Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Mol Cell ; 54(4): 626-38, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24768535

RESUMEN

In response to DNA damage, PCNA is mono-ubiquitinated and triggers translesion DNA synthesis (TLS) by recruiting polymerase-η. However, it remained unknown how error-prone TLS is turned off after DNA lesion bypass to prevent mutagenesis. Here we showed that ISG15 modification (ISGylation) of PCNA plays a key role in TLS termination. Upon UV irradiation, EFP, an ISG15 E3 ligase, bound to mono-ubiquitinated PCNA and promoted its ISGylation. ISGylated PCNA then tethered USP10 for deubiquitination and in turn the release of polymerase-η from PCNA. Eventually, PCNA was deISGylated by UBP43 for reloading of replicative DNA polymerases and resuming normal DNA replication. However, ISGylation-defective Lys-to-Arg mutations in PCNA or knockdown of any of ISG15, EFP, or USP10 led to persistent recruitment of mono-ubiquitinated PCNA and polymerase-η to nuclear foci, causing an increase in mutation frequency. These findings establish a crucial role of PCNA ISGylation in termination of error-prone TLS for preventing excessive mutagenesis.


Asunto(s)
Citocinas/metabolismo , Daño del ADN , Replicación del ADN , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitinas/metabolismo , Arginina/metabolismo , Sitios de Unión/genética , Citocinas/genética , ADN Polimerasa II/metabolismo , Reparación del ADN , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Lisina/metabolismo , Mutagénesis , Tasa de Mutación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Ubiquitinas/genética
2.
Biochem Biophys Res Commun ; 433(2): 194-9, 2013 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-23454125

RESUMEN

CHFR ubiquitin ligase plays an important role in cell cycle progression and tumorigenesis. CHFR tumor suppressor function is highly associated with its protein level. We recently reported that CHFR protein levels are negatively regulated by SUMOylation-mediated proteasomal degradation. In the present study, we uncover a detailed molecular mechanism how SUMOylation promotes CHFR destabilization. We demonstrate that SUMO modification of CHFR promotes its ubiquitylation and subsequent proteasomal degradation. However, SUMOylation of CHFR does not affect its auto-ubiquitylation, which generally serves as a maintenance mechanism for most ubiquitin ligases. Moreover, the E3 ubiquitin ligase activity of CHFR is dispensable for this SUMOylation-mediated ubiquitylation and degradation. Conversely, SENP2 deSUMOylating enzyme reduces SUMOylation-induced ubiquitylation of CHFR, leading to elevated CHFR protein levels. Taken together, our results present a new regulatory mechanism for CHFR that sequential post-translational modifications of CHFR by SUMO and ubiquitin coordinately regulates its stability.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Sumoilación , Ubiquitinación , Proteínas de Ciclo Celular/genética , Células HeLa , Humanos , Proteínas de Neoplasias/genética , Proteínas de Unión a Poli-ADP-Ribosa , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Proteína SUMO-1/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
3.
Biochem Biophys Res Commun ; 430(1): 213-7, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23131550

RESUMEN

CHFR ubiquitin ligase acts as a checkpoint upon DNA damage and its functional inactivation is one of key characteristics of tumor development and metastasis. Despite the crucial role in maintaining genome integrity and cell cycle progression, little is known how CHFR stability is regulated. Here, we showed that CHFR is covalently modified by SUMO-1 at lysine 663 and subsequently destabilized by ubiquitin-proteasome system. While CHFR(K663R) substitution mutation does not alter its subcellular localization, SUMOylation-defective CHFR(K663R)-stable cells exhibit substantial growth suppression due to the increased stability of CHFR(K663R). Moreover, protein level of CHFR, not CHFR(K663R), is rapidly declined under SUMOylation-promoting conditions, and SENP2 deSUMOylating enzyme reverses its SUMO-modification. Collectively, we demonstrated that CHFR stability is regulated by SUMOylation-dependent proteasomal degradation. Therefore, our study underscores the importance of CHFR SUMOylation as a new regulatory mechanism of CHFR and highlights the emerging role of SUMOylation in modulating protein stability.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Sumoilación , Proteínas de Ciclo Celular/genética , Cisteína Endopeptidasas/metabolismo , Células HeLa , Humanos , Lisina/genética , Lisina/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Unión a Poli-ADP-Ribosa , Estabilidad Proteica , Proteolisis , Ubiquitina-Proteína Ligasas
4.
Nat Cell Biol ; 8(12): 1424-31, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17086174

RESUMEN

The p53 tumour suppressor has a key role in the control of cell growth and differentiation, and in the maintenance of genome integrity. p53 is kept labile under normal conditions, but in response to stresses, such as DNA damage, it accumulates in the nucleus for induction of cell-cycle arrest, DNA repair or apoptosis. Mdm2 is an ubiquitin ligase that promotes p53 ubiquitination and degradation. Mdm2 is also self-ubiquitinated and degraded. Here, we identified a novel cascade for the increase in p53 level in response to DNA damage. A new SUMO-specific protease, SUSP4, removed SUMO-1 from Mdm2 and this desumoylation led to promotion of Mdm2 self-ubiquitination, resulting in p53 stabilization. Moreover, SUSP4 competed with p53 for binding to Mdm2, also resulting in p53 stabilization. Overexpression of SUSP4 inhibited cell growth, whereas knockdown of susp4 by RNA interference (RNAi) promoted of cell growth. UV damage induced SUSP4 expression, leading to an increase in p53 levels in parallel with a decrease in Mdm2 levels. These findings establish a new mechanism for the elevation of cellular p53 levels in response to UV damage.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo , Animales , Procesos de Crecimiento Celular/efectos de la radiación , Cisteína Endopeptidasas/genética , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Células 3T3 NIH , Unión Proteica/efectos de la radiación , Transporte de Proteínas/efectos de la radiación , Proteínas Proto-Oncogénicas c-mdm2/deficiencia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Termodinámica , Proteína p53 Supresora de Tumor/deficiencia , Rayos Ultravioleta
5.
Rapid Commun Mass Spectrom ; 27(2): 339-46, 2013 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23239382

RESUMEN

RATIONALE: Protein ubiquitination plays a critical role in regulating many cellular events, such as protein localization and stability, cellular signal transduction and DNA repair. Recent studies have shown that polyubiquitin (polyUb) chains elongate through heterogeneous isopeptide linkages to K11, K29, K48 and K63. In this study we have investigated the usage of isopeptide linkages of polyUb chains in different molecular weight regions by using quantitative mass spectrometry. METHODS: Recombinant Chfr protein was autoubiquitinated by E1 enzyme, E2 enzyme UbcH5 and ubiquitin (WT Ub, K11R Ub, K48R Ub and K63R Ub) in vitro, and different molecular weight regions of ubiquitinated Chfr were then subjected to liquid chromatography coupled with tandem mass spectrometry (LC/MS/MS) following sodium dodecyl sulfate/polyacrylamide gel electrophoresis (SDS-PAGE) and in-gel digestion. RESULTS: Absolute QUantitative Analysis (AQUA) of polyUb chain formation with wild-type (WT) and point mutants of ubiquitin was performed, and the results suggested that the K11 polyUb chain was most frequently used in the high ubiquitin conjugates of WT Ub. Furthermore, the extent of polyUb chain formation with K11R Ub was decreased about 10-fold compared to polyUb chain formation with WT Ub through the entire molecular weight region. The present study suggests that the linkage through K11 plays crucial roles in polyUb chain formation. CONCLUSIONS: Topologies of polyUb chains in the low and high Ub conjugates were studied using mass spectrometry. K48 and K63 were the primary ubiquitination sites of the low molecular weight Ub conjugates, whereas K11 was the critical site of polyUb chain formation in high molecular weight Ub conjugates.


Asunto(s)
Lisina/química , Lisina/metabolismo , Poliubiquitina/química , Poliubiquitina/metabolismo , Espectrometría de Masas en Tándem/métodos , Ubiquitinación/fisiología , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Cromatografía Líquida de Alta Presión/métodos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
6.
Biochem Biophys Res Commun ; 418(3): 512-7, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22285184

RESUMEN

The murine SWI/SNF-like BAF complex is an ATP-dependent chromatin remodeling complex that functions as a transcriptional regulator in cell proliferation, differentiation and development. The SWI/SNF-like BAF complex consists of several components including core subunits such as BRG1, BAF155/SRG3, BAF47/SNF5/INI1, and BAF170. We have previously shown that the interaction between SRG3/mBAF155 and other components of the complex stabilizes them by attenuating their proteasomal degradation. However, it has not been known how the major components of the SWI/SNF-like BAF complex such as BRG1, SNF5, and BAF60a are targeted for the ubiquitination and degradation, and how SRG3/mBAF155 protects them from the degradation process. Here we report that CHFR interacts with BRG1, SNF5, and BAF60a of the SWI/SNF-like BAF complex and ubiquitinates them to target for degradation through a proteasome-mediated pathway, and that SRG3/mBAF155 stabilizes these components by blocking their interaction with CHFR.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Animales , Células COS , Chlorocebus aethiops , ADN Helicasas/metabolismo , Estabilidad de Enzimas , Humanos , Ratones , Proteínas Nucleares/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa , Proteolisis , Proteína SMARCB1 , Factores de Transcripción/genética
7.
Nature ; 441(7097): 1162-6, 2006 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-16672981

RESUMEN

Parkinson's disease is the second most common neurodegenerative disorder and is characterized by the degeneration of dopaminergic neurons in the substantia nigra. Mitochondrial dysfunction has been implicated as an important trigger for Parkinson's disease-like pathogenesis because exposure to environmental mitochondrial toxins leads to Parkinson's disease-like pathology. Recently, multiple genes mediating familial forms of Parkinson's disease have been identified, including PTEN-induced kinase 1 (PINK1; PARK6) and parkin (PARK2), which are also associated with sporadic forms of Parkinson's disease. PINK1 encodes a putative serine/threonine kinase with a mitochondrial targeting sequence. So far, no in vivo studies have been reported for pink1 in any model system. Here we show that removal of Drosophila PINK1 homologue (CG4523; hereafter called pink1) function results in male sterility, apoptotic muscle degeneration, defects in mitochondrial morphology and increased sensitivity to multiple stresses including oxidative stress. Pink1 localizes to mitochondria, and mitochondrial cristae are fragmented in pink1 mutants. Expression of human PINK1 in the Drosophila testes restores male fertility and normal mitochondrial morphology in a portion of pink1 mutants, demonstrating functional conservation between human and Drosophila Pink1. Loss of Drosophila parkin shows phenotypes similar to loss of pink1 function. Notably, overexpression of parkin rescues the male sterility and mitochondrial morphology defects of pink1 mutants, whereas double mutants removing both pink1 and parkin function show muscle phenotypes identical to those observed in either mutant alone. These observations suggest that pink1 and parkin function, at least in part, in the same pathway, with pink1 functioning upstream of parkin. The role of the pink1-parkin pathway in regulating mitochondrial function underscores the importance of mitochondrial dysfunction as a central mechanism of Parkinson's disease pathogenesis.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Mitocondrias/fisiología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Drosophila melanogaster/enzimología , Drosophila melanogaster/fisiología , Epistasis Genética , Prueba de Complementación Genética , Humanos , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Longevidad/genética , Longevidad/fisiología , Masculino , Mitocondrias/patología , Músculos/metabolismo , Músculos/patología , Mutación/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Fenotipo , Transporte de Proteínas , Espermátides/metabolismo , Espermátides/patología , Ubiquitina-Proteína Ligasas
8.
J Biol Chem ; 284(48): 33475-84, 2009 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-19801685

RESUMEN

HslVU is a bacterial ATP-dependent protease distantly related to eukaryotic proteasomes consisting of hexameric HslU ATPase and dodecameric HslV protease. As a homolog of the 20 S proteasome beta-subunits, HslV also uses the N-terminal threonine as the active site residue. However, unlike the proteasome that has only 6 active sites among the 14 beta-subunits, HslV has 12 active sites that could potentially contribute to proteolytic activity. Here, by using a series of HslV dodecamers containing different numbers of active sites, we demonstrate that like the proteasome, HslV with only approximately 6 active sites is sufficient to support full catalytic activity. However, a further reduction of the number of active sites leads to a proportional decrease in activity. Using proteasome inhibitors, we also demonstrate that substrate-mediated stabilization of the HslV-HslU interaction remains unchanged until the number of the active sites is decreased to approximately 6 but is gradually compromised upon further reduction. These results with a mathematical model suggest HslVU utilizes no more than 6 active sites at any given time, presumably because of the action of HslU. These results also suggest that each ATP-bound HslU subunit activates one HslV subunit and that substrate bound to the HslV active site stimulates the HslU ATPase activity by stabilizing the HslV-HslU interaction. We propose this mechanism plays an important role in supporting complete degradation of substrates while preventing wasteful ATP hydrolysis in the resting state by controlling the interaction between HslV and HslU through the catalytic engagement of the proteolytic active sites.


Asunto(s)
Adenosina Trifosfato/metabolismo , Endopeptidasa Clp/metabolismo , Proteínas de Escherichia coli/metabolismo , Treonina/metabolismo , Adenosina Trifosfato/análogos & derivados , Alanina/genética , Alanina/metabolismo , Sustitución de Aminoácidos , Sitios de Unión , Catálisis , Electroforesis en Gel de Poliacrilamida , Endopeptidasa Clp/genética , Pruebas de Enzimas , Proteínas de Escherichia coli/genética , Hidrólisis , Cinética , Modelos Biológicos , Mutación , Péptidos/metabolismo , Unión Proteica , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Espectrometría de Fluorescencia , Especificidad por Sustrato , Treonina/genética
9.
Biochem Biophys Res Commun ; 395(4): 515-20, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20388495

RESUMEN

CHFR functions as a mitotic checkpoint by delaying entry into metaphase in response to mitotic stress. CHFR is frequently silenced by hypermethylation in human cancers, indicating that CHFR is a tumor suppressor. To further elucidate the role of CHFR in tumorigenesis, we studied the relationship between CHFR and a novel CHFR-interacting protein, HLTF, helicase-like transcription factor. Here we show that CHFR binds to and ubiquitinates HLTF, leading to its degradation. HLTF modulates basal expression of PAI-1 involved in regulation of cell migration. Consistently, overexpression of CHFR inhibits cell migration, resulting from reduced HLTF followed by decreased PAI-1 expression. HLTF expression is also higher in human breast cancer cells where CHFR is not expressed. Taken together, this is the first report identifying the regulatory mechanism of HLTF by CHFR, suggesting that CHFR-mediated downregulation of HLTF may help protect against cancer.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas de Neoplasias/fisiología , Neoplasias/enzimología , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Línea Celular Tumoral , Células HeLa , Humanos , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica
10.
Biol Open ; 8(8)2019 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-31362948

RESUMEN

Cilia are extended from mother centrioles in quiescent G0/G1 cells and retracted in dividing cells. Diverse post-translational modifications play roles in the assembly and disassembly of the cilium. Here, we examined class I histone deacetylases (HDACs) as positive regulators of cilia assembly in serum-deprived RPE1 and HK2 cells. We observed that the number of cells with cilia was significantly reduced in HDAC3- and HDAC8-depleted cells. The ciliary length also decreased in HDAC3- and HDAC8-depleted cells compared to that in control cells. A knockdown-rescue experiment showed that wild-type HDAC3 and HDAC8 rescued the cilia assembly and ciliary length in HDAC3- and HDAC8-depleted cells, respectively; however, deacetylase-dead HDAC3 and HDAC8 mutants did not. This suggests that deacetylase activity is critical for both HDAC3 and HDAC8 function in cilia assembly and ciliary length control. This is the first study to report that HDACs are required for the assembly and elongation of the primary cilia.

11.
Biochem Biophys Res Commun ; 377(3): 946-51, 2008 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-18952062

RESUMEN

The intracellular negatively regulatory mechanism which affects IL-1beta-induced MUC8 gene expression remains unclear. We found that SOCS3 overexpression suppressed IL-1beta-induced MUC8 gene expression in NCI-H292 cells, whereas silencing of SOCS3 restored IL-1beta-induced MUC8 gene expression. Sequentially activated ERK1/2, RSK1, and CREB by IL-1beta were not affected by SOCS3, indicating that SOCS3 has an independent mechanism of action. Using immunoprecipitaion and nano LC mass analysis, we found that SOCS3 bound NonO (non-POU-domain containing, octamer-binding domain protein) in the absence of IL-1beta, whereas IL-1beta treatment dissociated the direct binding of SOCS3 and NonO. A dominant-negative SOCS3 mutant (Y204F/Y221F) did not bind to NonO. Interestingly, SOCS3 overexpression dramatically suppressed MUC8 gene expression in cells transfected with wild-type or siRNA of NonO. Moreover, silencing of SOCS3 dramatically increased NonO-mediated MUC8 gene expression caused by IL-1beta compared to NonO overexpression alone, suggesting that SOCS3 acts as a suppressor by regulating the action of NonO.


Asunto(s)
Interleucina-1beta/metabolismo , Mucinas/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Factores de Transcripción de Octámeros/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Activación Transcripcional , Línea Celular , Proteínas de Unión al ADN , Expresión Génica , Humanos , Interleucina-1beta/genética , Proteínas Asociadas a Matriz Nuclear/genética , Factores de Transcripción de Octámeros/genética , Proteínas de Unión al ARN/genética , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/antagonistas & inhibidores , Proteínas Supresoras de la Señalización de Citocinas/genética , Ubiquitinación
12.
Cell Host Microbe ; 23(3): 338-352.e5, 2018 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-29503179

RESUMEN

DUOX, a member of the NADPH oxidase family, acts as the first line of defense against enteric pathogens by producing microbicidal reactive oxygen species. DUOX is activated upon enteric infection, but the mechanisms regulating DUOX activity remain incompletely understood. Using Drosophila genetic tools, we show that enteric infection results in "pro-catabolic" signaling that initiates metabolic reprogramming of enterocytes toward lipid catabolism, which ultimately governs DUOX homeostasis. Infection induces signaling cascades involving TRAF3 and kinases AMPK and WTS, which regulate TOR kinase to control the balance of lipogenesis versus lipolysis. Enhancing lipogenesis blocks DUOX activity, whereas stimulating lipolysis via ATG1-dependent lipophagy is required for DUOX activation. Drosophila with altered activity in TRAF3-AMPK/WTS-ATG1 pathway components exhibit abolished infection-induced lipolysis, reduced DUOX activation, and enhanced susceptibility to enteric infection. Thus, this work uncovers signaling cascades governing inflammation-induced metabolic reprogramming and provides insight into the pathophysiology of immune-metabolic interactions in the microbe-laden gut epithelia.


Asunto(s)
Sistema Digestivo/inmunología , Drosophila/inmunología , Oxidasas Duales/metabolismo , Interacciones Huésped-Patógeno/inmunología , Inflamación/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Sistemas CRISPR-Cas/genética , Técnicas de Cultivo de Célula , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Enterocitos/metabolismo , Femenino , Microbioma Gastrointestinal , Edición Génica , Regulación de la Expresión Génica , Homeostasis , Inmunidad Innata , Metabolismo de los Lípidos , Lipólisis , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Masculino , Pectobacterium carotovorum/patogenicidad , Especies Reactivas de Oxígeno/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/metabolismo
13.
Mol Cells ; 23(2): 252-7, 2007 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-17464204

RESUMEN

Escherichia coli HslVU is an ATP-dependent protease consisting of two heat shock proteins, the HslU ATPase and HslV peptidase. In the reconstituted enzyme, HslU stimulates the proteolytic activity of HslV by one to two orders of magnitude, while HslV increases the rate of ATP hydrolysis by HslU several-fold. Here we show that HslV alone can efficiently degrade certain unfolded proteins, such as unfolded lactalbumin and lysozyme prepared by complete reduction of disulfide bonds, but not their native forms. Furthermore, HslV alone cleaved a lactalbumin fragment sandwiched by two thioredoxin molecules, indicating that it can hydrolyze the internal peptide bonds of lactalbumin. Surprisingly, ATP inhibited the degradation of unfolded proteins by HslV. This inhibitory effect of ATP was markedly diminished by substitution of the Arg86 residue located in the apical pore of HslV with Gly, suggesting that interaction of ATP with the Arg residue blocks access of unfolded proteins to the proteolytic chamber of HslV. These results suggest that uncomplexed HslV is inactive under normal conditions, but may can degrade unfolded proteins when the ATP level is low, as it is during carbon starvation.


Asunto(s)
Adenosina Trifosfato/farmacología , Endopeptidasas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Pliegue de Proteína , Proteasas ATP-Dependientes/genética , Proteasas ATP-Dependientes/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Carbono/metabolismo , Endopeptidasas/genética , Activación Enzimática , Proteínas de Escherichia coli/genética , Proteínas de Choque Térmico/metabolismo , Hidrólisis , Lactalbúmina/metabolismo , Datos de Secuencia Molecular , Muramidasa/metabolismo
14.
Sci Rep ; 6: 37578, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27883020

RESUMEN

SIRT1, the NAD+-dependent protein deacetylase, controls cell-cycle progression and apoptosis by suppressing p53 tumour suppressor. Although SIRT1 is known to be phosphorylated by JNK1 upon oxidative stress and subsequently down-regulated, it still remains elusive how SIRT1 stability and activity are controlled. Here, we have unveiled that CHFR functions as an E3 Ub-ligase of SIRT1, responsible for its proteasomal degradation under oxidative stress conditions. CHFR interacts with and destabilizes SIRT1 by ubiquitylation and subsequent proteolysis. Such CHFR-mediated SIRT1 inhibition leads to the increase of p53 acetylation and its target gene transcription. Notably, CHFR facilitates SIRT1 destabilization when SIRT1 is phosphorylated by JNK1 upon oxidative stress, followed by prominent apoptotic cell death. Meanwhile, JNK inhibitor prevents SIRT1 phosphorylation, leading to elevated SIRT1 protein levels even in the presence of H2O2. Taken together, our results indicate that CHFR plays a crucial role in the cellular stress response pathway by controlling the stability and function of SIRT1.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteínas de Neoplasias/genética , Estrés Oxidativo/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Sirtuina 1/genética , Ubiquitina-Proteína Ligasas/genética , Acetilación/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas de Ciclo Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Peróxido de Hidrógeno/toxicidad , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/metabolismo
15.
Nat Commun ; 7: 12513, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27545325

RESUMEN

p53 plays a pivotal role in tumour suppression under stresses, such as DNA damage. ISG15 has been implicated in the control of tumorigenesis. Intriguingly, the expression of ISG15, UBE1L and UBCH8 is induced by DNA-damaging agents, such as ultraviolet and doxorubicin, which are known to induce p53. Here, we show that the genes encoding ISG15, UBE1L, UBCH8 and EFP, have the p53-responsive elements and their expression is induced in a p53-dependent fashion under DNA damage conditions. Furthermore, DNA damage induces ISG15 conjugation to p53 and this modification markedly enhances the binding of p53 to the promoters of its target genes (for example, CDKN1 and BAX) as well as of its own gene by promoting phosphorylation and acetylation, leading to suppression of cell growth and tumorigenesis. These findings establish a novel feedback circuit between p53 and ISG15-conjugating system for positive regulation of the tumour suppressive function of p53 under DNA damage conditions.


Asunto(s)
Carcinogénesis/patología , Citocinas/genética , Daño del ADN/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinas/genética , Acetilación , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citocinas/metabolismo , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Doxorrubicina/farmacología , Humanos , Fosforilación , Regiones Promotoras Genéticas/genética , Enzimas Activadoras de Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinas/metabolismo , Rayos Ultravioleta/efectos adversos , Proteína X Asociada a bcl-2/metabolismo
16.
Cancer Res ; 76(4): 855-65, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26781991

RESUMEN

The generation of reactive oxygen species (ROS) is required for proper cell signaling, but must be tightly regulated to minimize deleterious oxidizing effects. Activation of the NADPH oxidases (Nox) triggers ROS production and, thus, regulatory mechanisms exist to properly control Nox activity. In this study, we report a novel mechanism in which Nox1 activity is regulated through the proteasomal degradation of Nox organizer 1 (NoxO1). We found that through the interaction between NoxO1 and growth receptor-bound protein 2 (Grb2), the Casitas B-lineage lymphoma (Cbl) E3 ligase was recruited, leading to decreased NoxO1 stability and a subsequent reduction in ROS generation upon epidermal growth factor (EGF) stimulation. Additionally, we show that EGF-mediated phosphorylation of NoxO1 induced its release from Grb2 and facilitated its association with Nox activator 1 (NoxA1) to stimulate ROS production. Consistently, overexpression of Grb2 resulted in decreased Nox1 activity, whereas knockdown of Grb2 led to increased Nox1 activity in response to EGF. CRISPR/Cas9-mediated NoxO1 knockout in human colon cancer cells abrogated anchorage-independent growth on soft agar and tumor-forming ability in athymic nude mice. Moreover, the expression and stability of NoxO1 were significantly increased in human colon cancer tissues compared with normal colon. Taken together, these results support a model whereby Nox1 activity and ROS generation are regulated by Grb2/Cbl-mediated proteolysis of NoxO1 in response to EGF, providing new insight into the processes by which excessive ROS production may promote oncogenic signaling to drive colorectal tumorigenesis.


Asunto(s)
Neoplasias del Colon/genética , Proteína Adaptadora GRB2/genética , NADH NADPH Oxidorreductasas/metabolismo , Animales , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Humanos , Ratones , NADH NADPH Oxidorreductasas/genética , NADPH Oxidasa 1 , Especies Reactivas de Oxígeno , Transducción de Señal , Transfección
17.
Nat Commun ; 6: 6314, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25692647

RESUMEN

The Hippo pathway plays crucial roles in regulating organ size and stem cell homeostasis. Although the signalling cascade of the core Hippo kinases is relatively well understood, little is known about the mechanisms that modulate the activity of the Hippo pathway. Here, we report identification of NEDD4, a HECT-type E3 ubiquitin ligase, as a regulatory component of the Hippo pathway. We demonstrate that NEDD4 ubiquitylates and destabilizes WW45 and LATS kinase, both of which are required for active Hippo signalling. Interestingly, MST1 protects WW45, but not LATS2, against NEDD4. We also provide evidence indicating that NEDD4 inactivation at high cell density is a prerequisite for the elevated Hippo activity linked to contact inhibition. Moreover, NEDD4 promotes intestinal stem cell renewal in Drosophila by suppressing Hippo signalling. Collectively, we present a regulatory mechanism by which NEDD4 controls the Hippo pathway leading to coordinated cell proliferation and apoptosis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Tracto Gastrointestinal/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Drosophila melanogaster/fisiología , Regulación de la Expresión Génica , Células HEK293 , Factor de Crecimiento de Hepatocito/metabolismo , Homeostasis , Humanos , Ratones , Células 3T3 NIH , Ubiquitina-Proteína Ligasas Nedd4 , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , Transducción de Señal , Ubiquitina/metabolismo
18.
Neoplasia ; 17(6): 518-24, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26152360

RESUMEN

c-Cbl, a RING-type ubiquitin E3 ligase, downregulates various receptor tyrosine kinases (e.g., epidermal growth factor receptor (EGFR)), leading to inhibition of cell proliferation. Moreover, patients with myeloid neoplasm frequently harbor c-Cbl mutations, implicating the role of c-Cbl as a tumor suppressor. Recently, we have shown that c-Cbl downregulates αPix-mediated cell migration and invasion, and the lack of c-Cbl in the rat C6 and human A172 glioma cells is responsible for their malignant behavior. Here, we showed that c-Cbl exon skipping occurs in the glioma cells and the brain tissues from glioblastoma patients lacking c-Cbl. This exon skipping resulted in generation of two types of c-Cbl isoforms: type I lacking exon-9 and type II lacking exon-9 and exon-10. However, the c-Cbl isoforms in the cells and tissues could not be detected as they were rapidly degraded by proteasome. Consequently, C6 and A172 cells showed sustained EGFR activation. However, no splice site mutation was found in the region from exon-7 to exon-11 of the c-Cbl gene in C6 cells and a glioblastoma tissue lacking c-Cbl. In addition, c-Cbl exon skipping could be induced when cells transfected with a c-Cbl mini-gene were grown to high density or under hypoxic stress. These results suggest that unknown alternations (e.g., mutation) of splicing machinery in C6 and A172 cells and the glioblastoma brain tissues are responsible for the deleterious exon skipping. Collectively, these findings indicate that the c-Cbl exon skipping contributes to human glioma and its malignant behavior.


Asunto(s)
Exones , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/patología , Hipoxia/fisiopatología , Proteínas Proto-Oncogénicas c-cbl/genética , Animales , Western Blotting , Receptores ErbB/genética , Humanos , Inmunoprecipitación , Mutación/genética , ARN Mensajero/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Elementos Reguladores de la Transcripción , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
19.
Diabetes ; 64(7): 2420-31, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25784542

RESUMEN

Small ubiquitin-like modifier (SUMO)-specific proteases (SENPs) that reverse protein modification by SUMO are involved in the control of numerous cellular processes, including transcription, cell division, and cancer development. However, the physiological function of SENPs in energy metabolism remains unclear. Here, we investigated the role of SENP2 in fatty acid metabolism in C2C12 myotubes and in vivo. In C2C12 myotubes, treatment with saturated fatty acids, like palmitate, led to nuclear factor-κB-mediated increase in the expression of SENP2. This increase promoted the recruitment of peroxisome proliferator-activated receptor (PPAR)δ and PPARγ, through desumoylation of PPARs, to the promoters of the genes involved in fatty acid oxidation (FAO), such as carnitine-palmitoyl transferase-1 (CPT1b) and long-chain acyl-CoA synthetase 1 (ACSL1). In addition, SENP2 overexpression substantially increased FAO in C2C12 myotubes. Consistent with the cell culture system, muscle-specific SENP2 overexpression led to a marked increase in the mRNA levels of CPT1b and ACSL1 and thereby in FAO in the skeletal muscle, which ultimately alleviated high-fat diet-induced obesity and insulin resistance. Collectively, these data identify SENP2 as an important regulator of fatty acid metabolism in skeletal muscle and further implicate that muscle SENP2 could be a novel therapeutic target for the treatment of obesity-linked metabolic disorders.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Ácidos Grasos/metabolismo , Músculo Esquelético/metabolismo , Animales , Carnitina O-Palmitoiltransferasa/genética , Células Cultivadas , Coenzima A Ligasas/genética , Cisteína Endopeptidasas/genética , Humanos , Resistencia a la Insulina , Ratones , Fibras Musculares Esqueléticas/metabolismo , FN-kappa B/fisiología , Oxidación-Reducción , PPAR delta/fisiología , PPAR gamma/fisiología , Ácido Palmítico/farmacología
20.
J Mol Biol ; 425(16): 2940-54, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23707406

RESUMEN

In all cells, ATP-dependent proteases play central roles in the controlled degradation of short-lived regulatory or misfolded proteins. A hallmark of these enzymes is that proteolytic active sites are sequestered within a compartmentalized space, which is accessible to substrates only when they are fed into the cavity by protein-unfolding ATPases. HslVU is a prototype of such enzymes, consisting of the hexameric HslU ATPase and the dodecameric HslV protease. HslV forms a barrel-shaped proteolytic chamber with two constricted axial pores. Here, we report that structural alterations of HslV's pore motif dramatically affect the proteolytic activities of both HslV and HslVU complexes. Mutations of a conserved pore residue in HslV (Leu88 to Ala, Gly, or Ser) led to a tighter binding between HslV and HslU and a dramatic stimulation of both the proteolytic and ATPase activities. Furthermore, the HslV mutants alone showed a marked increase of basal hydrolytic activities toward small peptides and unstructured proteins. A synthetic peptide of the HslU C-terminal tail further stimulated the proteolytic activities of these mutants, even allowing degradation of certain folded proteins in the absence of HslU. Moreover, expression of the L88A mutant in Escherichia coli inhibited cell growth, suggesting that HslV pore mutations dysregulate the protease through relaxing the pore constriction, which normally prevents essential cellular proteins from random degradation. Consistent with these observations, an X-ray crystal structure shows that the pore loop of L88A-HslV is largely disordered. Collectively, these results suggest that substrate degradation by HslV is controlled by gating of its pores.


Asunto(s)
Endopeptidasa Clp/genética , Endopeptidasa Clp/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Cristalografía por Rayos X , Endopeptidasa Clp/química , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Modelos Moleculares , Proteínas Mutantes/química , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Proteolisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA