Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Discov ; 13(3): 702-723, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36445254

RESUMEN

LZTR1 is the substrate-specific adaptor of a CUL3-dependent ubiquitin ligase frequently mutated in sporadic and syndromic cancer. We combined biochemical and genetic studies to identify LZTR1 substrates and interrogated their tumor-driving function in the context of LZTR1 loss-of-function mutations. Unbiased screens converged on EGFR and AXL receptor tyrosine kinases as LZTR1 interactors targeted for ubiquitin-dependent degradation in the lysosome. Pathogenic cancer-associated mutations of LZTR1 failed to promote EGFR and AXL degradation, resulting in dysregulated growth factor signaling. Conditional inactivation of Lztr1 and Cdkn2a in the mouse nervous system caused tumors in the peripheral nervous system including schwannoma-like tumors, thus recapitulating aspects of schwannomatosis, the prototype tumor predisposition syndrome sustained by LZTR1 germline mutations. Lztr1- and Cdkn2a-deleted tumors aberrantly accumulated EGFR and AXL and exhibited specific vulnerability to EGFR and AXL coinhibition. These findings explain tumorigenesis by LZTR1 inactivation and offer therapeutic opportunities to patients with LZTR1-mutant cancer. SIGNIFICANCE: EGFR and AXL are substrates of LZTR1-CUL3 ubiquitin ligase. The frequent somatic and germline mutations of LZTR1 in human cancer cause EGFR and AXL accumulation and deregulated signaling. LZTR1-mutant tumors show vulnerability to concurrent inhibition of EGFR and AXL, thus providing precision targeting to patients affected by LZTR1-mutant cancer. This article is highlighted in the In This Issue feature, p. 517.


Asunto(s)
Neurilemoma , Factores de Transcripción , Animales , Humanos , Ratones , Carcinogénesis , Transformación Celular Neoplásica , Receptores ErbB/genética , Mutación , Neurilemoma/genética , Neurilemoma/metabolismo , Neurilemoma/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitinas/genética
2.
Nat Commun ; 13(1): 2089, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440621

RESUMEN

Tissue-specific transcriptional activity is silenced in mitotic cells but it remains unclear whether the mitotic regulatory machinery interacts with tissue-specific transcriptional programs. We show that such cross-talk involves the controlled interaction between core subunits of the anaphase-promoting complex (APC) and the ID2 substrate. The N-terminus of ID2 is independently and structurally compatible with a pocket composed of core APC/C subunits that may optimally orient ID2 onto the APCCDH1 complex. Phosphorylation of serine-5 by CDK1 prevented the association of ID2 with core APC, impaired ubiquitylation and stabilized ID2 protein at the mitosis-G1 transition leading to inhibition of basic Helix-Loop-Helix (bHLH)-mediated transcription. The serine-5 phospho-mimetic mutant of ID2 that inefficiently bound core APC remained stable during mitosis, delayed exit from mitosis and reloading of bHLH transcription factors on chromatin. It also locked cells into a "mitotic stem cell" transcriptional state resembling the pluripotent program of embryonic stem cells. The substrates of APCCDH1 SKP2 and Cyclin B1 share with ID2 the phosphorylation-dependent, D-box-independent interaction with core APC. These results reveal a new layer of control of the mechanism by which substrates are recognized by APC.


Asunto(s)
Anafase , Proteínas de Ciclo Celular , Ciclosoma-Complejo Promotor de la Anafase/genética , Proteínas de Ciclo Celular/metabolismo , Mitosis , Serina
3.
Nat Cancer ; 2(2): 141-156, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33681822

RESUMEN

The transcriptomic classification of glioblastoma (GBM) has failed to predict survival and therapeutic vulnerabilities. A computational approach for unbiased identification of core biological traits of single cells and bulk tumors uncovered four tumor cell states and GBM subtypes distributed along neurodevelopmental and metabolic axes, classified as proliferative/progenitor, neuronal, mitochondrial and glycolytic/plurimetabolic. Each subtype was enriched with biologically coherent multiomic features. Mitochondrial GBM was associated with the most favorable clinical outcome. It relied exclusively on oxidative phosphorylation for energy production, whereas the glycolytic/plurimetabolic subtype was sustained by aerobic glycolysis and amino acid and lipid metabolism. Deletion of the glucose-proton symporter SLC45A1 was the truncal alteration most significantly associated with mitochondrial GBM, and the reintroduction of SLC45A1 in mitochondrial glioma cells induced acidification and loss of fitness. Mitochondrial, but not glycolytic/plurimetabolic, GBM exhibited marked vulnerability to inhibitors of oxidative phosphorylation. The pathway-based classification of GBM informs survival and enables precision targeting of cancer metabolism.


Asunto(s)
Glioblastoma , Glioma , Glioblastoma/genética , Glioma/metabolismo , Glucólisis/genética , Humanos , Mitocondrias/genética , Fosforilación Oxidativa
4.
Nat Commun ; 11(1): 6236, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33262409

RESUMEN

A Correction to this paper has been published: https://doi.org/10.1038/s41467-020-20178-0.

5.
Mol Cell ; 79(3): 376-389.e8, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32640193

RESUMEN

Activation of dual-specificity tyrosine-phosphorylation-regulated kinases 1A and 1B (DYRK1A and DYRK1B) requires prolyl hydroxylation by PHD1 prolyl hydroxylase. Prolyl hydroxylation of DYRK1 initiates a cascade of events leading to the release of molecular constraints on von Hippel-Lindau (VHL) ubiquitin ligase tumor suppressor function. However, the proline residue of DYRK1 targeted by hydroxylation and the role of prolyl hydroxylation in tyrosine autophosphorylation of DYRK1 are unknown. We found that a highly conserved proline in the CMGC insert of the DYRK1 kinase domain is hydroxylated by PHD1, and this event precedes tyrosine autophosphorylation. Mutation of the hydroxylation acceptor proline precludes tyrosine autophosphorylation and folding of DYRK1, resulting in a kinase unable to preserve VHL function and lacking glioma suppression activity. The consensus proline sequence is shared by most CMGC kinases, and prolyl hydroxylation is essential for catalytic activation. Thus, formation of prolyl-hydroxylated intermediates is a novel mechanism of kinase maturation and likely a general mechanism of regulation of CMGC kinases in eukaryotes.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Isoenzimas/genética , Prolina/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Cristalografía por Rayos X , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Glioma/patología , Células HEK293 , Xenoinjertos , Humanos , Hidroxilación , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Isoenzimas/química , Isoenzimas/metabolismo , Ratones , Ratones Desnudos , Proteína Quinasa 14 Activada por Mitógenos/química , Proteína Quinasa 14 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Modelos Moleculares , Mutación , Neuroglía/metabolismo , Neuroglía/patología , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Secundaria de Proteína , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Quinasas DyrK
7.
Nat Commun ; 9(1): 3404, 2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-30143610

RESUMEN

AMP-activated protein kinase (AMPK) plays a key role in controlling energy metabolism in response to physiological and nutritional status. Although AMPK activation has been proposed as a promising molecular target for treating obesity and its related comorbidities, the use of pharmacological AMPK activators has been met with contradictory therapeutic challenges. Here we show a regulatory mechanism for AMPK through its ubiquitination and degradation by the E3 ubiquitin ligase makorin ring finger protein 1 (MKRN1). MKRN1 depletion promotes glucose consumption and suppresses lipid accumulation due to AMPK stabilisation and activation. Accordingly, MKRN1-null mice show chronic AMPK activation in both liver and adipose tissue, resulting in significant suppression of diet-induced metabolic syndrome. We demonstrate also its therapeutic effect by administering shRNA targeting MKRN1 into obese mice that reverses non-alcoholic fatty liver disease. We suggest that ubiquitin-dependent AMPK degradation represents a target therapeutic strategy for metabolic disorders.


Asunto(s)
Síndrome Metabólico/metabolismo , Ribonucleoproteínas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Animales , Dieta Alta en Grasa/efectos adversos , Hígado Graso/genética , Hígado Graso/metabolismo , Femenino , Hígado/metabolismo , Hígado/patología , Masculino , Síndrome Metabólico/genética , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ribonucleoproteínas/genética , Ubiquitina-Proteína Ligasas/genética
8.
Mol Cells ; 41(5): 381-389, 2018 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-29665672

RESUMEN

ARF is a tumor suppressor protein that has a pivotal role in the prevention of cancer development through regulating cell proliferation, senescence, and apoptosis. As a factor that induces senescence, the role of ARF as a tumor suppressor is closely linked to the p53-MDM2 axis, which is a key process that restrains tumor formation. Thus, many cancer cells either lack a functional ARF or p53, which enables them to evade cell oncogenic stress-mediated cycle arrest, senescence, or apoptosis. In particular, the ARF gene is a frequent target of genetic and epigenetic alterations including promoter hyper-methylation or gene deletion. However, as many cancer cells still express ARF, pathways that negatively modulate transcriptional or post-translational regulation of ARF could be potentially important means for cancer cells to induce cellular proliferation. These recent findings of regulators affecting ARF protein stability along with its low levels in numerous human cancers indicate the significance of an ARF post-translational mechanism in cancers. Novel findings of regulators stimulating or suppressing ARF function would provide new therapeutic targets to manage cancer- and senescence-related diseases. In this review, we present the current knowledge on the regulation and alterations of ARF expression in human cancers, and indicate the importance of regulators of ARF as a prognostic marker and in potential therapeutic strategies.


Asunto(s)
Transformación Celular Neoplásica/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Proteínas de Neoplasias/fisiología , Proteína p14ARF Supresora de Tumor/fisiología , Apoptosis , División Celular , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Genes p16 , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Proteína p14ARF Supresora de Tumor/deficiencia , Proteína p14ARF Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/fisiología , Ubiquitinación
9.
Cell Death Differ ; 25(6): 1050-1062, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29472714

RESUMEN

Oncogene-induced senescence (OIS) is a critical tumor-suppressor mechanism, which prevents hyper-proliferation and transformation of cells. c-Myc promotes OIS through the transcriptional activation of p14ARF followed by p53 activation. Although the oncogene-mediated transcriptional regulation of p14ARF has been well addressed, the post-translational modification of p14ARF regulated by oncogenic stress has yet to be investigated. Here, we found that c-Myc increased p14ARF protein stability by inducing the transcription of ubiquitin-specific protease 10 (USP10). USP10, in turn, mediated the deubiquitination of p14ARF, preventing its proteasome-dependent degradation. USP10-null mouse embryonic fibroblasts and human primary cells depleted of USP10 bypassed c-Myc-induced senescence via the destabilization of p14ARF, and these cells displayed accelerated hyper-proliferation and transformation. Clinically the c-Myc-USP10-p14ARF axis was disrupted in non-small cell lung cancer patients, resulting in significantly worse overall survival. Our studies indicate that USP10 induced by c-Myc has a crucial role in OIS by maintaining the stability of key tumor suppressor p14ARF.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Senescencia Celular , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Células MCF-7 , Ratones , Ratones Noqueados , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Proteína p14ARF Supresora de Tumor/genética , Ubiquitina Tiolesterasa/genética
10.
Cancer Res ; 77(2): 343-354, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27793846

RESUMEN

The tumor suppressor function of p14ARF is regulated at a posttranslational level via mechanisms yet to be fully understood. Here, we report the identification of an unconventional p14ARF degradation pathway induced by the chaperone HSP90 in association with the E3 ubiquitin ligase C-terminus of HSP70-interacting protein (CHIP). The ternary complex of HSP90, CHIP, and p14ARF was required to induce the lysosomal degradation of p14ARF by an ubiquitination-independent but LAMP2A-dependent mechanism. Depletion of HSP90 or CHIP induced p14ARF-dependent senescence in human fibroblasts. Premature senescence observed in cells genetically deficient in CHIP was rescued in cells that were doubly deficient in CHIP and p14ARF. Notably, non-small cell lung cancer cells (NSCLC) positive for p14ARF were sensitive to treatment with the HSP90 inhibitor geldanamycin. Furthermore, overexpression of HSP90 and CHIP with a concomitant loss of p14ARF correlated with poor prognosis in patients with NSCLC. Our findings identify a relationship between p14ARF and its chaperones that suggest new therapeutic strategies in cancers that overexpress HSP90. Cancer Res; 77(2); 343-54. ©2016 AACR.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Senescencia Celular/fisiología , Proteínas HSP90 de Choque Térmico/metabolismo , Neoplasias Pulmonares/patología , Proteína p14ARF Supresora de Tumor/metabolismo , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos de Riesgos Proporcionales , Ubiquitina-Proteína Ligasas/metabolismo
11.
BMB Rep ; 49(11): 598-606, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27470213

RESUMEN

ARF is an alternative reading frame product of the INK4a/ARF locus, inactivated in numerous human cancers. ARF is a key regulator of cellular senescence, an irreversible cell growth arrest that suppresses tumor cell growth. It functions by sequestering MDM2 (a p53 E3 ligase) in the nucleolus, thus activating p53. Besides MDM2, ARF has numerous other interacting partners that induce either cellular senescence or apoptosis in a p53-independent manner. This further complicates the dynamics of the ARF network. Expression of ARF is frequently disrupted in human cancers, mainly due to epigenetic and transcriptional regulation. Vigorous studies on various transcription factors that either positively or negatively regulate ARF transcription have been carried out. However, recent focus on posttranslational modifications, particularly ubiquitination, indicates wider dynamic controls of ARF than previously known. In this review, we discuss the role and dynamic regulation of ARF in senescence and cancer. [BMB Reports 2016; 49(11): 598-606].


Asunto(s)
Envejecimiento/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Neoplasias/patología , Factor 1 de Ribosilacion-ADP , Animales , Apoptosis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Sitios Genéticos , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
12.
Nat Commun ; 6: 7769, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26183061

RESUMEN

The activity of the phosphatase and tensin homologue (PTEN) is known to be suppressed via post-translational modification. However, the mechanism and physiological significance by which post-translational modifications lead to PTEN suppression remain unclear. Here we demonstrate that PTEN destabilization is induced by EGFR- or oncogenic PI3K mutation-mediated AKT activation in cervical cancer. EGFR/PI3K/AKT-mediated ubiquitination and degradation of PTEN are dependent on the MKRN1 E3 ligase. These processes require the stabilization of MKRN1 via AKT-mediated phosphorylation. In cervical cancer patients with high levels of pAKT and MKRN1 expression, PTEN protein levels are low and correlate with a low 5-year survival rate. Taken together, our results demonstrate that PI3K/AKT signals enforce positive-feedback regulation by suppressing PTEN function.


Asunto(s)
Carcinoma/genética , Regulación Neoplásica de la Expresión Génica , Proteínas del Tejido Nervioso/metabolismo , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ribonucleoproteínas/metabolismo , Displasia del Cuello del Útero/genética , Neoplasias del Cuello Uterino/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Carcinogénesis/genética , Carcinoma/metabolismo , Carcinoma/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular , Receptores ErbB/metabolismo , Retroalimentación Fisiológica , Femenino , Células HeLa , Humanos , Inmunohistoquímica , Técnicas In Vitro , Mutación , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas , Fosforilación , Pronóstico , Procesamiento Proteico-Postraduccional , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina-Treonina Quinasas TOR/metabolismo , Ubiquitinación , Neoplasias del Cuello Uterino/metabolismo , Displasia del Cuello del Útero/metabolismo
13.
J Org Chem ; 78(19): 9975-80, 2013 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-23984866

RESUMEN

Rhodium-catalyzed asymmetric 1,4-addition of arylboronic acids to α,ß-unsaturated carbonyl compounds was achieved at temperatures below 0 °C using a Rh/MeO-F12-BIPHEP catalyst. The reaction of cyclohexenone or N-R-maleimide with arylboronic acids proceeded even at -80 °C in the presence of the Rh catalyst. In the latter case, high enantioselectivity was observed because a low-temperature method was used, regardless of the type of substituent on maleimide.

14.
J Natl Cancer Inst ; 104(21): 1660-72, 2012 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-23104211

RESUMEN

BACKGROUND: We investigated whether Makorin ring finger protein 1 (MKRN1), an E3 ligase, affects p14ARF-associated cellular senescence and tumorigenesis by posttranslational modification in gastric tumorigenesis. METHODS: A link between MKRN1 and ARF was examined in MKRN1 null mouse embryonic fibroblasts (MEFs) and in human fibroblasts and gastric cancer cells by silencing MKRN1 using small interfering RNA (siRNA) and short hairpin RNA (shRNA). Ubiquitination and proteasomal degradation assays were used to assess p14ARF degradation associated with MKRN1. MKRN1 and p14ARF expression levels were analyzed with immunohistochemistry in malignant and normal tissues from gastric cancer patients and with χ(2) tests. The tumor growth of gastric cancer cells stably expressing MKRN1 shRNA, p14ARF shRNA, or both was examined in mouse xenograft models (n = 4-6) and analyzed with unpaired t tests. All statistical tests were two-sided. RESULTS: MKRN1 knockout MEFs exhibited premature senescence and growth retardation with increased p19ARF protein expression. Similar results were obtained for human fibroblasts or gastric cancer cell lines by MKRN1 knockdown. Biochemical analyses confirmed that MKRN1 targets p14ARF for ubiquitination and subsequent proteasome-dependent degradation. A statistically significant association was shown between MKRN1 overexpression and p14ARF underexpression (P = .016). Xenograft analyses using p53-functional AGS or -dysfunctional SNU601 cells displayed statistically significant tumor growth retardation by silencing MKRN1, which was reversed under depletion of p14ARF (AGS cells, MKRN1 knockdown tumors vs MKRN1 and p14ARF knockdown tumors: 164.6 vs 464.8mm(3), difference = 300.2mm(3), 95% CI = 189.1 to 411.3mm(3), P < .001). CONCLUSIONS: We demonstrated that MKRN1 functions as a novel E3 ligase of p14ARF and that it potentially regulates cellular senescence and tumorigenesis in gastric cancer.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Senescencia Celular , Proteínas del Tejido Nervioso/metabolismo , Ribonucleoproteínas/metabolismo , Neoplasias Gástricas/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Ratones Desnudos , Proteínas del Tejido Nervioso/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Biosíntesis de Proteínas , ARN Interferente Pequeño , Ribonucleoproteínas/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Neoplasias Gástricas/fisiopatología , Análisis de Matrices Tisulares , Trasplante Heterólogo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación
15.
Nat Commun ; 3: 978, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22864571

RESUMEN

Fas-associated protein with death domain (FADD) is a pivotal component of death receptor-mediated extrinsic apoptosis and necroptosis. Here we show that FADD is regulated by Makorin Ring Finger Protein 1 (MKRN1) E3 ligase-mediated ubiquitination and proteasomal degradation. MKRN1 knockdown results in FADD protein stabilization and formation of the rapid death-inducing signalling complex, which causes hypersensitivity to extrinsic apoptosis by facilitating caspase-8 and caspase-3 cleavage in response to death signals. We also show that MKRN1 and FADD are involved in the regulation of necrosome formation and necroptosis upon caspase inhibition. Downregulation of MKRN1 results in severe defects of tumour growth upon tumour necrosis factor-related apoptosis-inducing ligand treatment in a xenograft model using MDA-MB-231 breast cancer cells. Suppression of tumour growth by MKRN1 depletion is relieved by simultaneous FADD knockdown. Our data reveal a novel mechanism by which fas-associated protein with death domain is regulated via an ubiquitination-induced degradation pathway.


Asunto(s)
Apoptosis/fisiología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Necrosis/metabolismo , Receptores de Muerte Celular/metabolismo , Ubiquitinación/fisiología , Animales , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Proteína de Dominio de Muerte Asociada a Fas/genética , Citometría de Flujo , Células HeLa , Humanos , Inmunoprecipitación , Técnicas In Vitro , Ratones , Necrosis/genética , Receptores de Muerte Celular/genética , Análisis de Matrices Tisulares , Ubiquitinación/genética
17.
J Virol ; 84(1): 426-36, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19846531

RESUMEN

West Nile virus capsid protein (WNVCp) displays pathogenic toxicity via the apoptotic pathway. However, a cellular mechanism protective against this toxic effect has not been observed so far. Here, we identified Makorin ring finger protein 1 (MKRN1) as a novel E3 ubiquitin ligase for WNVCp. The cytotoxic effects of WNVCp as well as its expression levels were inhibited in U2OS cells that stably expressed MKRN1. Immunoprecipitation analyses revealed an interaction between MKRN1 and WNVCp. Domain analysis indicated that the C terminus of MKRN1 and the N terminus of WNVCp were required for the interaction. MKRN1 could induce WNVCp ubiquitination and degradation in a proteasome-dependent manner. Interestingly, the WNVCp mutant with amino acids 1 to 105 deleted WNVCp was degraded by MKRN1, whereas the mutant with amino acids 1 to 90 deleted was not. When three lysine sites at positions 101, 103, and 104 of WNVCp were replaced with alanine, MKRN1-mediated ubiquitination and degradation of the mutant were significantly inhibited, suggesting that these sites are required for the ubiquitination. Finally, U2OS cell lines stably expressing MKRN1 were resistant to cytotoxic effects of WNV. In contrast, cells depleted of MKRN1 were more susceptible to WNVCp cytotoxicity. Confirming this, overexpression of MKRN1 significantly reduced, but depletion of MKRN1 increased, WNV proliferation in 293T cells. Taken together, our results suggest that MKRN1 can protect cells from WNV by inducing WNVCp degradation.


Asunto(s)
Proteínas de la Cápside/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ribonucleoproteínas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Virus del Nilo Occidental/patogenicidad , Sustitución de Aminoácidos , Sitios de Unión , Proteínas de la Cápside/fisiología , Línea Celular Tumoral , Humanos , Lisina , Proteínas del Tejido Nervioso/genética , Complejo de la Endopetidasa Proteasomal , Ribonucleoproteínas/genética , Ubiquitinación , Virus del Nilo Occidental/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA