Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Mol Cell ; 81(22): 4722-4735.e5, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34626566

RESUMEN

Rapid protein degradation enables cells to quickly modulate protein abundance. Dysregulation of short-lived proteins plays essential roles in disease pathogenesis. A focused map of short-lived proteins remains understudied. Cycloheximide, a translational inhibitor, is widely used in targeted studies to measure degradation kinetics for short-lived proteins. Here, we combined cycloheximide chase assays with advanced quantitative proteomics to map short-lived proteins under translational inhibition in four human cell lines. Among 11,747 quantified proteins, we identified 1,017 short-lived proteins (half-lives ≤ 8 h). These short-lived proteins are less abundant, evolutionarily younger, and less thermally stable than other proteins. We quantified 103 proteins with different stabilities among cell lines. We showed that U2OS and HCT116 cells express truncated forms of ATRX and GMDS, respectively, which have lower stability than their full-length counterparts. This study provides a large-scale resource of human short-lived proteins under translational arrest, leading to untapped avenues of protein regulation for therapeutic interventions.


Asunto(s)
Proteínas/química , Proteoma , Proteómica/métodos , Alanina/análogos & derivados , Alanina/química , Línea Celular , Línea Celular Tumoral , Cicloheximida/química , Cicloheximida/farmacología , Fucosa/química , Geminina/química , Células HCT116 , Células HEK293 , Humanos , Péptidos/química , Análisis de Componente Principal , Biosíntesis de Proteínas , Proteínas/efectos de los fármacos , Control de Calidad , ARN Interferente Pequeño/metabolismo , Telómero/química
2.
Mol Cell ; 81(21): 4509-4526.e10, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34560002

RESUMEN

The interferon (IFN) pathway is critical for cytotoxic T cell activation, which is central to tumor immunosurveillance and successful immunotherapy. We demonstrate here that PKCλ/ι inactivation results in the hyper-stimulation of the IFN cascade and the enhanced recruitment of CD8+ T cells that impaired the growth of intestinal tumors. PKCλ/ι directly phosphorylates and represses the activity of ULK2, promoting its degradation through an endosomal microautophagy-driven ubiquitin-dependent mechanism. Loss of PKCλ/ι results in increased levels of enzymatically active ULK2, which, by direct phosphorylation, activates TBK1 to foster the activation of the STING-mediated IFN response. PKCλ/ι inactivation also triggers autophagy, which prevents STING degradation by chaperone-mediated autophagy. Thus, PKCλ/ι is a hub regulating the IFN pathway and three autophagic mechanisms that serve to maintain its homeostatic control. Importantly, single-cell multiplex imaging and bioinformatics analysis demonstrated that low PKCλ/ι levels correlate with enhanced IFN signaling and good prognosis in colorectal cancer patients.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Interferones/metabolismo , Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal , Adulto , Anciano , Anciano de 80 o más Años , Animales , Autofagia , Linfocitos T CD8-positivos/metabolismo , Carcinogénesis , Transformación Celular Neoplásica , Neoplasias Colorrectales/mortalidad , Cicloheximida/química , Femenino , Células HEK293 , Humanos , Inmunofenotipificación , Factor 3 Regulador del Interferón/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Persona de Mediana Edad , Trasplante de Neoplasias , Fosforilación , Pronóstico , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción , Regulación hacia Arriba
3.
Nat Prod Rep ; 37(6): 752-762, 2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32428051

RESUMEN

Covering: 2000 to 2020 The translation of mRNA into proteins is a precisely regulated, complex process that can be divided into three main stages, i.e. initiation, elongation, termination, and recycling. This contribution is intended to highlight how natural products interfere with the elongation phase of eukaryotic protein biosynthesis. Cycloheximide, isolated from Streptomyces griseus, has long been the prototype inhibitor of eukaryotic translation elongation. In the last three decades, a variety of natural products from different origins were discovered to also address the elongation step in different manners, including interference with the elongation factors eEF1 and eEF2 as well as binding to A-, P- or E-sites of the ribosome itself. Recent advances in the crystallization of the ribosomal machinery together with natural product inhibitors allowed characterizing similarities as well as differences in their mode of action. Since aberrations in protein synthesis are commonly observed in tumors, and malfunction or overexpression of translation factors can cause cellular transformation, the protein synthesis machinery has been realized as an attractive target for anticancer drugs. The therapeutic use of the first natural products that reached market approval, plitidepsin (Aplidin®) and homoharringtonine (Synribo®), will be introduced. In addition, we will highlight two other potential indications for translation elongation inhibitors, i.e. viral infections and genetic disorders caused by premature termination of translation.


Asunto(s)
Productos Biológicos/química , Productos Biológicos/farmacología , Extensión de la Cadena Peptídica de Translación/efectos de los fármacos , Antineoplásicos/farmacología , Cicloheximida/química , Cicloheximida/farmacología , Humanos , Extensión de la Cadena Peptídica de Translación/fisiología , Factor 1 de Elongación Peptídica/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Ribosomas/metabolismo
4.
Angew Chem Int Ed Engl ; 58(16): 5387-5391, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30802354

RESUMEN

Cycloheximide (CHX) is an inhibitor of eukaryotic translation elongation that has played an essential role in the study of protein synthesis. Despite its ubiquity, few studies have been directed towards accessing synthetic CHX derivatives, even though such efforts may lead to protein synthesis inhibitors with improved or alternate properties. Described here is the total synthesis of CHX and analogues, and the establishment of structure-activity relationships (SAR) responsible for translation inhibition. The SAR studies aided the design of more potent compounds, one of which irreversibly blocks ribosomal elongation, preserves polysome profiles, and may be a broadly useful tool for investigating protein synthesis.


Asunto(s)
Productos Biológicos/farmacología , Cicloheximida/farmacología , Células Eucariotas/efectos de los fármacos , Ribosomas/efectos de los fármacos , Productos Biológicos/síntesis química , Productos Biológicos/química , Cicloheximida/síntesis química , Cicloheximida/química , Relación Dosis-Respuesta a Droga , Células Eucariotas/metabolismo , Conformación Molecular , Biosíntesis de Proteínas/efectos de los fármacos , Ribosomas/metabolismo , Relación Estructura-Actividad
5.
Cancer Res ; 78(21): 6121-6133, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30217930

RESUMEN

The sodium iodide symporter (NIS) is a classical iodide pump typically localized within the cell plasma membrane in thyroid cells, where NIS expression is believed to ensure success of mainstay radioiodide therapy in thyroid cancers. Although radioiodide uptake is generally reduced in thyroid cancer tissue, intracellular nonmembranous NIS has been reported to increase, suggesting that NIS serves a pump-independent function. Thyroid cancer is one of the major component cancers of Cowden syndrome, a subset of which is caused by germline mutations in PTEN In this study, we explored the noncanonical tumorigenic role of NIS in thyroid cancer cells in relation to PTEN signaling. PTEN knockdown in thyroid cancer cell lines stabilized intracellular NIS protein by promoting an interaction with NIS-LARG (leukemia-associated RhoA guanine exchange factor). Increased protein levels of cytoplasmic NIS enhanced RhoA activation and resulted in a promigration tumorigenic phenotype. Inhibition of NIS glycosylation through activation of the PI3K/AKT/mTOR signaling pathway contributed to mislocalization of NIS in the cytoplasm, facilitating its nonpump tumorigenic function through an interaction with LARG, which predominantly localized in the cytoplasm. Moreover, PTEN or PI3K/AKT/mTOR signaling could affect DPAGT1, a glycosylating enzyme involved in the initial step of N-linked glycosylation, to inhibit glycosylation of NIS. In summary, our results elucidate a pump-independent, protumorigenic role for NIS in thyroid cancer via its cross-talk with PTEN signaling.Significance: A novel pump-independent protumorigenic role of nonmembranous NIS challenges the presumption that radioiodine treatment of thyroid cancer is ineffective when transmembrane NIS is not expressed. Cancer Res; 78(21); 6121-33. ©2018 AACR.


Asunto(s)
Fosfohidrolasa PTEN/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Transducción de Señal , Simportadores/metabolismo , Neoplasias de la Tiroides/metabolismo , Carcinogénesis , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular , Cicloheximida/química , Citoplasma/metabolismo , Perfilación de la Expresión Génica , Mutación de Línea Germinal , Glicosilación , Humanos , Radioisótopos de Yodo , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Interferente Pequeño/metabolismo , Neoplasias de la Tiroides/genética , Proteína de Unión al GTP rhoA/metabolismo
6.
Nat Commun ; 9(1): 1270, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29593334

RESUMEN

Gain-of-function p53 mutants such as p53-R175H form stable aggregates that accumulate in cells and play important roles in cancer progression. Selective degradation of gain-of-function p53 mutants has emerged as a highly attractive therapeutic strategy to target cancer cells harboring specific p53 mutations. We identified a small molecule called MCB-613 to cause rapid ubiquitination, nuclear export, and degradation of p53-R175H through a lysosome-mediated pathway, leading to catastrophic cancer cell death. In contrast to its effect on the p53-R175H mutant, MCB-613 causes slight stabilization of p53-WT and has weaker effects on other p53 gain-of-function mutants. Using state-of-the-art genetic and chemical approaches, we identified the deubiquitinase USP15 as the mediator of MCB-613's effect on p53-R175H, and established USP15 as a selective upstream regulator of p53-R175H in ovarian cancer cells. These results confirm that distinct pathways regulate the turnover of p53-WT and the different p53 mutants and open new opportunities to selectively target them.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Lisosomas/metabolismo , Neoplasias Ováricas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Línea Celular Tumoral , Cicloheximida/química , Femenino , Células HEK293 , Humanos , Células MCF-7 , Mutación , Plásmidos/metabolismo , Receptores de Esteroides/metabolismo
7.
J Biol Chem ; 292(19): 8059-8072, 2017 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-28325838

RESUMEN

The phagocytic elimination of cells undergoing apoptosis is an evolutionarily conserved innate immune mechanism for eliminating unnecessary cells. Previous studies showed an increase in the level of engulfment receptors in phagocytes after the phagocytosis of apoptotic cells, which leads to the enhancement of their phagocytic activity. However, precise mechanisms underlying this phenomenon require further clarification. We found that the pre-incubation of a Drosophila phagocyte cell line with the fragments of apoptotic cells enhanced the subsequent phagocytosis of apoptotic cells, accompanied by an augmented expression of the engulfment receptors Draper and integrin αPS3. The DNA-binding activity of the transcription repressor Tailless was transiently raised in those phagocytes, depending on two partially overlapping signal-transduction pathways for the induction of phagocytosis as well as the occurrence of engulfment. The RNAi knockdown of tailless in phagocytes abrogated the enhancement of both phagocytosis and engulfment receptor expression. Furthermore, the hemocyte-specific RNAi of tailless reduced apoptotic cell clearance in Drosophila embryos. Taken together, we propose the following mechanism for the activation of Drosophila phagocytes after an encounter with apoptotic cells: two partially overlapping signal-transduction pathways for phagocytosis are initiated; transcription repressor Tailless is activated; expression of engulfment receptors is stimulated; and phagocytic activity is enhanced. This phenomenon most likely ensures the phagocytic elimination of apoptotic cells by stimulated phagocytes and is thus considered as a mechanism to prime phagocytes in innate immunity.


Asunto(s)
Apoptosis , Fagocitos/citología , Transducción de Señal , Animales , Línea Celular , Núcleo Celular/metabolismo , Cicloheximida/química , Proteínas del Citoesqueleto/metabolismo , ADN/análisis , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Hemocitos/citología , Inmunidad Innata , Cadenas alfa de Integrinas/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Oncogénica v-crk/metabolismo , Fagocitosis , Interferencia de ARN , Proteínas Represoras/metabolismo
8.
Cell Chem Biol ; 24(4): 458-470.e18, 2017 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-28343940

RESUMEN

USP2a is a deubiquitinase responsible for stabilization of cyclin D1, a crucial regulator of cell-cycle progression and a proto-oncoprotein overexpressed in numerous cancer types. Here we report that lithocholic acid (LCA) derivatives are inhibitors of USP proteins, including USP2a. The most potent LCA derivative, LCA hydroxyamide (LCAHA), inhibits USP2a, leading to a significant Akt/GSK3ß-independent destabilization of cyclin D1, but does not change the expression of p27. This leads to the defects in cell-cycle progression. As a result, LCAHA inhibits the growth of cyclin D1-expressing, but not cyclin D1-negative cells, independently of the p53 status. We show that LCA derivatives may be considered as future therapeutics for the treatment of cyclin D1-addicted p53-expressing and p53-defective cancer types.


Asunto(s)
Ciclina D1/metabolismo , Endopeptidasas/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Ácido Litocólico/análogos & derivados , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina D1/antagonistas & inhibidores , Ciclina D1/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Cicloheximida/química , Cicloheximida/farmacología , Regulación hacia Abajo/efectos de los fármacos , Endopeptidasas/química , Endopeptidasas/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HCT116 , Humanos , Ácido Litocólico/farmacología , Células MCF-7 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina Tiolesterasa
9.
J Steroid Biochem Mol Biol ; 165(Pt B): 430-437, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27601169

RESUMEN

BACKGROUND: Axin1 is a scaffold protein in the ß-catenin destruction complex, which, if disrupted, contributes to pathogenesis of various human diseases, including colorectal carcinogenesis and inflammatory bowel diseases (IBD). We have previously demonstrated that Salmonella infection promotes the degradation and plasma sequestration of Axin1, leading to bacterial invasiveness and inflammatory responses. Vitamin D and the vitamin D receptor (VDR) appear to be important regulators of IBD and colon cancer. Although VDR and Axin1 are all involved in intestinal inflammation, it remains unclear whether these processes are related or function independently. In the current study, we hypothesize that VDR is an important regulator for the maintenance of physiological level of Axin1. METHODS: Using the intestinal epithelial conditional VDR knockout mouse model (VDRΔIEC) and cultured cell lines, influences of VDR status on the expression of Axin1 was evaluated by Western blots and real-time PCR. Loss- and gain-of-function assays were used to investigate the regulation of VDR on Axin1 at the transcriptional and translational levels. Cells were treated with cycloheximide or actinomycin for molecular mechanistic studies. Candidate genomic VDR binding sites for Axin1 were tested by chromatin immunoprecipitation (ChIP) assay. Physical interactions among VDR, Axin1, and ß-catenin were tested by immunoprecipitation. Cellular localization of Axin1 with different VDR status was determined by fractionation and immunohistochemistry. RESULTS: We found that VDR deletion led to lower protein and mRNA levels of Axin1, whereas knockdown of Axin1 did not change the expression level of VDR protein. Immunoprecipitation data did not support physical interaction between VDR and Axin1. The VDR regulation of Axin1 was through a VDR genomic binding site for Axin1 gene on the regulatory region. Fractionation data showed that cytosolic Axin1 was significantly reduced due to VDR deletion, leaving the nuclear fraction unchanged. In ileum, Axin1 was distributed in the cytosol of apical epithelium and crypts. CONCLUSION: VDR is important for the maintenance of physiological level of Axin1. The discovery of Axin1 as a VDR target gene provides novel and fundamental insights into the interactions between the VDR and ß-catenin signaling pathways.


Asunto(s)
Proteína Axina/metabolismo , Receptores de Calcitriol/metabolismo , Animales , Sitios de Unión , Núcleo Celular/metabolismo , Neoplasias del Colon/metabolismo , Cicloheximida/química , Citosol/metabolismo , Dactinomicina/química , Epitelio/metabolismo , Fibroblastos/metabolismo , Células HCT116 , Células HT29 , Humanos , Inflamación , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Transducción de Señal , beta Catenina/metabolismo
10.
Autophagy ; 12(9): 1575-92, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27310465

RESUMEN

Spermiogenesis is a complex and highly ordered spermatid differentiation process that requires reorganization of cellular structures. We have previously found that Atg7 is required for acrosome biogenesis. Here, we show that autophagy regulates the round and elongating spermatids. Specifically, we found that Atg7 is required for spermatozoa flagella biogenesis and cytoplasm removal during spermiogenesis. Spermatozoa motility of atg7-null mice dropped significantly with some extra-cytoplasm retained on the mature sperm head. These defects are associated with an impairment of the cytoskeleton organization. Functional screening revealed that the negative cytoskeleton organization regulator, PDLIM1 (PDZ and LIM domain 1 [elfin]), needs to be degraded by the autophagy-lysosome-dependent pathway to facilitate the proper organization of the cytoskeleton. Our results thus provide a novel mechanism showing that autophagy regulates cytoskeleton organization mainly via degradation of PDLIM1 to facilitate the differentiation of spermatids.


Asunto(s)
Autofagia , Regulación de la Expresión Génica , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Espermátides/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Proteína 7 Relacionada con la Autofagia/genética , Diferenciación Celular , Biología Computacional , Cicloheximida/química , Citoplasma/metabolismo , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Flagelos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microtúbulos/metabolismo , Espermatogénesis , Espermatozoides/citología
11.
Arch Biochem Biophys ; 603: 102-9, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27246478

RESUMEN

Glucocorticoids (GCs) are a class of steroid hormones that regulate multiple aspects of glucose homeostasis. In skeletal muscle, it is well established that prolonged GC excess inhibits glucose uptake and utilization through glucocorticoid receptor (GR)-mediated transcriptional changes. However, it remains obscure that whether the rapid non-genomic effects of GC on glucose uptake are involved in acute exercise stress. Therefore, we used electric pulse stimulation (EPS)-evoked contracting myotubes to determine whether the non-genomic actions of GC were involved and its underlying mechanism(s). Pretreatment with dexamethasone (Dex, 10 µM) significantly prevented contraction-stimulated glucose uptake and glucose transporter 4 (Glut4) translocation within 20 min in C2C12 myotubes. Neither GC nuclear receptor antagonist (RU486) nor protein synthesis inhibitor (cycloheximide, Chx) affected the rapid inhibition effects of Dex. AMPK and CaMKII-dependent signaling pathways were associated with the non-genomic effects of Dex. These results provide evidence that GC rapidly suppresses glucose uptake in contracting myotubes via GR-independent non-genomic mechanisms. AMPK and CaMKII-mediated Glut4 translocation may play a critical role in GC-induced rapid inhibition of glucose uptake.


Asunto(s)
Dexametasona/química , Glucosa/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antiinflamatorios/química , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Diferenciación Celular , Línea Celular , Membrana Celular/metabolismo , Cicloheximida/química , Genómica , Transportador de Glucosa de Tipo 4/metabolismo , Ratones , Mifepristona/química , Músculo Esquelético/metabolismo , Fosforilación , Condicionamiento Físico Animal , Transporte de Proteínas , Receptores de Glucocorticoides/metabolismo , Transducción de Señal , Esteroides/química , Transcripción Genética
12.
J Biol Chem ; 291(5): 2196-222, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26670607

RESUMEN

Rev-erbα and Rev-erbß are heme-binding nuclear receptors (NR) that repress the transcription of genes involved in regulating metabolism, inflammation, and the circadian clock. Previous gene expression and co-immunoprecipitation studies led to a model in which heme binding to Rev-erbα recruits nuclear receptor corepressor 1 (NCoR1) into an active repressor complex. However, in contradiction, biochemical and crystallographic studies have shown that heme decreases the affinity of the ligand-binding domain of Rev-erb NRs for NCoR1 peptides. One explanation for this discrepancy is that the ligand-binding domain and NCoR1 peptides used for in vitro studies cannot replicate the key features of the full-length proteins used in cellular studies. However, the combined in vitro and cellular results described here demonstrate that heme does not directly promote interactions between full-length Rev-erbß (FLRev-erbß) and an NCoR1 construct encompassing all three NR interaction domains. NCoR1 tightly binds both apo- and heme-replete FLRev-erbß·DNA complexes; furthermore, heme, at high concentrations, destabilizes the FLRev-erbß·NCoR1 complex. The interaction between FLRev-erbß and NCoR1 as well as Rev-erbß repression at the Bmal1 promoter appear to be modulated by another cellular factor(s), at least one of which is related to the ubiquitin-proteasome pathway. Our studies suggest that heme is involved in regulating the degradation of Rev-erbß in a manner consistent with its role in circadian rhythm maintenance. Finally, the very slow rate constant (10(-6) s(-1)) of heme dissociation from Rev-erbß rules out a prior proposal that Rev-erbß acts as an intracellular heme sensor.


Asunto(s)
Regulación de la Expresión Génica , Hemo/química , Co-Represor 1 de Receptor Nuclear/química , Receptores Citoplasmáticos y Nucleares/química , Proteínas Represoras/química , Secuencias de Aminoácidos , Apoproteínas/química , Ritmo Circadiano , Cicloheximida/química , Células HEK293 , Humanos , Inflamación , Iones , Ligandos , Espectrometría de Masas , Metales/química , Mioglobina/química , Regiones Promotoras Genéticas , Unión Proteica , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Transfección , Ubiquitina/química
13.
J Biol Chem ; 291(5): 2246-59, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26644469

RESUMEN

Cell signaling pathways are noisy communication channels, and statistical measures derived from information theory can be used to quantify the information they transfer. Here we use single cell signaling measures to calculate mutual information as a measure of information transfer via gonadotropin-releasing hormone (GnRH) receptors (GnRHR) to extracellular signal-regulated kinase (ERK) or nuclear factor of activated T-cells (NFAT). This revealed mutual information values <1 bit, implying that individual GnRH-responsive cells cannot unambiguously differentiate even two equally probable input concentrations. Addressing possible mechanisms for mitigation of information loss, we focused on the ERK pathway and developed a stochastic activation model incorporating negative feedback and constitutive activity. Model simulations revealed interplay between fast (min) and slow (min-h) negative feedback loops with maximal information transfer at intermediate feedback levels. Consistent with this, experiments revealed that reducing negative feedback (by expressing catalytically inactive ERK2) and increasing negative feedback (by Egr1-driven expression of dual-specificity phosphatase 5 (DUSP5)) both reduced information transfer from GnRHR to ERK. It was also reduced by blocking protein synthesis (to prevent GnRH from increasing DUSP expression) but did not differ for different GnRHRs that do or do not undergo rapid homologous desensitization. Thus, the first statistical measures of information transfer via these receptors reveals that individual cells are unreliable sensors of GnRH concentration and that this reliability is maximal at intermediate levels of ERK-mediated negative feedback but is not influenced by receptor desensitization.


Asunto(s)
Retroalimentación Fisiológica , Regulación Enzimológica de la Expresión Génica , Hormona Liberadora de Gonadotropina/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factores de Transcripción NFATC/metabolismo , Receptores LHRH/metabolismo , Catálisis , Simulación por Computador , Cicloheximida/química , Fosfatasas de Especificidad Dual/metabolismo , Células HeLa , Humanos , Modelos Teóricos , Inhibidores de la Síntesis de la Proteína/química , Transducción de Señal , Procesos Estocásticos
14.
Oncotarget ; 6(25): 20933-45, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26369285

RESUMEN

The TRAF-interacting protein (TRAIP) is an E3 ubiquitin ligase required for cell proliferation. TRAIP mRNA is downregulated in human keratinocytes after inhibition of the PI3K/AKT/mTOR signaling. Since E2F transcription factors are downstream of PI3K/AKT/mTOR we investigated whether they regulate TRAIP expression. E2F1 expression significantly increased the TRAIP mRNA level in HeLa cells. Reporter assays with the 1400 bp 5'-upstream promoter in HeLa cells and human keratinocytes showed that E2F1-, E2F2- and E2F4-induced upregulation of TRAIP expression is mediated by 168 bp upstream of the translation start site. Mutating the E2F binding site within this fragment reduced the E2F1- and E2F2-dependent promoter activities and protein-DNA complex formation in gel shift assays. Abundance of TRAIP mRNA and protein was regulated by the cell cycle with a peak in G2/M. Expression of GFP and TRAIP-GFP demonstrated that TRAIP-GFP protein has a lower steady-state concentration than GFP despite similar mRNA levels. Cycloheximide inhibition experiments indicated that the TRAIP protein has a half-life of around four hours. Therefore, the combination of cell cycle-dependent transcription of the TRAIP gene by E2F and rapid protein degradation leads to cell cycle-dependent expression with a maximum in G2/M. These findings suggest that TRAIP has important functions in mitosis and tumorigenesis.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F2/metabolismo , Factor de Transcripción E2F4/metabolismo , Regulación Neoplásica de la Expresión Génica , Mitosis , Ubiquitina-Proteína Ligasas/metabolismo , Células 3T3 , Animales , Ciclo Celular , Núcleo Celular/metabolismo , Proliferación Celular , Cicloheximida/química , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Queratinocitos/citología , Ratones , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , Inhibidores de la Síntesis de la Proteína/química , ARN Mensajero/metabolismo
15.
Oncotarget ; 6(27): 24393-403, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26405158

RESUMEN

The oncogenic RAS-selective lethal small molecule Erastin triggers a unique iron-dependent form of nonapoptotic cell death termed ferroptosis. Ferroptosis is dependent upon the production of intracellular iron-dependent reactive oxygen species (ROS), but not other metals. However, key regulators remain unknown. The heme oxygenase (HO) is a major intracellular source of iron. In this study, the role of heme oxygenase in Erastin-triggered ferroptotic cancer cell death has been investigated. Zinc protoporphyrin IX (ZnPP), a HO-1 inhibitor, prevented Erastin-triggered ferroptotic cancer cell death. Furthermore, Erastin induced the protein and mRNA levels of HO-1 in HT-1080 fibrosarcoma cells. HO-1+/+ and HO-1-/- fibroblast, HO-1 overexpression, and chycloheximide-treated experiments revealed that the expression of HO-1 has a decisive effects in Erastin-triggered cell death. Hemin and CO-releasing molecules (CORM) promote Erastin-induced ferroptotic cell death, not by biliverdin and bilirubin. In addition, hemin and CORM accelerate the HO-1 expression in the presence of Erastin and increase membranous lipid peroxidation. Thus, HO-1 is an essential enzyme for iron-dependent lipid peroxidation during ferroptotic cell death.


Asunto(s)
Muerte Celular , Regulación Neoplásica de la Expresión Génica , Hemo-Oxigenasa 1/metabolismo , Hierro/química , Proteínas de la Membrana/metabolismo , Piperazinas/química , Animales , Bilirrubina/química , Biliverdina/química , Línea Celular Tumoral , Núcleo Celular/metabolismo , Cicloheximida/química , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Hemina/química , Humanos , Peroxidación de Lípido , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Protoporfirinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
16.
Proc Natl Acad Sci U S A ; 112(12): 3728-33, 2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25775599

RESUMEN

Eukaryotic cells have evolved robust mechanisms to counter excess cholesterol including redistribution of lipids into different compartments and compensatory up-regulation of phospholipid biosynthesis. We demonstrate here that excess cellular cholesterol increased the activity of the endoplasmic reticulum (ER) enzyme serine palmitoyl-CoA transferase (SPT), the rate-limiting enzyme in sphingomyelin synthesis. This increased SPT activity was not due to altered levels of SPTLC1 or SPTLC2, the major subunits of SPT. Instead, cholesterol loading decreased the levels of ORMDL1, a negative regulator of SPT activity, due to its increased turnover. Several lines of evidence demonstrated that free-cholesterol-induced autophagy, which led to increased turnover of ORMDL1. Cholesterol loading induced ORMDL1 redistribution from the ER to cytoplasmic p62 positive autophagosomes. Coimmunoprecipitation analysis of cholesterol-loaded cells showed increased association between ORMDL1 and p62. The lysosomal inhibitor chloroquine or siRNA knockdown of Atg7 inhibited ORMDL1 degradation by cholesterol, whereas proteasome inhibitors showed no effect. ORMDL1 degradation was specific to free-cholesterol loading as autophagy induced by serum starvation or general ER stress did not lead to ORMDL1 degradation. ORMDL proteins are thus previously unidentified responders to excess cholesterol, exiting the ER to activate SPT and increase sphingomyelin biosynthesis, which may buffer excess cellular cholesterol.


Asunto(s)
Autofagia , Orosomucoide/metabolismo , Animales , Asma/metabolismo , Aterosclerosis/metabolismo , Transporte Biológico , Línea Celular , Colesterol/metabolismo , Cicloheximida/química , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Eosinófilos/metabolismo , Homeostasis , Lípidos/química , Macrófagos/metabolismo , Microdominios de Membrana/química , Proteínas de la Membrana , Ratones , Transporte de Proteínas , Serina C-Palmitoiltransferasa/química , Esfingolípidos/química , Esfingomielinas/química
17.
J Biol Chem ; 290(4): 2489-95, 2015 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-25492870

RESUMEN

Proteins are typically targeted for proteasomal degradation by the attachment of a polyubiquitin chain to ϵ-amino groups of lysine residues. Non-lysine ubiquitylation of proteasomal substrates has been considered an atypical and rare event limited to complex eukaryotes. Here we report that a fully functional lysine-less mutant of an inner nuclear membrane protein in yeast, Asi2, is polyubiquitylated and targeted for proteasomal degradation. Efficient degradation of lysine-free Asi2 requires E3-ligase Doa10 and E2 enzymes Ubc6 and Ubc7, components of the endoplasmic reticulum-associated degradation pathway. Together, our data suggest that non-lysine ubiquitylation may be more prevalent than currently considered.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico , Regulación Fúngica de la Expresión Génica , Lisina/genética , Proteínas de la Membrana/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Núcleo Celular/metabolismo , Cicloheximida/química , Retículo Endoplásmico/metabolismo , Epítopos/química , Lisina/química , Mutación , Plásmidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
18.
J Zhejiang Univ Sci B ; 15(12): 1032-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25471832

RESUMEN

The p53 tumor suppressor protein coordinates the cellular responses to a broad range of cellular stresses, leading to DNA repair, cell cycle arrest or apoptosis. The stability of p53 is essential for its tumor suppressor function, which is tightly controlled by ubiquitin-dependent degradation primarily through its negative regulator murine double minute 2 (Mdm2). To better understand the regulation of p53, we tested the interaction between p53 and USP11 using co-immunoprecipitation. The results show that USP11, an ubiquitin-specific protease, forms specific complexes with p53 and stabilizes p53 by deubiquitinating it. Moreover, down-regulation of USP11 dramatically attenuated p53 induction in response to DNA damage stress. These findings reveal that USP11 is a novel regulator of p53, which is required for p53 activation in response to DNA damage.


Asunto(s)
Daño del ADN , Tioléster Hidrolasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Cicloheximida/química , Reparación del ADN , Células HEK293 , Humanos , Plásmidos/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Interferencia de ARN , Ubiquitinación
19.
Biochem Biophys Res Commun ; 455(3-4): 241-5, 2014 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-25449278

RESUMEN

The chaperone αB-crystallin (αBC) is a member of the small heat shock protein family and its point or truncated mutants cause the muscular disorder α-crystallinopathy. The illness is histologically characterized by accumulation of protein aggregates in muscle cells. Expression of the myopathy-causing R120G mutant of αBC, harboring an arginine-to-glycine mutation at position 120, results in aggregate formation. We demonstrated that tethering αBC to the endoplasmic reticulum (ER) membrane represses the protein aggregation mediated by the R120G mutant. ER-anchored αBC decreased the amount of the R120G mutant through autophagic proteolysis. In contrast, knockdown of ATG5, an E3 ligase essential for autophagy, in ER-anchored αBC-transfected cells restored the quantity of the R120G mutant. In this context, aggregate formation was still suppressed, indicating that ER-anchored αBC profoundly constrains aggregation competency of the R120G mutant separately from downregulating the abundance of the mutant. We have proposed that protein aggregation is prevented by manipulation of the ER microenvironment with αBC, and have shed light on a novel aspect of the ER as a therapeutic target.


Asunto(s)
Retículo Endoplásmico/metabolismo , Agregación Patológica de Proteínas/prevención & control , Cadena B de alfa-Cristalina/metabolismo , Autofagia , Cicloheximida/química , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Inmunoprecipitación , Microscopía Fluorescente , Enfermedades Musculares/patología , Mutación , Unión Proteica , ARN Interferente Pequeño/metabolismo , Transfección , Cadena B de alfa-Cristalina/genética
20.
J Food Prot ; 77(10): 1682-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25285484

RESUMEN

Campylobacter is the most frequent cause of bacterial gastroenteritis in Canada, and the illness is commonly associated with poultry consumption. Whereas Canadian retail poultry is often contaminated with campylobacters, studies on the prevalence of this organism are inconsistent due to variability in sampling and microbiological methodology. To determine the current microbiological status of Canadian poultry, and to evaluate two commonly used microbiological methods, 348 raw poultry samples were collected at retail across Canada over a period of 3 years (2007 to 2010) and were analyzed for the presence of thermophilic Campylobacter species. The overall prevalence of Campylobacter spp. was found to be 42.8% by a combination of the two testing methods, with 33.9% of the samples positive for C. jejuni, 3.7% of the samples positive for C. coli, and 5.2% of the samples positive for both. Variability in Campylobacter spp. prevalence was observed in samples obtained from different regions across Canada and from poultry with or without skin, but this was not statistically significant. In co-contaminated samples, C. jejuni was preferentially recovered from Preston agar compared with mCCDA and Campy-Cefex agar, with an increase in recovery of C. coli on all selective media after 48 h of enrichment. A subset of 214 of the poultry rinses were analyzed by both Health Canada's standard method, MFLP-46 (enrichment in Park and Sanders broth), and a second method requiring enrichment in Bolton broth. Significantly more positive samples were obtained with the MFLP-46 method (40.6%) than with the alternate method (35.0%). This improved recovery with MFLP-46 may be due to the omission of cycloheximide from this method. These results demonstrate that determination of prevalence of Campylobacter spp. on poultry products may be significantly impacted by the choice of microbiological methods used. Canadian poultry continues to be a source of exposure to Campylobacter spp.


Asunto(s)
Campylobacter coli/aislamiento & purificación , Campylobacter jejuni/aislamiento & purificación , Microbiología de Alimentos/métodos , Carne/microbiología , Aves de Corral/microbiología , Agar , Animales , Canadá , Cicloheximida/química , Contaminación de Alimentos/análisis , Productos Avícolas/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...