Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 14(5): 470-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23542741

RESUMEN

Uncontrolled activation of tumor necrosis factor receptor-associated factor (TRAF) proteins may result in profound tissue injury by linking surface signals to cytokine release. Here we show that a ubiquitin E3 ligase component, Fbxo3, potently stimulates cytokine secretion from human inflammatory cells by destabilizing a sentinel TRAF inhibitor, Fbxl2. Fbxo3 and TRAF protein in circulation positively correlated with cytokine responses in subjects with sepsis, and we identified a polymorphism in human Fbxo3, with one variant being hypofunctional. A small-molecule inhibitor targeting Fbxo3 was sufficient to lessen severity of cytokine-driven inflammation in several mouse disease models. These studies identified a pathway of innate immunity that may be useful to detect subjects with altered immune responses during critical illness or provide a basis for therapeutic intervention targeting TRAF protein abundance.


Asunto(s)
Proteínas F-Box/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Sepsis/inmunología , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Ciego/inmunología , Ciego/cirugía , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Secuencias F-Box/genética , Proteínas F-Box/genética , Humanos , Inmunomodulación , Inflamación/genética , Ratones , Ratones Endogámicos C57BL , Polimorfismo Genético , Estabilidad Proteica , Infecciones por Pseudomonas/genética , Pseudomonas aeruginosa/genética , ARN Interferente Pequeño/genética , Sepsis/genética , Transgenes/genética
2.
Nat Immunol ; 13(7): 651-8, 2012 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-22660580

RESUMEN

The ST2L receptor for interleukin 33 (IL-33) mediates pulmonary inflammation and immune system-related disorders, such as asthma and rheumatoid arthritis. At present, very little is known about the molecular regulation of ST2L expression. Here we found that FBXL19, an 'orphan' member of the Skp1-Cullin-F-box family of E3 ubiquitin ligases, selectively bound to ST2L to mediate its polyubiquitination and elimination in the proteasome. Degradation of ST2L involved phosphorylation of ST2L at Ser442 catalyzed by the kinase GSK3ß. Overexpression of FBXL19 abrogated the proapoptotic and inflammatory effects of IL-33 and lessened the severity of lung injury in mouse models of pneumonia. Our results suggest that modulation of the IL-33-ST2L axis by ubiquitin ligases might serve as a unique strategy for lessening pulmonary inflammation.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Proteínas F-Box/inmunología , Neumonía/inmunología , Receptores de Interleucina/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/inmunología , Animales , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Proteínas F-Box/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteína 1 Similar al Receptor de Interleucina-1 , Ratones , Ratones Endogámicos C57BL , Fosforilación , Neumonía/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Serina/metabolismo , Índice de Severidad de la Enfermedad
3.
COPD ; 21(1): 2342797, 2024 12.
Artículo en Inglés | MEDLINE | ID: mdl-38712759

RESUMEN

Objective: To investigate the effects of cigarette smoke (CS) on Serine/Threonine Kinase 11 (STK11) and to determine STK11's role in CS-induced airway epithelial cell cytotoxicity.Methods: STK11 expression levels in the lung tissues of smokers with or without COPD and mice exposed to CS or room air (RA) were determined by immunoblotting and RT-PCR. BEAS-2Bs-human bronchial airway epithelial cells were exposed to CS extract (CSE), and the changes in STK11 expression levels were determined by immunoblotting and RT-PCR. BEAS-2B cells were transfected with STK11-specific siRNA or STK11 expression plasmid, and the effects of CSE on airway epithelial cell cytotoxicity were measured. To determine the specific STK11 degradation-proteolytic pathway, BEAS-2Bs were treated with cycloheximide alone or combined with MG132 or leupeptin. Finally, to identify the F-box protein mediating the STK11 degradation, a screening assay was performed using transfection with a panel of FBXL E3 ligase subunits.Results: STK11 protein levels were significantly decreased in the lung tissues of smokers with COPD relative to smokers without COPD. STK11 protein levels were also significantly decreased in mouse lung tissues exposed to CS compared to RA. Exposure to CSE shortened the STK11 mRNA and protein half-life to 4 h in BEAS-2B cells. STK11 protein overexpression attenuated the CSE-induced cytotoxicity; in contrast, its knockdown augmented CSE-induced cytotoxicity. FBXL19 mediates CSE-induced STK11 protein degradation via the ubiquitin-proteasome pathway in cultured BEAS-2B cells. FBXL19 overexpression led to accelerated STK11 ubiquitination and degradation in a dose-dependent manner.Conclusions: Our results suggest that CSE enhances the degradation of STK11 protein in airway epithelial cells via the FBXL19-mediated ubiquitin-proteasomal pathway, leading to augmented cell death.HIGHLIGHTSLung tissues of COPD-smokers exhibited a decreased STK11 RNA and protein expression.STK11 overexpression attenuates CS-induced airway epithelial cell cytotoxicity.STK11 depletion augments CS-induced airway epithelial cell cytotoxicity.CS diminishes STK11 via FBXL19-mediated ubiquitin-proteasome degradation.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Células Epiteliales , Proteínas F-Box , Proteínas Serina-Treonina Quinasas , Humo , Animales , Humanos , Masculino , Ratones , Quinasas de la Proteína-Quinasa Activada por el AMP , Línea Celular , Fumar Cigarrillos/efectos adversos , Cicloheximida/farmacología , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Proteínas F-Box/metabolismo , Proteínas F-Box/genética , Leupeptinas/farmacología , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteolisis/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/genética , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/efectos de los fármacos , ARN Interferente Pequeño , Humo/efectos adversos
4.
Int J Mol Sci ; 25(2)2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38256231

RESUMEN

The pandemic of coronavirus disease 2019 (COVID-19) has been the foremost modern global public health challenge. The airway is the primary target in severe acute respiratory distress syndrome coronavirus 2 (SARS-CoV-2) infection, with substantial cell death and lung injury being signature hallmarks of exposure. The viral factors that contribute to cell death and lung injury remain incompletely understood. Thus, this study investigated the role of open reading frame 7b (Orf7b), an accessory protein of the virus, in causing lung injury. In screening viral proteins, we identified Orf7b as one of the major viral factors that mediates lung epithelial cell death. Overexpression of Orf7b leads to apoptosis and ferroptosis in lung epithelial cells, and inhibitors of apoptosis and ferroptosis ablate Orf7b-induced cell death. Orf7b upregulates the transcription regulator, c-Myc, which is integral in the activation of lung cell death pathways. Depletion of c-Myc alleviates both apoptotic and ferroptotic cell deaths and lung injury in mouse models. Our study suggests a major role of Orf7b in the cell death and lung injury attributable to COVID-19 exposure, supporting it as a potential therapeutic target.


Asunto(s)
COVID-19 , Ferroptosis , Lesión Pulmonar , Proteínas Virales , Animales , Ratones , Apoptosis , Lesión Pulmonar/virología , Sistemas de Lectura Abierta , SARS-CoV-2 , Proteínas Virales/genética
5.
Thorax ; 78(4): 402-408, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35301243

RESUMEN

BACKGROUND: Birt-Hogg-Dubé syndrome (BHD) is a clinical syndrome manifesting with cystic lung disease and pneumothorax. Features of BHD result from the loss-of-function mutations of the folliculin (FLCN) gene. Chronic obstructive pulmonary disease (COPD), characterised by an irreversible airflow limitation, is primarily caused by cigarette smoking. OBJECTIVE: Given that COPD often shares structural features with BHD, we investigated the link between COPD, cigarette smoke (CS) exposure and FLCN expression. METHODS: We measured the expression of FLCN in human COPD lungs and CS-exposed mouse lungs, as well as in CS extract (CSE)-exposed immortalised human airway epithelial cells by immunoblotting. RESULTS: We found that the lung FLCN protein levels in smokers with COPD and CS exposure mice exhibit a marked decrease compared with smokers without COPD and room air exposure mice, respectively. We confirmed CS induced degradation of FLCN in immortalised human bronchial epithelial Beas-2B cells via ubiquitin proteasome system. Further, siRNA targeting FLCN enhanced CSE-induced cytotoxicity. By contrast, FLCN overexpression protected cells from CSE-induced cytotoxicity. We found that FBXO23, the ubiquitin E3 ligase subunit, specifically binds to and targets FLCN for degradation. Inhibition of ATM (ataxia-telangiectasia mutated) attenuated CSE induced FLCN degradation, suggesting a role of ATM in FLCN proteolysis. We further confirmed that the mutant of major FLCN phosphorylation site serine 62A is resistant to CSE-induced degradation and cytotoxicity. CONCLUSIONS: Our study demonstrates that CS exposure is a secondary cause of FLCN deficiency due to the enhanced proteolysis, which promoted airway epithelial cell death.


Asunto(s)
Fumar Cigarrillos , Enfermedad Pulmonar Obstructiva Crónica , Animales , Humanos , Ratones , Fumar Cigarrillos/efectos adversos , Pulmón/química , Pulmón/metabolismo , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Proteínas Supresoras de Tumor/análisis , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinas/metabolismo
6.
Thorax ; 78(4): 383-393, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35354645

RESUMEN

BACKGROUND: One hallmark of sepsis is the reduced number of lymphocytes, termed lymphopenia, that occurs from decreased lymphocyte proliferation or increased cell death contributing to immune suppression. Histone modification enzymes regulate immunity by their epigenetic and non-epigenetic functions; however, the role of these enzymes in lymphopenia remains elusive. METHODS: We used molecular biological approaches to investigate the high expression and function of a chromatin modulator protein arginine N-methyltransferase 4 (PRMT4)/coactivator-associated arginine methyltransferase 1 in human samples from septic patients and cellular and animal septic models. RESULTS: We identified that PRMT4 is elevated systemically in septic patients and experimental sepsis. Gram-negative bacteria and their derived endotoxin lipopolysaccharide (LPS) increased PRMT4 in B and T lymphocytes and THP-1 monocytes. Single-cell RNA sequencing results indicate an increase of PRMT4 gene expression in activated T lymphocytes. Augmented PRMT4 is crucial for inducing lymphocyte apoptosis but not monocyte THP-1 cells. Ectopic expression of PRMT4 protein caused substantial lymphocyte death via caspase 3-mediated cell death signalling, and knockout of PRMT4 abolished LPS-mediated lymphocyte death. PRMT4 inhibition with a small molecule compound attenuated lymphocyte death in complementary models of sepsis. CONCLUSIONS: These findings demonstrate a previously uncharacterised role of a key chromatin modulator in lymphocyte survival that may shed light on devising therapeutic modalities to lessen the severity of septic immunosuppression.


Asunto(s)
Linfopenia , Proteína-Arginina N-Metiltransferasas , Sepsis , Animales , Humanos , Arginina/genética , Caspasa 3/genética , Caspasa 3/inmunología , Cromatina , Lipopolisacáridos/farmacología , Linfopenia/etiología , Linfopenia/genética , Linfopenia/inmunología , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Sepsis/complicaciones , Sepsis/genética , Sepsis/inmunología
7.
Respir Res ; 22(1): 100, 2021 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-33823868

RESUMEN

BACKGROUND: Whole lung tissue transcriptomic profiling studies in chronic obstructive pulmonary disease (COPD) have led to the identification of several genes associated with the severity of airflow limitation and/or the presence of emphysema, however, the cell types driving these gene expression signatures remain unidentified. METHODS: To determine cell specific transcriptomic changes in severe COPD, we conducted single-cell RNA sequencing (scRNA seq) on n = 29,961 cells from the peripheral lung parenchymal tissue of nonsmoking subjects without underlying lung disease (n = 3) and patients with severe COPD (n = 3). The cell type composition and cell specific gene expression signature was assessed. Gene set enrichment analysis (GSEA) was used to identify the specific cell types contributing to the previously reported transcriptomic signatures. RESULTS: T-distributed stochastic neighbor embedding and clustering of scRNA seq data revealed a total of 17 distinct populations. Among them, the populations with more differentially expressed genes in cases vs. controls (log fold change >|0.4| and FDR = 0.05) were: monocytes (n = 1499); macrophages (n = 868) and ciliated epithelial cells (n = 590), respectively. Using GSEA, we found that only ciliated and cytotoxic T cells manifested a trend towards enrichment of the previously reported 127 regional emphysema gene signatures (normalized enrichment score [NES] = 1.28 and = 1.33, FDR = 0.085 and = 0.092 respectively). Among the significantly altered genes present in ciliated epithelial cells of the COPD lungs, QKI and IGFBP5 protein levels were also found to be altered in the COPD lungs. CONCLUSIONS: scRNA seq is useful for identifying transcriptional changes and possibly individual protein levels that may contribute to the development of emphysema in a cell-type specific manner.


Asunto(s)
Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Pulmón/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/genética , Proteínas de Unión al ARN/genética , ARN/genética , Análisis de Secuencia de ARN/métodos , Transcriptoma/genética , Adulto , Anciano , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Pulmón/patología , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , ARN/metabolismo , Proteínas de Unión al ARN/biosíntesis , Índice de Severidad de la Enfermedad , Adulto Joven
8.
Am J Physiol Lung Cell Mol Physiol ; 318(2): L252-L263, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31746627

RESUMEN

Cigarette smoking increases susceptibility for microbial infection in respiratory system. However, the underlying molecular mechanism(s) is not fully elucidated. Here we report that cigarette smoking extract (CSE) increases bacterial load in lung epithelial cells via downregulation of the ubiquitin-specific protease 25 (USP25)/histone deacetylase 11 (HDAC11) axis. CSE treatment decreases HDAC11 at protein level in lung epithelial cells without significant changes of its transcription. Concomitantly, CSE treatment accelerates a ubiquitin-specific protease USP25 ubiquitination and degradation. Coimmunoprecipitation studies showed that USP25 associated with HDAC11. USP25 catalyzes deubiquitination of HDAC11, which regulates HDAC11 protein stability. CSE-mediated degradation of USP25 thereafter reduces HDAC11 at the protein level. Interestingly, CSE-downregulated USP25/HDAC11 axis increases the bacterial load of Pseudomonas aeruginosa in lung epithelial cells. These findings suggest that CSE-downregulated USP25 and HDAC11 may contribute to high susceptibility of bacterial infection in the cigarette smoking population.


Asunto(s)
Carga Bacteriana/fisiología , Fumar Cigarrillos/efectos adversos , Histona Desacetilasas/metabolismo , Pulmón/metabolismo , Pulmón/microbiología , Pseudomonas aeruginosa/fisiología , Transducción de Señal , Ubiquitina Tiolesterasa/metabolismo , Animales , Línea Celular , Estabilidad de Enzimas , Femenino , Humanos , Lisina/metabolismo , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Ubiquitinación
9.
Int J Mol Sci ; 21(15)2020 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-32731491

RESUMEN

Pseudomonas aeruginosa is an important opportunistic pathogen responsible for the cause of acute lung injury and acute respiratory distress syndrome. P. aeruginosa isthe leading species isolated from patients with nosocomial infection and is detected in almost all the patients with long term ventilation in critical care units. P. aeruginosa infection is also the leading cause of deleterious chronic lung infections in patients suffering from cystic fibrosis as well as the major reason for morbidity in people with chronic obstructive pulmonary disease. P. aeruginosa infections are linked to diseases with high mortality rates and are challenging for treatment, for which no effective remedies have been developed. Massive lung epithelial cell death is a hallmark of severe acute lung injury and acute respiratory distress syndrome caused by P. aeruginosa infection. Lung epithelial cell death poses serious challenges to air barrier and structural integrity that may lead to edema, cytokine secretion, inflammatory infiltration, and hypoxia. Here we review different types of cell death caused by P. aeruginosa serving as a starting point for the diseases it is responsible for causing. We also review the different mechanisms of cell death and potential therapeutics in countering the serious challenges presented by this deadly bacterium.


Asunto(s)
Lesión Pulmonar Aguda , Células Epiteliales , Pulmón , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Síndrome de Dificultad Respiratoria , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Muerte Celular/inmunología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/patología , Humanos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/metabolismo , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/microbiología , Síndrome de Dificultad Respiratoria/patología
10.
Int J Mol Sci ; 21(14)2020 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-32650621

RESUMEN

Acute lung injury and acute respiratory distress syndrome (ALI/ARDS) are characterized by an inflammatory response, alveolar edema, and hypoxemia. ARDS occurs most often in the settings of pneumonia, sepsis, aspiration of gastric contents, or severe trauma. The prevalence of ARDS is approximately 10% in patients of intensive care. There is no effective remedy with mortality high at 30-40%. Most functional proteins are dynamic and stringently governed by ubiquitin proteasomal degradation. Protein ubiquitination is reversible, the covalently attached monoubiquitin or polyubiquitin moieties within the targeted protein can be removed by a group of enzymes called deubiquitinating enzymes (DUBs). Deubiquitination plays an important role in the pathobiology of ALI/ARDS as it regulates proteins critical in engagement of the alveolo-capillary barrier and in the inflammatory response. In this review, we provide an overview of how DUBs emerge in pathogen-induced pulmonary inflammation and related aspects in ALI/ARDS. Better understanding of deubiquitination-relatedsignaling may lead to novel therapeutic approaches by targeting specific elements of the deubiquitination pathways.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Enzimas Desubicuitinizantes/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Animales , Humanos , Neumonía/metabolismo , Transducción de Señal/fisiología , Ubiquitina/metabolismo , Ubiquitinación/fisiología
11.
BMC Genomics ; 20(1): 22, 2019 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-30626320

RESUMEN

BACKGROUND: Aging is affected by genetic and environmental factors, and cigarette smoking is strongly associated with accumulation of senescent cells. In this study, we wanted to identify genes that may potentially be beneficial for cell survival in response to cigarette smoke and thereby may contribute to development of cellular senescence. RESULTS: Primary human bronchial epithelial cells from five healthy donors were cultured, treated with or without 1.5% cigarette smoke extract (CSE) for 24 h or were passaged into replicative senescence. Transcriptome changes were monitored using RNA-seq in CSE and non-CSE exposed cells and those passaged into replicative senescence. We found that, among 1534 genes differentially regulated during senescence and 599 after CSE exposure, 243 were altered in both conditions, representing strong enrichment. Pathways and gene sets overrepresented in both conditions belonged to cellular processes that regulate reactive oxygen species, proteasome degradation, and NF-κB signaling. CONCLUSIONS: Our results offer insights into gene expression responses during cellular aging and cigarette smoke exposure, and identify potential molecular pathways that are altered by cigarette smoke and may also promote airway epithelial cell senescence.


Asunto(s)
Bronquios/metabolismo , Senescencia Celular/genética , Fumar Cigarrillos/genética , Bronquios/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Fumar Cigarrillos/efectos adversos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , FN-kappa B/genética , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos
12.
J Cell Sci ; 130(20): 3578-3587, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28883095

RESUMEN

E3 ubiquitin ligase recognizes its protein substrates via specific molecular signatures for ubiquitin proteasomal degradation. However, the role of acetylation/deacetylation in the process of E3 ubiquitin ligase recognizing its protein substrates is not fully studied. Here, we report that a tandem IK motif in protein arginine methyltransferase 1 (PRMT1) forms an acetyldegron to recruit the F-box/LRR-repeat protein 17 (FBXL17), a component of the SKP1-CUL1-F-box protein (SCF)-type E3 ubiquitin ligase complex. PRMT1 is polyubiquitylated for proteasome degradation with a half-life of approximately 4 h in lung epithelial cells. SCFFbxl17 mediates PRMT1 polyubiquitylation at K117. SCFFbxl17 specifically binds PRMT1 via a unique motif IKxxxIK. Strikingly, the acetylation/deacetylation status of the lysine residues within the motif determines Fbxl17 binding. Deacetylation on both K200 and K205 by Sirtuin 1 (Sirt1) and acetylation of p300 (EP300) on K205 collaboratively prepare the motif for SCFFbxl17 binding thereby triggering PRMT1 protein degradation. Pathogen-derived lipopolysaccharide (LPS) downregulates Sirt1 and p300 to protect PRMT1 from degradation. This study demonstrates that LPS promotes PRMT1 stability by blockade of PRMT1 and SCFFbxl17 binding via an acetylation/deacetylation-modified acetyldegron; and LPS-elevated levels of PRMT1 lead to bronchial epithelial cell overgrowth in pulmonary inflammatory diseases.


Asunto(s)
Proteínas F-Box/metabolismo , Lipopolisacáridos/farmacología , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Sirtuina 1/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Secuencias de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Humanos , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Estabilidad Proteica , Proteolisis , Ubiquitinación/inmunología
13.
J Cell Sci ; 129(1): 51-64, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26604221

RESUMEN

Cardiolipin (also known as PDL6) is an indispensable lipid required for mitochondrial respiration that is generated through de novo synthesis and remodeling. Here, the cardiolipin remodeling enzyme, acyl-CoA:lysocardiolipin-acyltransferase-1 (Alcat1; SwissProt ID, Q6UWP7) is destabilized in epithelia by lipopolysaccharide (LPS) impairing mitochondrial function. Exposure to LPS selectively decreased levels of carbon 20 (C20)-containing cardiolipin molecular species, whereas the content of C18 or C16 species was not significantly altered, consistent with decreased levels of Alcat1. Alcat1 is a labile protein that is lysosomally degraded by the ubiquitin E3 ligase Skp-Cullin-F-box containing the Fbxo28 subunit (SCF-Fbxo28) that targets Alcat1 for monoubiquitylation at residue K183. Interestingly, K183 is also an acetylation-acceptor site, and acetylation conferred stability to the enzyme. Histone deacetylase 2 (HDAC2) interacted with Alcat1, and expression of a plasmid encoding HDAC2 or treatment of cells with LPS deacetylated and destabilized Alcat1, whereas treatment of cells with a pan-HDAC inhibitor increased Alcat1 levels. Alcat1 degradation was partially abrogated in LPS-treated cells that had been silenced for HDAC2 or treated with MLN4924, an inhibitor of Cullin-RING E3 ubiquitin ligases. Thus, LPS increases HDAC2-mediated Alcat1 deacetylation and facilitates SCF-Fbxo28-mediated disposal of Alcat1, thus impairing mitochondrial integrity.


Asunto(s)
Aciltransferasas/metabolismo , Epitelio/metabolismo , Lipopolisacáridos/farmacología , Mitocondrias/enzimología , Oxígeno/metabolismo , Proteolisis/efectos de los fármacos , Animales , Cardiolipinas/metabolismo , Línea Celular , Epitelio/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Histona Desacetilasa 2/metabolismo , Lisina/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Modelos Biológicos , Proteínas Ligasas SKP Cullina F-box/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/efectos de los fármacos
14.
Am J Physiol Cell Physiol ; 313(3): C285-C294, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28637674

RESUMEN

Oxidative stress impacts normal cellular function leading to the pathogenesis of various diseases including pulmonary illnesses. Protein arginine methyltransferase 4 (PRMT4) is critical for normal lung alveolar epithelial cell development; however, the regulation of PRMT4 within such pulmonary diseases has yet to be elucidated. Using biochemical approaches, we uncovered that peroxide (H2O2) treatment decreases PRMT4 protein stability in murine lung epithelial (MLE12) cells to impede cell migration. Protein kinase glycogen synthase kinase 3ß (GSK-3ß) interacts with PRMT4 and catalyzes PRMT4 T132 phosphorylation that protects PRMT4 from ubiquitin proteasomal degradation. H2O2 downregulates GSK-3ß to reduce PRMT4 at protein level. PRMT4 promotes cell migration and H2O2 degrades PRMT4 to inhibit lung epithelial cell migration. These observations demonstrate that oxidative stress destabilizes PRMT4 via GSK-3ß signaling to impede lung epithelial cell migration that may hinder the lung repair and regeneration process.


Asunto(s)
Movimiento Celular/fisiología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Estrés Oxidativo/fisiología , Proteína-Arginina N-Metiltransferasas/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/fisiología , Cicatrización de Heridas/fisiología , Animales , Línea Celular , Estabilidad de Enzimas , Células Epiteliales/citología , Células Epiteliales/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Ratones
15.
J Biol Chem ; 290(29): 18124-18133, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26037928

RESUMEN

The inflammasome is a multiprotein complex that augments the proinflammatory response by increasing the generation and cellular release of key cytokines. Specifically, the NALP3 inflammasome requires two-step signaling, priming and activation, to be functional to release the proinflammatory cytokines IL-1ß and IL-18. The priming process, through unknown mechanisms, increases the protein levels of NALP3 and pro-IL-1ß in cells. Here we show that LPS increases the NALP3 protein lifespan without significantly altering steady-state mRNA in human cells. LPS exposure reduces the ubiquitin-mediated proteasomal processing of NALP3 by inducing levels of an E3 ligase component, FBXO3, which targets FBXL2. The latter is an endogenous mediator of NALP3 degradation. FBXL2 recognizes Trp-73 within NALP3 for interaction and targets Lys-689 within NALP3 for ubiquitin ligation and degradation. A unique small molecule inhibitor of FBXO3 restores FBXL2 levels, resulting in decreased NALP3 protein levels in cells and, thereby, reducing the release of IL-1ß and IL-18 in human inflammatory cells after NALP3 activation. Our findings uncover NALP3 as a molecular target for FBXL2 and suggest that therapeutic targeting of the inflammasome may serve as a platform for preclinical intervention.


Asunto(s)
Proteínas Portadoras/inmunología , Proteínas F-Box/inmunología , Inflamasomas/inmunología , Lipopolisacáridos/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Proteínas Portadoras/metabolismo , Línea Celular , Proteínas F-Box/metabolismo , Humanos , Inmunidad Innata , Inflamasomas/metabolismo , Interleucina-18/inmunología , Interleucina-1beta/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Proteolisis , Proteínas Ligasas SKP Cullina F-box/inmunología , Proteínas Ligasas SKP Cullina F-box/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
16.
J Biol Chem ; 289(10): 7092-7098, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24451372

RESUMEN

Histone acetyltransferase mortality factor 4-like 1 (MORF4L1) is a relatively new histone acetyltransferase component that exists as a homodimer to exert its epigenetic function. The mechanism of MORF4L1 self-assembly is unknown. Here we report that Lys-148 deacetylation is indispensable for facilitating MORF4L1 self-assembly into a homodimeric unit. Among a stretch of ∼10 amino acids in the NH2 terminus between the chromodomain and MORF4-related gene (MRG) domain within MORF4L1, Lys-148 is normally acetylated. Substitution of Lys-148 with arginine augments MORF4L1 self-assembly. However, acetylation mimics of MORF4L1, including K148L and K148Q, abolished its self-assembly of the histone acetyltransferase component. HDAC2, a deacetylase, interacts with and keeps MORF4L1 in a deacetylation status at Lys(148) that triggers MORF4L1 self-assembly. Knockdown of HDAC2 reduces MORF4L1 self-assembly. HDAC2-dependent deacetylation of MORF4L1 enhances MORF4L1 homodimerization, thus facilitating the functionality of complex formation to repress cell proliferation.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Histona Desacetilasa 2/metabolismo , Multimerización de Proteína , Transactivadores/metabolismo , Acetilación , Secuencia de Aminoácidos , Animales , Arginina/química , Línea Celular , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Técnicas de Silenciamiento del Gen , Histona Desacetilasa 2/química , Histona Desacetilasa 2/genética , Ratones , Datos de Secuencia Molecular , Eliminación de Secuencia , Transactivadores/química , Transactivadores/genética
17.
Biochim Biophys Acta ; 1843(4): 694-702, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24389248

RESUMEN

Histone post-translational modification is a key step that may result in an epigenetic mark that regulates chromatin structure and gene transcriptional activity thereby impacting many fundamental aspects of human biology. Subtypes of post-translational modification such as acetylation and methylation are executed by a variety of distinct modification enzymes. The cytoplasmic and nuclear concentrations of these enzymes are dynamically and tightly controlled at the protein level to precisely fine-tune transcriptional activity in response to environmental clues and during pathophysiological states. Recent data have emerged demonstrating that the life span of these critical nuclear enzymes involved in histone modification that impact chromatin structure and gene expression are controlled at the level of protein turnover by ubiquitin-proteasomal processing. This review focuses on the recent progress on mechanisms for ubiquitin-proteasomal degradation of histone modification enzymes and the potential pathophysiological significance of this process.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Proteolisis , Acetilación , Cromatina/metabolismo , Epigénesis Genética , Histona Acetiltransferasas/química , Histona Desacetilasas/química , Histonas/química , Humanos , Metilación , Complejo de la Endopetidasa Proteasomal/genética , Procesamiento Proteico-Postraduccional/genética , Ubiquitina/genética , Ubiquitina/metabolismo
18.
Hepatology ; 59(5): 2010-21, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24122846

RESUMEN

UNLABELLED: Met, the transmembrane tyrosine kinase receptor for hepatocyte growth factor (HGF), is known to function as a potent antiapoptotic mediator in normal and neoplastic cells. Herein we report that the intracellular cytoplasmic tail of Met has evolved to harbor a tandem pair of caspase-3 cleavage sites, which bait, trap, and disable the active site of caspase-3, thereby blocking the execution of apoptosis. We call this caspase-3 cleavage motif the Death Defying Domain (DDD). This site consists of the following sequence: DNAD-DEVD-T (where the hyphens denote caspase cleavage sites). Through functional and mechanistic studies, we show that upon DDD cleavage by caspase-3 the resulting DEVD-T peptide acts as a competitive inhibitor and entraps the active site of caspase-3 akin to DEVD-CHO, which is a potent, synthetic inhibitor of caspase-3 activity. By gain- and loss-of-function studies using restoration of DDD expression in DDD-deficient hepatocytic cells, we found that both caspase-3 sites in DDD are necessary for inhibition of caspase-3 and promotion of cell survival. Employing mutagenesis studies, we show that DDD could operate independently of Met's enzymatic activity as determined by using kinase-dead human Met mutant constructs. Studies of both human liver cancer tissues and cell lines uncovered that DDD cleavage and entrapment of caspase-3 by DDD occur in vivo, further proving that this site has physiological and pathophysiological relevance. CONCLUSION: Met can directly inhibit caspase-3 by way of a novel mechanism and promote hepatocyte survival. The results presented here will further our understanding of the mechanisms that control not only normal tissue homeostasis but also abnormal tissue growth such as cancer and degenerative diseases in which apoptotic caspases are at play.


Asunto(s)
Apoptosis , Caspasa 3/química , Hepatocitos/fisiología , Proteínas Proto-Oncogénicas c-met/fisiología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Caspasa 3/fisiología , Inhibidores de Caspasas/farmacología , Citoprotección , Humanos , Ratones , Datos de Secuencia Molecular , Oligopéptidos/farmacología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-met/química
19.
J Immunol ; 191(10): 5247-55, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24123678

RESUMEN

Cytokine-driven inflammation underlies the pathobiology of a wide array of infectious and immune-related disorders. The TNFR-associated factor (TRAF) proteins have a vital role in innate immunity by conveying signals from cell surface receptors to elicit transcriptional activation of genes encoding proinflammatory cytokines. We discovered that a ubiquitin E3 ligase F box component, termed Fbxo3, potently stimulates cytokine secretion from human inflammatory cells by mediating the degradation of the TRAF inhibitory protein, Fbxl2. Analysis of the Fbxo3 C-terminal structure revealed that the bacterial-like ApaG molecular signature was indispensible for mediating Fbxl2 disposal and stimulating cytokine secretion. By targeting this ApaG motif, we developed a highly unique, selective genus of small-molecule Fbxo3 inhibitors that by reducing TRAF protein levels, potently inhibited cytokine release from human blood mononuclear cells. The Fbxo3 inhibitors effectively lessened the severity of viral pneumonia, septic shock, colitis, and cytokine-driven inflammation systemically in murine models. Thus, pharmacological targeting of Fbxo3 might be a promising strategy for immune-related disorders characterized by a heightened host inflammatory response.


Asunto(s)
Bencilaminas/uso terapéutico , Proteínas F-Box/metabolismo , Inflamación/tratamiento farmacológico , Piridinas/uso terapéutico , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Bencilaminas/farmacología , Línea Celular , Colitis/tratamiento farmacológico , Citocinas/biosíntesis , Citocinas/metabolismo , Proteínas F-Box/antagonistas & inhibidores , Inflamación/inmunología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Piridinas/farmacología , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/metabolismo
20.
J Biol Chem ; 288(9): 6306-16, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23319590

RESUMEN

Histone acetyltransferase binding to origin recognition complex (HBO1) plays a crucial role in DNA replication licensing and cell proliferation, yet its molecular regulation in cells is relatively unknown. Here an uncharacterized protein, Fbxw15, directly interacts with HBO1, a labile protein (t½ = ∼3 h), to mediate its ubiquitination (Lys(338)) and degradation in the cytoplasm. Fbxw15-mediated HBO1 depletion required mitogen-activated protein kinase 1 (Mek1), which was sufficient to trigger HBO1 phosphorylation and degradation in cells. Mek1 ability to produce HBO1 degradation was blocked by Fbxw15 silencing. Lipopolysaccharide induced HBO1 degradation, an effect abrogated by Fbxw15 or Mek1 cellular depletion. Modulation of Fbxw15 levels was able to differentially regulate histone H3K14 acetylation and cellular proliferation by altering HBO1 levels. These studies authenticate Fbxw15 as a ubiquitin E3 ligase subunit that mediates endotoxin-induced HBO1 depletion in cells, thereby controlling cell replicative capacity.


Asunto(s)
Proliferación Celular , Proteínas F-Box/metabolismo , Histona Acetiltransferasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Ubiquitina/metabolismo , Acetilación , Animales , Línea Celular , Proteínas F-Box/genética , Silenciador del Gen , Histona Acetiltransferasas/genética , Histonas/genética , Histonas/metabolismo , Humanos , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , Ratones , Complejo de la Endopetidasa Proteasomal/genética , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA