decrease in the self-renewal capacity of NSCs and may contribute to their dysregulated differentiation into neurons.


Asunto(s)
Autorrenovación de las Células , Células-Madre Neurales/citología , Poliubiquitina/genética , Ubiquitina/genética , Animales , Células Cultivadas , Eliminación de Gen , Técnicas de Inactivación de Genes , Ratones , Células-Madre Neurales/metabolismo , Células-Madre Neurales/trasplante
5.
Exp Dermatol ; 29(6): 562-569, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32298492

RESUMEN

Melanoma is a deadly type of skin cancer that is particularly difficult to treat owing to its resistance to radiation therapy. Here, we attempted to determine the key proteins responsible for melanoma radioresistance, with the aim of improving disease response to radiation therapy. Two melanoma cell lines, SK-Mel5 and SK-Mel28, with different radiosensitivities were analysed via RNA-Seq (Quant-Seq) and target proteins with higher abundance in the more radioresistant cell line, SK-Mel28, identified. Among these proteins, integrin αvß3, a well-known molecule in cell adhesion, was selected for analysis. Treatment of SK-Mel28 cells with cilengitide, an integrin αvß3 inhibitor, as well as γ-irradiation resulted in more significant cell death than γ-irradiation alone. In addition, Akt, a downstream signal transducer of integrin αvß3, showed high basic activation in SK-Mel28 and was significantly decreased upon co-treatment with cilengitide and γ-irradiation. MK-2206, an Akt inhibitor, exerted similar effects on the SK-Mel28 cell line following γ-irradiation. Our results collectively demonstrate that the integrin αvß3-Akt signalling pathway contributes to radioresistance in SK-Mel28 cells, which may be manipulated to improve therapeutic options for melanoma.


Asunto(s)
Integrina alfaVbeta3/metabolismo , Melanoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tolerancia a Radiación , Neoplasias Cutáneas/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Rayos gamma , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Melanoma/radioterapia , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal , Neoplasias Cutáneas/radioterapia , Venenos de Serpiente/farmacología
6.
Biochem Biophys Res Commun ; 513(1): 35-40, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30935687

RESUMEN

We have previously demonstrated that a reduction in ubiquitin (Ub) levels via disruption of the polyubiquitin gene Ubb results in reactive gliosis and hypothalamic neurodegeneration in mice. However, it is not known whether other neural tissues, apart from the brain, can also be affected by Ubb disruption. We examined the retina, which, being derived from the diencephalon, has the same developmental origin as the hypothalamus. We found that expression levels of Ubb were much higher than those of the other polyubiquitin gene Ubc in the retina. In retinal tissues from Ubb knockout (KO) mice, we found that Ubc expression was upregulated to compensate for the loss of Ubb; however, the Ub pool remained disrupted, with reduced levels of free Ub. To directly demonstrate whether the disrupted Ub pools affect neural integrity in retinal tissues, we investigated retinal layers in control and Ubb KO mice. Using optical coherence tomography and histological analysis, we demonstrated that the thickness of the outer nuclear layer of the retina was decreased in Ubb KO mice compared to control mice, suggesting that retinal degeneration was induced by Ub deficiency. Furthermore, the mRNA and protein levels of rhodopsin decreased and those of glial fibrillary acidic protein increased in Ubb KO mouse retinas. Therefore, the maintenance of Ub pools in the retina appears to be crucial for the survival of photoreceptor cells and the prevention of excessive glial cell activation.


Asunto(s)
Poliubiquitina/genética , Retina/patología , Degeneración Retiniana/genética , Ubiquitina/genética , Animales , Técnicas de Inactivación de Genes , Ratones , Ratones Noqueados , Poliubiquitina/análisis , Degeneración Retiniana/patología , Ubiquitina/análisis
7.
Int J Mol Sci ; 20(13)2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-31261719

RESUMEN

Ubiquitin is required under both normal and stress conditions. Under stress conditions, upregulation of the polyubiquitin gene UBC is essential to meet the requirement of increased ubiquitin levels to confer stress resistance. However, UBC upregulation is usually observed only under stress conditions and not under normal conditions. Therefore, it has not been possible to upregulate UBC under normal conditions to study the effect of excess ubiquitin on cellular machinery. Recently, the CRISPR/Cas9 system has been widely used in biological research as a useful tool to study gene disruption effects. In this study, using an inducible CRISPR/Cas9 variant, a dCas9-VP64 fusion protein, combined with a single guide RNA (sgRNA) containing MS2 aptamer loops and MS2-p65-HSF1, we developed a system to increase the ubiquitin pool via upregulation of UBC. Although it is challenging to upregulate the expression of a gene that is already expressed at high levels, the significance of our system is that UBC upregulation can be induced in an efficient, reversible manner that is compatible with cellular processes, even under normal conditions. This system can be used to study ubiquitin pool dynamics and it will be a useful tool in identifying the role of ubiquitin under normal and stress conditions.


Asunto(s)
Sistemas CRISPR-Cas , Ingeniería Genética/métodos , Enzimas Ubiquitina-Conjugadoras/genética , Aptámeros de Nucleótidos/genética , Aptámeros de Nucleótidos/metabolismo , Células HEK293 , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Regulación hacia Arriba
8.
Biochem Biophys Res Commun ; 500(2): 344-350, 2018 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-29654755

RESUMEN

Ubiquitin (Ub) homeostasis is important for cellular function and survival, especially under stress conditions. Recently, we have demonstrated that Ubc-/- (Ub-deficient) mouse embryonic fibroblasts (MEFs) exhibited reduced viability under oxidative stress induced by arsenite, which was not due to dysregulation of the antioxidant response pathway, but rather due to the potential toxicity caused by the misfolded protein aggregates. However, it is still not clear whether Ub deficiency is directly related to the accumulation of toxic protein aggregates, as arsenite itself triggers protein aggregation and renders cells into aberrant conditions such as reduced proteasome function and inhibition of autophagic flux. Therefore, under arsenite treatment, the outcome could be derived from the combination of multiple defective pathways. Furthermore, it has also been suggested that ubiquitination status of misfolded proteins may not be important for the formation of inclusion bodies composed of misfolded protein aggregates. We therefore wondered whether Ub deficiency is sufficient to trigger the accumulation of toxic protein aggregates inside the cells. In this study, we ectopically expressed polyQ-expanded aggregates (Q103) in MEFs and observed inclusion body formation at the juxtanuclear region, which was independent of cellular Ub levels. In contrast to arsenite treatment, polyQ expression did not affect proteasome function. However, we observed an increased accumulation of Q103 aggregates in Ubc-/- MEFs, which was due to impaired autophagic clearance. Finally, we demonstrated that the increased accumulation of Q103 aggregates under Ub deficiency dramatically reduced the viability of cells. Therefore, our results suggest that the maintenance of proper levels of cellular Ub is important to protect cells against the toxicity induced by the accumulation of protein aggregates.


Asunto(s)
Citoprotección/efectos de los fármacos , Péptidos/toxicidad , Agregado de Proteínas , Expansión de Repetición de Trinucleótido , Ubiquitina/farmacología , Animales , Autofagia/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Embrión de Mamíferos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo
9.
Biochem Biophys Res Commun ; 485(2): 234-240, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28237703

RESUMEN

The polyubiquitin genes Ubb and Ubc are upregulated under oxidative stress induced by arsenite [As(III)]. However, the role of ubiquitin (Ub) under As(III) exposure is not known in detail. In a previous study, we showed that the reduced viability observed in Ubc-/- mouse embryonic fibroblasts under As(III) exposure was not due to dysregulation of the Nrf2-Keap1 pathway, which prompted us to investigate another NFE2 family protein, nuclear factor erythroid 2-related factor 1 (Nrf1). In this study, we found that Ub deficiency due to Ubc knockdown in N2a cells reduced cell viability and proteasome activity under As(III) exposure. Furthermore, mRNA levels of the proteasome subunit Psma1 were also reduced. In addition, Ub deficiency led to the nuclear accumulation of the p65 isoform of Nrf1 under As(III) exposure. Interestingly, the overexpression of p65-Nrf1 recapitulated the phenotypes of Ub-deficient N2a cells under As(III) exposure. On the other hand, Nrf1 knockdown suppressed the death of Ub-deficient N2a cells upon exposure to As(III). Therefore, the levels of p65-Nrf1 may play an important role in the maintenance of cell viability under oxidative stress induced by As(III).


Asunto(s)
Arsenitos/toxicidad , Factor Nuclear 1 de Respiración/metabolismo , Estrés Oxidativo/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Ratones , Células 3T3 NIH , Factor Nuclear 1 de Respiración/genética , Complejo de la Endopetidasa Proteasomal/genética , Ubiquitina/genética , Ubiquitina C/genética , Ubiquitina C/metabolismo
10.
Biochem Biophys Res Commun ; 486(1): 171-177, 2017 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-28285139

RESUMEN

Reduced levels of cellular ubiquitin (Ub) caused by disruption of the polyubiquitin gene Ubb lead to dysregulated differentiation of neural stem/progenitor cells (NSCs) and apoptosis in cells cultured in vitro. However, the underlying mechanisms responsible for these phenotypes in Ub-deficient cells have not been studied extensively. In the present study, we found that levels of repressor element-1 silencing transcription factor (REST) are elevated in Ubb-/- cells. To determine whether dysregulation of NSC differentiation is caused by the increased REST levels, we investigated the effect of reduced REST levels in Ubb-/- cells. Rest knockdown was found to increase the expression of the neuronal marker ßIII-tubulin (TUJ1) and restore the expression pattern of the early neuronal marker α-internexin (α-INX) in Ubb-/- cells. Furthermore, Rest knockdown reduced Ub deficiency-induced apoptosis in cells cultured in vitro. Therefore, our study validates that cellular Ub levels are crucial for precise control of the levels of regulatory proteins such as REST during neurogenesis. We propose that regulation of Rest levels is a promising approach to overcome dysregulation of NSC differentiation caused by disruption of the polyubiquitin gene Ubb.


Asunto(s)
Diferenciación Celular/genética , Regulación de la Expresión Génica , Células-Madre Neurales/metabolismo , Proteínas Represoras/genética , Ubiquitina/genética , Animales , Apoptosis/genética , Células Cultivadas , Immunoblotting , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/metabolismo , Ratones Noqueados , Células-Madre Neurales/citología , Interferencia de ARN , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Ubiquitina/deficiencia
11.
Biochem Biophys Res Commun ; 486(3): 839-844, 2017 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-28359760

RESUMEN

Oxidative stress induced by arsenite [As(III)] affects protein folding and results in increased levels of misfolded proteins or protein aggregates. Accumulation of misfolded protein aggregates may act as a cue signal for the oligomerization of the autophagic adaptor protein p62, which facilitates recognition of misfolded protein aggregates that are polyubiquitinated with K63 linkages. However, as the autophagic flux is impaired under exposure to As(III), p62 oligomers cannot be cleared by autophagy and accumulate as aggregates with Keap1. This results in the sequestration of Keap1 and the stabilization of Nrf2, which activates the non-canonical Nrf2-Keap1 pathway as an antioxidant response. In this study, we found that polyubiquitination of p62 itself increased upon exposure to As(III) to prevent further oligomerization of p62 and to increase the availability of functional free monomeric p62. We also found that monomeric p62 could also interact with ubiquitinated proteins and that the forced dimerization of p62 was sufficient to increase the interactions with ubiquitinated proteins, probably polyubiquitinated with K63 linkages. Upon exposure to As(III), (1) inability to form oligomeric p62 because of a mutation in the PB1 dimerization domain, or (2) reduced capability to generate monomeric p62 owing to diminished polyubiquitination of p62 itself, resulted in reduced viability of cells. Therefore, upon exposure to As(III), p62 initially needs to form oligomers to activate an antioxidant response pathway. Subsequently, p62 is polyubiquitinated to prevent further oligomerization and ensure the availability of free p62 monomers. We propose that the polyubiquitination of p62 under exposure to As(III) plays an important role in overcoming the impaired autophagic flux by regulating the oligomerization status of p62.


Asunto(s)
Arsenitos/toxicidad , Autofagia/efectos de los fármacos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo/efectos de los fármacos , Proteína Sequestosoma-1/genética , Animales , Supervivencia Celular/efectos de los fármacos , Embrión de Mamíferos , Fibroblastos , Regulación de la Expresión Génica , Células HEK293 , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Cultivo Primario de Células , Pliegue de Proteína/efectos de los fármacos , Multimerización de Proteína , Proteína Sequestosoma-1/metabolismo , Transducción de Señal , Ubiquitinación
12.
PLoS Genet ; 10(12): e1004861, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25474007

RESUMEN

The kinase PINK1 and the E3 ubiquitin (Ub) ligase Parkin participate in mitochondrial quality control. The phosphorylation of Ser65 in Parkin's ubiquitin-like (UBl) domain by PINK1 stimulates Parkin activation and translocation to damaged mitochondria, which induces mitophagy generating polyUb chain. However, Parkin Ser65 phosphorylation is insufficient for Parkin mitochondrial translocation. Here we report that Ser65 in polyUb chain is also phosphorylated by PINK1, and that phosphorylated polyUb chain on mitochondria tethers Parkin at mitochondria. The expression of Tom70MTS-4xUb SE, which mimics phospho-Ser65 polyUb chains on the mitochondria, activated Parkin E3 activity and its mitochondrial translocation. An E3-dead form of Parkin translocated to mitochondria with reduced membrane potential in the presence of Tom70(MTS)-4xUb SE, whereas non-phospho-polyUb mutant Tom70(MTS)-4xUb SA abrogated Parkin translocation. Parkin binds to the phospho-polyUb chain through its RING1-In-Between-RING (IBR) domains, but its RING0-linker is also required for mitochondrial translocation. Moreover, the expression of Tom70(MTS)-4xUb SE improved mitochondrial degeneration in PINK1-deficient, but not Parkin-deficient, Drosophila. Our study suggests that the phosphorylation of mitochondrial polyUb by PINK1 is implicated in both Parkin activation and mitochondrial translocation, predicting a chain reaction mechanism of mitochondrial phospho-polyUb production by which rapid translocation of Parkin is achieved.


Asunto(s)
Mitocondrias/metabolismo , Poliubiquitina/metabolismo , Proteínas Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Animales Modificados Genéticamente , Células Cultivadas , Drosophila , Células HEK293 , Células HeLa , Humanos , Ratones , Fosforilación , Unión Proteica , Transporte de Proteínas , Ubiquitinación
13.
Biochim Biophys Acta ; 1853(5): 996-1009, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25701757

RESUMEN

The polyubiquitin gene Ubc is upregulated under oxidative stress induced by arsenite [As(III)]. However, the detailed mechanism of Ubc upregulation and the exact role of ubiquitin (Ub) to protect cells against As(III)-induced toxicity remain unknown. Here, we found that Ubc-/- mouse embryonic fibroblasts (MEFs) exhibited reduced viability under As(III) exposure, although the Nrf2-Keap1 pathway was activated as a cytoprotective response. Intriguingly, due to the reduced polyubiquitination and delayed onset of degradation of Nrf2 in Ubc-/- MEFs, the basal expression levels of Nrf2 target genes were elevated. As(III)-induced accumulation of Ub conjugates occurred in an Nrf2-independent manner, probably due to cellular stress conditions, including reduced proteasomal activity. Increased cellular Ub levels were essential to polyubiquitinate misfolded proteins generated under As(III) exposure and to degrade them by the proteasome. However, when cellular Ub levels decreased, these misfolded proteins were not efficiently polyubiquitinated, but rather accumulated as large protein aggregates inside the cells, causing cytotoxicity. Furthermore, increased activity of the autophagic pathway to clear these aggregates was not observed in Ubc-/- MEFs. Therefore, reduced viability of Ubc-/- MEFs under As(III) exposure may not be due to dysregulation of the Nrf2-Keap1 pathway, but mostly to reduced efficacy to polyubiquitinate and degrade misfolded protein aggregates.


Asunto(s)
Arsenitos/toxicidad , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Eliminación de Gen , Poliubiquitina/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Proteína 1 Asociada A ECH Tipo Kelch , Ratones , Modelos Biológicos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Poliubiquitina/metabolismo , Pliegue de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ubiquitina/metabolismo , Ubiquitinación/efectos de los fármacos
14.
Biochem Biophys Res Commun ; 453(3): 443-8, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25280998

RESUMEN

Disruption of the polyubiquitin gene Ubb leads to hypothalamic neurodegeneration and metabolic disorders, including obesity and sleep abnormalities, in mice. However, it has yet to be determined whether or not these neural phenotypes in Ubb(-/-) mice are directly caused by cell autonomous defects in maintaining proper levels of ubiquitin (Ub). To directly demonstrate that reduced levels of Ub are sufficient to cause neuronal abnormalities, we investigated the characteristics of cultured neurons isolated from Ubb(-/-) mouse embryonic brains. We found that neuronal morphology, neurite outgrowth, and synaptic development were significantly impaired in Ubb(-/-) neurons. Furthermore, we observed the growth of astrocytes in Ubb(-/-) cell cultures despite the fact that cells were cultured under conditions promoting neuronal growth. When the reduced levels of free Ub, but not Ub conjugates, in Ubb(-/-) cells were restored to those of wild-type cells by providing exogenous Ub via lentivirus-mediated delivery, the increased apoptosis observed in Ubb(-/-) cells was almost completely abolished. Ectopic expression of Ub also improved neuronal and glial phenotypes observed in Ubb(-/-) cells. Therefore, our study suggests that Ub homeostasis, or the maintenance of cellular free Ub above certain threshold levels, is essential for proper neuronal development and survival.


Asunto(s)
Neuronas/citología , Ubiquitina/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Ratones , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Ubiquitina/genética , Ubiquitina/fisiología
15.
Biochem Biophys Res Commun ; 435(3): 434-40, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23669039

RESUMEN

We have previously demonstrated that disruption of polyubiquitin gene Ubc leads to mid-gestation embryonic lethality most likely due to a defect in fetal liver development, which can be partially rescued by ectopic expression of Ub. In a previous study, we assessed the cause of embryonic lethality with respect to the fetal liver hematopoietic system. We confirmed that Ubc(-/-) embryonic lethality could not be attributed to impaired function of hematopoietic stem cells, which raises the question of whether or not FLECs such as hepatocytes and bile duct cells, the most abundant cell types in the liver, are affected by disruption of Ubc and contribute to embryonic lethality. To answer this, we isolated FLCs from E13.5 embryos and cultured them in vitro. We found that proliferation capacity of Ubc(-/-) cells was significantly reduced compared to that of control cells, especially during the early culture period, however we did not observe the increased number of apoptotic cells. Furthermore, levels of Ub conjugate, but not free Ub, decreased upon disruption of Ubc expression in FLCs, and this could not be compensated for by upregulation of other poly- or mono-ubiquitin genes. Intriguingly, the highest Ubc expression levels throughout the entire culture period were observed in bipotent FLEPCs. Hepatocytes and bipotent FLEPCs were most affected by disruption of Ubc, resulting in defective proliferation as well as reduced cell numbers in vitro. These results suggest that defective proliferation of these cell types may contribute to severe reduction of fetal liver size and potentially mid-gestation lethality of Ubc(-/-) embryos.


Asunto(s)
Células Madre Embrionarias/patología , Hepatocitos/patología , Hígado/embriología , Células Madre Multipotentes/patología , Ubiquitina C/deficiencia , Ubiquitina C/genética , Animales , Conductos Biliares Intrahepáticos/citología , Conductos Biliares Intrahepáticos/embriología , Conductos Biliares Intrahepáticos/metabolismo , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Hepatocitos/metabolismo , Queratina-19/metabolismo , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Células Madre Multipotentes/metabolismo , Tamaño de los Órganos/genética , Tamaño de los Órganos/fisiología , alfa-Fetoproteínas/metabolismo
16.
Nature ; 448(7154): 704-8, 2007 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-17687326

RESUMEN

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by expansion of CAG triplet repeats in the huntingtin (HTT) gene (also called HD) and characterized by accumulation of aggregated fragments of polyglutamine-expanded HTT protein in affected neurons. Abnormal enrichment of HD inclusion bodies with ubiquitin, a diagnostic characteristic of HD and many other neurodegenerative disorders including Alzheimer's and Parkinson's diseases, has suggested that dysfunction in ubiquitin metabolism may contribute to the pathogenesis of these diseases. Because modification of proteins with polyubiquitin chains regulates many essential cellular processes including protein degradation, cell cycle, transcription, DNA repair and membrane trafficking, disrupted ubiquitin signalling is likely to have broad consequences for neuronal function and survival. Although ubiquitin-dependent protein degradation is impaired in cell-culture models of HD and of other neurodegenerative diseases, it has not been possible to evaluate the function of the ubiquitin-proteasome system (UPS) in HD patients or in animal models of the disease, and a functional role for UPS impairment in neurodegenerative disease pathogenesis remains controversial. Here we exploit a mass-spectrometry-based method to quantify polyubiquitin chains and demonstrate that the abundance of these chains is a faithful endogenous biomarker of UPS function. Lys 48-linked polyubiquitin chains accumulate early in pathogenesis in brains from the R6/2 transgenic mouse model of HD, from a knock-in model of HD and from human HD patients, establishing that UPS dysfunction is a consistent feature of HD pathology. Lys 63- and Lys 11-linked polyubiquitin chains, which are not typically associated with proteasomal targeting, also accumulate in the R6/2 mouse brain. Thus, HD is linked to global changes in the ubiquitin system to a much greater extent than previously recognized.


Asunto(s)
Enfermedad de Huntington/metabolismo , Ubiquitina/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/patología , Cuerpos de Inclusión/metabolismo , Lisina/metabolismo , Ratones , Ratones Transgénicos , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo
17.
Exp Mol Med ; 55(10): 2138-2146, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37779143

RESUMEN

Glial cell activation precedes neuronal cell death during brain aging and the progression of neurodegenerative diseases. Under neuroinflammatory stress conditions, lipocalin-2 (LCN2), also known as neutrophil gelatinase-associated lipocalin or 24p3, is produced and secreted by activated microglia and reactive astrocytes. Lcn2 expression levels are known to be increased in various cells, including reactive astrocytes, through the activation of the NF-κB signaling pathway. In the central nervous system, as LCN2 exerts neurotoxicity when secreted from reactive astrocytes, many researchers have attempted to identify various strategies to inhibit LCN2 production, secretion, and function to minimize neuroinflammation and neuronal cell death. These strategies include regulation at the transcriptional, posttranscriptional, and posttranslational levels, as well as blocking its functions using neutralizing antibodies or antagonists of its receptor. The suppression of NF-κB signaling is a strategy to inhibit LCN2 production, but it may also affect other cellular activities, raising questions about its effectiveness and feasibility. Recently, LCN2 was found to be a target of the autophagy‒lysosome pathway. Therefore, autophagy activation may be a promising therapeutic strategy to reduce the levels of secreted LCN2 and overcome neurodegenerative diseases. In this review, we focused on research progress on astrocyte-derived LCN2 in the central nervous system.


Asunto(s)
Lipocalinas , Enfermedades Neurodegenerativas , Humanos , Lipocalina 2/genética , Lipocalina 2/metabolismo , Lipocalinas/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Gliosis , FN-kappa B/metabolismo , Inflamación
18.
Autophagy ; 19(8): 2296-2317, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36781380

RESUMEN

LCN2/neutrophil gelatinase-associated lipocalin/24p3 (lipocalin 2) is a secretory protein that acts as a mammalian bacteriostatic molecule. Under neuroinflammatory stress conditions, LCN2 is produced and secreted by activated microglia and reactive astrocytes, resulting in neuronal apoptosis. However, it remains largely unknown whether inflammatory stress and neuronal loss can be minimized by modulating LCN2 production and secretion. Here, we first demonstrated that LCN2 was secreted from reactive astrocytes, which were stimulated by treatment with lipopolysaccharide (LPS) as an inflammatory stressor. Notably, we found two effective conditions that led to the reduction of induced LCN2 levels in reactive astrocytes: proteasome inhibition and macroautophagic/autophagic flux activation. Mechanistically, proteasome inhibition suppresses NFKB/NF-κB activation through NFKBIA/IκBα stabilization in primary astrocytes, even under inflammatory stress conditions, resulting in the downregulation of Lcn2 expression. In contrast, autophagic flux activation via MTOR inhibition reduced the intracellular levels of LCN2 through its pre-secretory degradation. In addition, we demonstrated that the N-terminal signal peptide of LCN2 is critical for its secretion and degradation, suggesting that these two pathways may be mechanistically coupled. Finally, we observed that LPS-induced and secreted LCN2 levels were reduced in the astrocyte-cultured medium under the above-mentioned conditions, resulting in increased neuronal viability, even under inflammatory stress.Abbreviations: ACM, astrocyte-conditioned medium; ALP, autophagy-lysosome pathway; BAF, bafilomycin A1; BTZ, bortezomib; CHX, cycloheximide; CNS, central nervous system; ER, endoplasmic reticulum; GFAP, glial fibrillary acidic protein; GFP, green fluorescent protein; JAK, Janus kinase; KD, knockdown; LCN2, lipocalin 2; LPS, lipopolysaccharide; MACS, magnetic-activated cell sorting; MAP1LC3/LC3, microtubule-associated protein 1 light chain 3; MTOR, mechanistic target of rapamycin kinase; NFKB/NF-κB, nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105; NFKBIA/IκBα, nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha; OVEX, overexpression; SLC22A17, solute carrier family 22 member 17; SP, signal peptide; SQSTM1, sequestosome 1; STAT3, signal transducer and activator of transcription 3; TNF/TNF-α, tumor necrosis factor; TUBA, tubulin, alpha; TUBB3/ß3-TUB, tubulin, beta 3 class III; UB, ubiquitin; UPS, ubiquitin-proteasome system.


Asunto(s)
Lipocalinas , FN-kappa B , Animales , Lipocalinas/genética , Lipocalinas/metabolismo , Lipocalinas/farmacología , Lipocalina 2/metabolismo , Lipocalina 2/farmacología , FN-kappa B/metabolismo , Astrocitos/metabolismo , Tubulina (Proteína)/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , Inhibidor NF-kappaB alfa/farmacología , Lipopolisacáridos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Autofagia , Sistema Nervioso Central/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Mamíferos/metabolismo
19.
Biochem Biophys Res Commun ; 418(3): 541-6, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22285186

RESUMEN

Previously, we demonstrated that disruption of polyubiquitin gene Ubb leads to hypothalamic neurodegeneration and metabolic abnormalities associated with hypothalamic dysfunction. However, we cannot exclude the possibility that defects in other brain regions where Ubb is highly expressed may also contribute to the phenotypes exhibited by Ubb(-/-) mice. Upon searching for such brain regions, we identified a region in the brainstem called the locus coeruleus where both polyubiquitin genes Ubb and Ubc were highly expressed. In contrast to other brain regions, Ubc was significantly upregulated in the locus coeruleus of Ubb(-/-) mice presumably to compensate for loss of Ubb, and this upregulation was sufficient to maintain levels of free Ub, but not total Ub, in the locus coeruleus. However, in the hypothalamus of Ubb(-/-) mice, both free and total Ub levels significantly decreased. This discrepancy resulted in completely different phenotypic outcomes between the two different brain regions. While we have reported dysfunction and degeneration of hypothalamic neurons in adult Ubb(-/-) mice, there were no signs of functional impairment or degeneration in the locus coeruleus neurons, suggesting that the maintenance of free Ub above threshold levels could be an important mechanism for neuronal protection. Accordingly, we propose that, upon stress induced by disruption of Ubb, neuronal vulnerability may be determined based on the ability of neurons or neighboring cells to maintain free Ub levels for the protection of neuronal function and survival.


Asunto(s)
Locus Coeruleus/metabolismo , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Poliubiquitina/metabolismo , Ubiquitina/metabolismo , Animales , Supervivencia Celular , Locus Coeruleus/anomalías , Locus Coeruleus/patología , Ratones , Ratones Mutantes , Degeneración Nerviosa/patología , Neuronas/patología , Poliubiquitina/genética , Ubiquitina/genética , Ubiquitina C/genética , Ubiquitina C/metabolismo
20.
J Hazard Mater ; 439: 129576, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-35850071

RESUMEN

Nanoplastics (NPs) are derived from microplastics and may cause health problems. We previously showed that 100 nm polystyrene (PS)-NPs enter cells, including mouse embryonic fibroblasts (MEFs), and their intracellular accumulation induces inflammatory and oxidative stress. Moreover, PS-NP uptake was found to occur via endocytosis, and they accumulated mostly at the juxtanuclear position, but never within the nucleus. We speculated that PS-NPs were cleared from cells when they were no longer exposed to PS-NPs. However, the effects of PS-NPs on the cellular machinery remain unknown. The accumulation of PS-NPs at the juxtanuclear position may be due to retrograde transport along microtubules. To confirm this, we treated PS-NP-exposed MEFs with inhibitors of histone deacetylase 6 (HDAC6), dynein, or microtubule polymerization and found greatly diminished intracellular and juxtanuclear accumulation. Moreover, rapid clearance of PS-NPs was observed when MEFs were treated with an HDAC6 inhibitor. PS-NPs were removed by exocytosis, as confirmed by treatment with an exocytosis inhibitor. Furthermore, inhibiting the retrograde transport of PS-NPs alleviated the activation of the antioxidant response pathway, inflammatory and oxidative stress, and reactive oxygen species generation. In summary, inhibition of the retrograde transport of non-biodegradable PS-NPs leads to their rapid export by exocytosis, which may reduce their cytotoxicity.


Asunto(s)
Nanopartículas , Contaminantes Químicos del Agua , Animales , Exocitosis , Fibroblastos , Ratones , Microplásticos , Plásticos , Poliestirenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA