Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
J Virol ; 98(3): e0000324, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38353538

RESUMEN

The microtubule (MT) is a highly dynamic polymer that functions in various cellular processes through MT hyperacetylation. Thus, many viruses have evolved mechanisms to hijack the MT network of the cytoskeleton to allow intracellular replication of viral genomic material. Coronavirus non-structural protein 8 (nsp8), a component of the viral replication transcriptional complex, is essential for viral survival. Here, we found that nsp8 of porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus with a zoonotic potential, inhibits interferon (IFN)-ß production by targeting melanoma differentiation gene 5 (MDA5), the main pattern recognition receptor for coronaviruses in the cytoplasm. Mechanistically, PDCoV nsp8 interacted with MDA5 and induced autophagy to degrade MDA5 in wild-type cells, but not in autophagy-related (ATG)5 or ATG7 knockout cells. Further screening for autophagic degradation receptors revealed that nsp8 interacts with sequestosome 1/p62 and promotes p62-mediated selective autophagy to degrade MDA5. Importantly, PDCoV nsp8 induced hyperacetylation of MTs, which in turn triggered selective autophagic degradation of MDA5 and subsequent inhibition of IFN-ß production. Overall, our study uncovers a novel mechanism employed by PDCoV nsp8 to evade host innate immune defenses. These findings offer new insights into the interplay among viruses, IFNs, and MTs, providing a promising target to develop anti-viral drugs against PDCoV.IMPORTANCECoronavirus nsp8, a component of the viral replication transcriptional complex, is well conserved and plays a crucial role in viral replication. Exploration of the role mechanism of nsp8 is conducive to the understanding of viral pathogenesis and development of anti-viral strategies against coronavirus. Here, we found that nsp8 of PDCoV, an emerging enteropathogenic coronavirus with a zoonotic potential, is an interferon antagonist. Further studies showed that PDCoV nsp8 interacted with MDA5 and sequestosome 1/p62, promoting p62-mediated selective autophagy to degrade MDA5. We further found that PDCoV nsp8 could induce hyperacetylation of MT, therefore triggering selective autophagic degradation of MDA5 and inhibiting IFN-ß production. These findings reveal a novel immune evasion strategy used by PDCoV nsp8 and provide insights into potential therapeutic interventions.


Asunto(s)
Infecciones por Coronavirus , Deltacoronavirus , Enfermedades de los Porcinos , Animales , Autofagia , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Deltacoronavirus/metabolismo , Interferones/metabolismo , Microtúbulos/metabolismo , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo , Porcinos , Enfermedades de los Porcinos/virología
2.
Int J Mol Sci ; 25(18)2024 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-39337655

RESUMEN

We have previously shown that the overexpression of acetyl-CoA carboxylase 1 (ACC1) was associated with the poor prognosis of cholangiocarcinoma (CCA) patients, and suppression of its expression in CCA cell lines deteriorated cell growth. The present study explored the mechanism by which ACC1 inhibition affects global protein acetylation, using genetic knockdown and pharmacological inhibition with an ACC1 inhibitor ND-646 as models. Both ACC1 knockdown and ACC1-inhibitor-treated cells displayed the hyperacetylation of proteins, accompanied by impaired growth and migration. The immunoprecipitation of hyperacetylated proteins using the anti-acetylated lysine antibody, followed by tandem mass spectrometry, identified three potential verification candidates, namely POTE ankyrin domain family member E, peroxisomal biogenesis factor 1, and heat shock protein 90 beta (HSP90B). HSP90 acetylation was the candidate selected for the verification of protein acetylation. To establish the effects of protein hyperacetylation, treatment with suberoylanilide hydroxamic acid (SAHA), a lysine deacetylase inhibitor, was conducted, and this served as an independent model. Decreased tumor growth but increased acetylated protein levels were observed in ACC1-KD xenograft tumors. Hyperacetylated-alleviated cell growth and migration were consistently observed in the SAHA-treated models. The molecular linkage between protein hyperacetylation and the AKT/GSK3ß/Snail pathway was demonstrated. This study highlighted the importance of protein acetylation in CCA progression, suggesting that ACC1 and KDAC are potential targets for CCA treatment.


Asunto(s)
Acetil-CoA Carboxilasa , Neoplasias de los Conductos Biliares , Movimiento Celular , Proliferación Celular , Colangiocarcinoma , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Colangiocarcinoma/genética , Acetilación , Humanos , Animales , Línea Celular Tumoral , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Neoplasias de los Conductos Biliares/genética , Ratones , Acetil-CoA Carboxilasa/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Genes Dev ; 29(14): 1507-23, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26220994

RESUMEN

NUT midline carcinoma (NMC), a subtype of squamous cell cancer, is one of the most aggressive human solid malignancies known. NMC is driven by the creation of a translocation oncoprotein, BRD4-NUT, which blocks differentiation and drives growth of NMC cells. BRD4-NUT forms distinctive nuclear foci in patient tumors, which we found correlate with ∼100 unprecedented, hyperacetylated expanses of chromatin that reach up to 2 Mb in size. These "megadomains" appear to be the result of aberrant, feed-forward loops of acetylation and binding of acetylated histones that drive transcription of underlying DNA in NMC patient cells and naïve cells induced to express BRD4-NUT. Megadomain locations are typically cell lineage-specific; however, the cMYC and TP63 regions are targeted in all NMCs tested and play functional roles in tumor growth. Megadomains appear to originate from select pre-existing enhancers that progressively broaden but are ultimately delimited by topologically associating domain (TAD) boundaries. Therefore, our findings establish a basis for understanding the powerful role played by large-scale chromatin organization in normal and aberrant lineage-specific gene transcription.


Asunto(s)
Carcinoma de Células Escamosas/fisiopatología , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Elementos de Facilitación Genéticos , Humanos , Proteínas de Neoplasias , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Estructura Terciaria de Proteína , Factores de Transcripción/genética
4.
Int J Mol Sci ; 22(16)2021 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-34445236

RESUMEN

Sepsis-induced liver dysfunction (SILD) is a common event and is strongly associated with mortality. Establishing a causative link between protein post-translational modification and diseases is challenging. We studied the relationship among lysine acetylation (Kac), sirtuin (SIRTs), and the factors involved in SILD, which was induced in LPS-stimulated HepG2 cells. Protein hyperacetylation was observed according to SIRTs reduction after LPS treatment for 24 h. We identified 1449 Kac sites based on comparative acetylome analysis and quantified 1086 Kac sites on 410 proteins for acetylation. Interestingly, the upregulated Kac proteins are enriched in glycolysis/gluconeogenesis pathways in the Kyoto Encyclopedia of Genes and Genomes (KEGG) category. Among the proteins in the glycolysis pathway, hyperacetylation, a key regulator of lactate level in sepsis, was observed at three pyruvate kinase M2 (PKM2) sites. Hyperacetylation of PKM2 induced an increase in its activity, consequently increasing the lactate concentration. In conclusion, this study is the first to conduct global profiling of Kac, suggesting that the Kac mechanism of PKM2 in glycolysis is associated with sepsis. Moreover, it helps to further understand the systematic information regarding hyperacetylation during the sepsis process.


Asunto(s)
Proteínas Portadoras/metabolismo , Lipopolisacáridos/toxicidad , Hígado/enzimología , Proteínas de la Membrana/metabolismo , Sepsis/enzimología , Hormonas Tiroideas/metabolismo , Acetilación/efectos de los fármacos , Células Hep G2 , Humanos , Lisina/metabolismo , Sepsis/inducido químicamente , Proteínas de Unión a Hormona Tiroide
5.
Immunology ; 160(2): 183-197, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32061096

RESUMEN

Under the condition of lipopolysaccharide (LPS)/interferon (IFN)-γ activation, macrophage metabolism is converted from oxidative phosphorylation to glycolysis. In the present work, we analysed whether glycolysis could affect interleukin (IL)-1ß expression through altering histone acetylation levels in mouse bone marrow-derived macrophages. Immunocytochemistry and Western blot analysis are used to characterize histone acetylation in macrophages stimulated by LPS/IFN-γ. Real-time polymerase chain reaction and enzyme-linked immunosorbent assay were used to determine IL-1ß production. The metabolism of macrophages was monitored in real-time by the Seahorse test. Our results showed that glycolytic metabolism could enhance histone acetylation and promote IL-1ß production in LPS/IFN-γ-activated macrophages. Moreover, increased production of IL-1ß by glycolysis was mediated through enhanced H3K9 acetylation. Importantly, it was found that a high dose of histone deacetylase inhibitor could also significantly increase the expression of IL-1ß in the absence of glycolytic metabolism. In conclusion, this study demonstrates that glycolytic metabolism could regulate IL-1ß expression by increasing histone acetylation levels in LPS/IFN-γ-stimulated macrophages.


Asunto(s)
Histonas/metabolismo , Interleucina-1beta/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Acetilación , Animales , Células Cultivadas , Glucólisis/inmunología , Interferón gamma/inmunología , Lipopolisacáridos/inmunología , Macrófagos/metabolismo , Ratones , Cultivo Primario de Células , Proteínas Recombinantes/inmunología
6.
Pharmacol Res ; 159: 104887, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32526680

RESUMEN

Sepsis induced myocardial dysfunction (SIMD) results in high morbidity and mortality. However, the effective therapeutic strategies for SIMD treatment remain limited. Sirt3 is the main mitochondrial Sirtuin member and is a key modulator of mitochondrial metabolism and function. In this study, we aimed to investigate the effect and mechanism of Sirt3 on SIMD. SIMD was induced by 20 mg/kg Lipopolysaccharides (LPS) injection for 6 h in mice. Sepsis could induce the reduction of cardiac Sirt3 expression and global deficiency of Sirt3 exacerbated cardiac function. Quantitative acetyl-proteomics and cardiac metabolomics analysis revealed that loss of Sirt3 led to hyper-acetylation of critical enzymes within cardiac tricarboxylic acid (TCA) cycle and generation of lactate and NADH, subsequently promotion of cardiac dysfunction after sepsis. Additionally, to evaluate whether Emodin could be utilized as a potential Sirt3 modulator to treat SIMD, male wild type mice (WT mice) or global Sirt3 deficient mice (Sirt3-/- mice) were intraperitoneally injected with 40 mg/kg Emodin for 5 days followed by 20 mg/kg LPS administration for another 6 h and observed that exogenous administration of Emodin could attenuate myocardial dysfunction in septic WT mice. However, septic Sirt3-/- mice can not gain benefit on cardiac performance from Emodin infusion. In conclusion, this study presented the protective role of Sirt3 targeting SIMD, which may provide a potential novel approach to maintain normal cardiac performance after sepsis.


Asunto(s)
Ciclo del Ácido Cítrico , Cardiopatías/enzimología , Mitocondrias Cardíacas/enzimología , Miocitos Cardíacos/enzimología , Sepsis/enzimología , Sirtuina 3/metabolismo , Acetilación , Animales , Ciclo del Ácido Cítrico/efectos de los fármacos , Modelos Animales de Enfermedad , Emodina/farmacología , Cardiopatías/etiología , Cardiopatías/fisiopatología , Cardiopatías/prevención & control , Lipopolisacáridos , Masculino , Metabolómica , Ratones Noqueados , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/ultraestructura , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/ultraestructura , Procesamiento Proteico-Postraduccional , Sepsis/inducido químicamente , Sepsis/tratamiento farmacológico , Sepsis/fisiopatología , Sirtuina 3/deficiencia , Sirtuina 3/genética
7.
Proc Natl Acad Sci U S A ; 114(21): E4184-E4192, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28484033

RESUMEN

To investigate the mechanism that drives dramatic mistargeting of active chromatin in NUT midline carcinoma (NMC), we have identified protein interactions unique to the BRD4-NUT fusion oncoprotein compared with wild-type BRD4. Using cross-linking, affinity purification, and mass spectrometry, we identified the EP300 acetyltransferase as uniquely associated with BRD4 through the NUT fusion in both NMC and non-NMC cell types. We also discovered ZNF532 associated with BRD4-NUT in NMC patient cells but not detectable in 293T cells. EP300 and ZNF532 are both implicated in feed-forward regulatory loops leading to propagation of the oncogenic chromatin complex in BRD4-NUT patient cells. Adding key functional significance to our biochemical findings, we independently discovered a ZNF532-NUT translocation fusion in a newly diagnosed NMC patient. ChIP sequencing of the major players NUT, ZNF532, BRD4, EP300, and H3K27ac revealed the formation of ZNF532-NUT-associated hyperacetylated megadomains, distinctly localized but otherwise analogous to those found in BRD4-NUT patient cells. Our results support a model in which NMC is dependent on ectopic NUT-mediated interactions between EP300 and components of BRD4 regulatory complexes, leading to a cascade of misregulation.


Asunto(s)
Carcinoma de Células Escamosas/patología , Cromatina/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Neoplasias Pulmonares/patología , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Carcinoma de Células Escamosas/genética , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/genética , Células Epiteliales/patología , Femenino , Células HEK293 , Humanos , Hibridación Fluorescente in Situ , Neoplasias Pulmonares/genética , Persona de Mediana Edad , Complejos Multiproteicos/genética , Proteínas de Neoplasias , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Dominios Proteicos/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Dedos de Zinc/genética
8.
FASEB J ; 32(9): 5143-5161, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29913563

RESUMEN

The histidine triad nucleotide-binding protein 2 (HINT-2) is a mitochondrial adenosine phosphoramidase expressed in hepatocytes. The phenotype of Hint2 knockout ( Hint2-/-) mice includes progressive hepatic steatosis and lysine hyperacetylation of mitochondrial proteins, which are features of respiratory chain malfunctions. We postulated that the absence of HINT-2 induces a defect in mitochondria bioenergetics. Isolated Hint2-/- hepatocytes produced less ATP and generated a lower mitochondrial membrane potential than did Hint2+/+ hepatocytes. In extracellular flux analyses with glucose, the basal, ATP-linked, and maximum oxygen consumption rates (OCRs) were decreased in Hint2-/- hepatocytes and in HepG2 cells lacking HINT-2. Conversely, in HINT-2 overexpressing SNU-449 and HepG2 cells, the basal, ATP-linked, and maximum OCRs were increased. Similarly, with palmitate, basal and maximum OCRs were decreased in Hint2-/- hepatocytes, but they were increased in HINT-2 overexpressing HepG2 cells. When assayed with radiolabeled substrate, palmitate oxidation was reduced by 25% in Hint2-/- mitochondria. In respirometry assays, complex I- and II-driven, coupled and uncoupled respirations and complex IV KCN-sensitive respiration were reduced in Hint2-/- mitochondria. Furthermore, HINT-2 associated with cardiolipin and glucose-regulated protein 75 kDa. Our study shows decreased electron transfer and oxidative phosphorylation capacity in the absence of HINT-2. The bioenergetics deficit accumulated over time in hepatocytes lacking HINT-2 likely leads to the secondary outcome of steatosis.-Rajasekaran, R., Felser, A., Nuoffer, J.-M., Dufour, J.-F., St-Pierre, M. V. The histidine triad nucleotide-binding protein 2 (HINT-2) positively regulates hepatocellular energy metabolism.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Metabolismo Energético/fisiología , Neoplasias Hepáticas/metabolismo , Proteínas Mitocondriales/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Carcinoma Hepatocelular/fisiopatología , Línea Celular Tumoral , Respiración de la Célula/fisiología , Transporte de Electrón/fisiología , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/fisiología , Humanos , Neoplasias Hepáticas/fisiopatología , Masculino , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/fisiología , Fosforilación Oxidativa
9.
Zygote ; 27(5): 305-314, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31412961

RESUMEN

Male germ cell development is a critical period during which epigenetic patterns are established and maintained. The progression from diploid spermatogonia to haploid spermatozoa involves the incorporation of testis-specific histone variants, mitotic and meiotic divisions, haploid gene expression, histone-protamine transitions and massive epigenetic reprogramming. Understanding the protein players and the epigenetic mark network involved in the setting of the epigenetic programme in spermatogenesis is an exciting new clue in the field of reproductive biology with translational outcomes. As information in the existing literature regarding cross-talk between DNA methylation and histone hyperacetylation in the advanced stages of murine spermatogenesis is still scarce and controversial we have investigated the effect of a DNA-methyltransferase inhibitor, 5-aza-2'-deoxycytidine, at the cytological and molecular level (by transmission electron microscopy, immunocytochemistry and immunoprecipitation methods). Our results revealed an important role for regulation of DNA methylation in controlling histone hyperacetylation and chromatin remodelling during spermatogenesis.


Asunto(s)
Metilación de ADN , Histonas/metabolismo , Espermatogénesis/fisiología , Acetilación , Animales , Inmunoprecipitación de Cromatina , Metilasas de Modificación del ADN/antagonistas & inhibidores , Decitabina/farmacología , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Histonas/genética , Masculino , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Espermatogonias/efectos de los fármacos , Espermatogonias/patología , Testículo/efectos de los fármacos , Testículo/patología
10.
J Cell Mol Med ; 22(3): 1909-1922, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29327812

RESUMEN

Therapeutic agents are urgently needed for treating metastatic castration-refractory prostate cancer (mCRPC) that is unresponsive to androgen deprivation and chemotherapy. Our screening assays demonstrated that chemotherapy-resistant prostate cancer (PCa) cells are more sensitive to HDAC inhibitors than paired sensitive PCa cells, as demonstrated by cell proliferation and apoptosis in vitro and in vivo. Kinetic study revealed that TSA-induced apoptosis was significantly dependent on enhanced transcription and protein synthesis in an early stage, which subsequently caused ER stress and apoptosis. ChIP analysis indicated that TSA increased H4K16 acetylation, promoting ER stress gene transcription. The changes in Ac-H4K16, ATF3 and ATF4 were also validated in TSA-treated animals. Further study revealed the higher enzyme activity of HDACs and an increase in acetylated proteins in resistant cells. The higher nucleocytoplasmic acetyl-CoA in resistant cells was responsible for elevated acetylation status of protein and a more vigorous growth state. These results strongly support the pre-clinical application of HDAC inhibitors for treating chemotherapy-resistant mCRPC.


Asunto(s)
Acetilcoenzima A/metabolismo , Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Adaptadoras Transductoras de Señales , Aloinjertos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Docetaxel/farmacología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factores Eucarióticos de Iniciación , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Carga Tumoral/efectos de los fármacos
11.
Biochim Biophys Acta Gen Subj ; 1862(8): 1729-1741, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29746960

RESUMEN

BACKGROUND: p300 (KAT3B) lysine acetyltransferase activity is modulated under different physiological and pathological contexts through the induction of trans-autoacetylation. This phenomenon is mediated by several factors, mechanisms of which are not fully understood. METHODS: Through acetyltransferase assays using full-length, baculovirus-expressed KATs, the specificity of NPM1-mediated enhancement of p300 autoacetylation was tested. Chaperone assays and tryptophan fluorescence studies were performed to evaluate the NPM1-induced protein folding. The NPM1 oligomer-defective mutant characterization was done by glutaraldehyde-crosslinking. The small-molecule inhibitor of NPM1 oligomerization was used to confirm the absolute requirement of multimeric NPM1 in vivo. Immunohistochemistry analysis of oral cancer patient samples was done to uncover the pathophysiological significance of NPM1-induced p300 autoacetylation. RESULTS: We find that the histone chaperone NPM1 is a specific inducer of p300 autoacetylation. Distinct from its histone chaperone activity, NPM1 is a molecular chaperone of p300. The biophysical experiments suggest that there is a reversible binding between NPM1 and p300 which can modulate p300 acetyltransferase activity. Disruption of NPM1 oligomerization suggests that oligomeric NPM1 is essential for the induction of p300 autoacetylation. Significantly, we observe a concomitant hyper-autoacetylation of p300 with overexpression of NPM1 in oral cancer samples. CONCLUSION: NPM1 can specifically modulate p300 acetyltransferase activity through the enhancement of autoacetylation. The molecular chaperone activity and oligomerization of NPM1 play a pivotal role in this phenomenon. GENERAL SIGNIFICANCE: NPM1 is overexpressed in several solid cancers, the significance of which is unknown. Induction of p300 autoacetylation could be the cause of NPM1-mediated tumorigenicity.


Asunto(s)
Proteína p300 Asociada a E1A/química , Proteína p300 Asociada a E1A/metabolismo , Histonas/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Pliegue de Proteína , Multimerización de Proteína , Neoplasias de la Lengua/metabolismo , Acetilación , Humanos , Nucleofosmina , Unión Proteica , Conformación Proteica , Neoplasias de la Lengua/patología , Células Tumorales Cultivadas
12.
Cell Biol Int ; 42(9): 1160-1169, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29741224

RESUMEN

The high-mobility group box-1 (HMGB1), as a highly conserved ubiquitous DNA-binding protein, has been widely studied in various diseases, including inflammation and tumor; however, fewer studies were focused on the mechanisms controlling HMGB1 release compared with the function of HMGB1. Previous studies have proven that ANG II can act as a pro-inflammatory cytokine, both of HMGB1 and ANG II were significantly upregulated in autoimmune diseases; however, the exact role of ANG II in regulating HMGB1 release have not been shown. The present study was to define the effects of ANG II on macrophages and the possible mechanisms in controlling HMGB1 release. Our results showed that ANG II can induce M1 macrophage polarization through upregulated the expression of HMGB1 and caused acetylation of HMGB1 and release via its dissociation from SIRT1, which in a positive feedback upregulates ANG II. Subsequently, HMGB1 inhibitors can reduce the ANG II-elicited polarize of macrophage. Meanwhile, we show that JAK/STAT pathways play an essential role in ANG II-induced HMGB1 nuclear translocation, JAK/STAT specific inhibitors can inhibit ANG II-induced HMGB1 expression. Taken together, our results provide a novel evidence that HMGB1 play a critical role in ANG II mediated macrophage polarization, and we suggest that ANG II mediated HMGB1 release via dissociation from SIRT1, induce hyperacetylation of HMGB1, thus for subsequent release, suggesting that the angiotensin II receptor antagonist is a potential drug target for inhibiting HMGB1 release in inflammation diseases.


Asunto(s)
Angiotensina II/metabolismo , Proteína HMGB1/metabolismo , Acetilación , Animales , Polaridad Celular/fisiología , Citocinas/sangre , Citocinas/metabolismo , Proteína HMGB1/biosíntesis , Proteína HMGB1/genética , Inflamación/metabolismo , Macrófagos Peritoneales/metabolismo , Ratones , Transporte de Proteínas , Células RAW 264.7 , Transducción de Señal , Sirtuina 1/metabolismo
13.
J Mol Cell Cardiol ; 112: 64-73, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28882480

RESUMEN

Heart failure is associated with mitochondrial dysfunction so that restoring or improving mitochondrial health is of therapeutic importance. Recently, reduction in NAD+ levels and NAD+-mediated deacetylase activity has been recognized as negative regulators of mitochondrial function. Using a cardiac specific KLF4 deficient mouse line that is sensitive to stress, we found mitochondrial protein hyperacetylation coupled with reduced Sirt3 and NAD+ levels in the heart before stress, suggesting that the KLF4-deficient heart is predisposed to NAD+-associated defects. Further, we demonstrated that short-term administration of Nicotinamide Mononucleotide (NMN) successfully protected the mutant mice from pressure overload-induced heart failure. Mechanically, we showed that NMN preserved mitochondrial ultrastructure, reduced ROS and prevented cell death in the heart. In cultured cardiomyocytes, NMN treatment significantly increased long-chain fatty acid oxidation despite no direct effect on pyruvate oxidation. Collectively, these results provide cogent evidence that hyperacetylation of mitochondrial proteins is critical in the pathogenesis of cardiac disease and that administration of NMN may serve as a promising therapy.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/prevención & control , Homeostasis , Mononucleótido de Nicotinamida/administración & dosificación , Mononucleótido de Nicotinamida/uso terapéutico , Acetilación , Animales , Muerte Celular , Ácidos Grasos/metabolismo , Insuficiencia Cardíaca/patología , Homeostasis/efectos de los fármacos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/metabolismo , NAD/metabolismo , Mononucleótido de Nicotinamida/farmacología , Nicotinamida Fosforribosiltransferasa/metabolismo , Oxidación-Reducción , Presión , Ratas , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3/metabolismo
14.
Apoptosis ; 22(9): 1079-1097, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28647884

RESUMEN

Fluoxetine (FLX) is an antidepressant drug that belongs to the class of selective serotonin reuptake inhibitors. FLX is known to induce apoptosis in multiple types of cancer cells. In this study, the molecular mechanisms underlying the anti-cancer effects of FLX were investigated in SK-N-BE(2)-M17 human neuroblastoma cells. FLX induced apoptotic cell death, activation of caspase-4, -9, and -3, and expression of endoplasmic reticulum (ER) stress-associated proteins, including C/EBP homologous protein (CHOP). Inhibition of ER stress by treatment with the ER stress inhibitors, salubrinal and 4-phenylbutyric acid or CHOP siRNA transfection reduced FLX-induced cell death. FLX induced phosphorylation of mitogen-activated protein kinases (MAPKs) family, p38, JNK, and ERK, and an upstream kinase apoptosis signal kinase 1 (ASK1). Inhibition of MAPKs and ASK1 reduced FLX-induced cell death and CHOP expression. We then showed that FLX reduced mitochondrial membrane potential (MMP) and ER stress inhibitors as well as MAPK inhibitors ameliorated FLX-induced loss of MMP. Interestingly, FLX induced hyperacetylation of histone H3 and H4, upregulation of p300 histone acetyltransferase (HAT), and downregulation of histone deacetylases (HDACs). Treatment with a HAT inhibitor anacardic acid or p300 HAT siRNA transfection blocked FLX-induced apoptosis in SK-N-BE(2)-M17 cells. However, FLX did not induce histone acetylation and anacardic acid had no protective effect on FLX-induced cell death and CHOP expression in MYCN non-amplified SH-SY5Y human neuroblastoma and MYCN knockdowned SK-N-BE(2)-M17 cells. These findings suggest that FLX induces apoptosis in neuroblastoma through ER stress and mitochondrial dysfunction via the ASK1 and MAPK pathways and through histone hyperacetylation in a MYCN-dependent manner.


Asunto(s)
Acetilación/efectos de los fármacos , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Fluoxetina/farmacología , Histonas/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Neuroblastoma/patología , Animales , Antineoplásicos/farmacología , Caspasas Iniciadoras/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , MAP Quinasa Quinasa Quinasa 5/genética , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción CHOP/antagonistas & inhibidores , Factor de Transcripción CHOP/genética
15.
Biochem Biophys Res Commun ; 486(1): 83-87, 2017 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-28263745

RESUMEN

Exercise brings changes on the chromatin ensuing the upregulation of many genes that confer protection from type 2 diabetes. In type-2 diabetes, critical genes are down-regulated such as those involved in glucose transport (GLUT4, MEF2A) and also oxidative phosphorylation (NRF-1 and its target genes). Recent reports have shown that NRF-1 not only regulate mitochondrial oxidative genes but also controls MEF2A, the main transcription factor for glucose transporter, GLUT4. Such dual control of the two pathways by NRF-1 place it as critical gene in the design of therapeutic modalities much needed to cure or better manage type 2 diabetes. Although it is known that NRF-1 controls these dual pathways (glucose transport and oxidative phosphorylation), the actual molecular mechanisms involved surrounding this regulation remains elusive. NRF-1 itself is regulated through posttranslational modifications (acetylation, methylation and phosphorylation) resulting in enhanced binding to its target genes. This study is therefore aimed at assessing whether CaMKII, a kinase activated by exercise brings about hyper-acetylation of histones in the vicinity of NRF-1 target gene, Mef2a. Five to six weeks old male Wistar rats were used in this study. Chromatin immunoprecipitation (ChIP) assay was used to investigate the extent through which NRF-1 is bound to the Mef2a gene and if this was associated with hyper-acetylation of histones in the region of NRF-1 binding site of the Mef2a gene. Quantitative real time PCR (qPCR) was used to determine the gene expression of MEF2A and NRF-1. Results from this study indicated that exercise-induced CaMKII activation increased hyper-acetylation of histones in the region of NRF-1 binding site on vicinity of Mef2a gene and this was associated with the increased binding of NRF-1 to Mef2a gene. Exercise also increased the expression of NRF-1 and MEF2A genes. Administration of CaMKII inhibitor (KN93) prior to exercise attenuated the observed exercise-induced increase of NRF-1 and MEF2A expressions. In conclusion, this study demonstrated for the first time in our knowledge one mechanism through which NRF-1 regulates MEF2A, pathway critical in glucose transport.


Asunto(s)
Histonas/metabolismo , Factor 1 Relacionado con NF-E2/genética , Condicionamiento Físico Animal , Regiones Promotoras Genéticas/genética , Acetilación , Animales , Bencilaminas/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Factor 1 Relacionado con NF-E2/metabolismo , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Ratas Wistar , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfonamidas/farmacología
16.
Invest New Drugs ; 35(6): 691-705, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28776290

RESUMEN

Histone deacetylases (HDACs) play an important role in the epigenetic regulation of gene expression through their effects on the compact chromatin structure. In clinical studies, several classes of histone deacetylase inhibitors (HDACi) have demonstrated potent anticancer activities with metal complexes. Hence, we synthesized cadmium-proline complexes using both the D- and L-isomers of proline and evaluated their biological activities by observing the efficiency of their inhibition of HDAC activity, ability to reduce the expression of HDAC isoforms in A549 cells and effect on apoptosis. The synthesized compounds were characterized by UV, IR, NMR spectroscopy and elemental analysis. In-vitro cell toxicity was evaluated by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, and the 50% inhibitory concentration (IC50; 2 µM) was obtained at 12 h. The morphological study at nuclear levels was performed by acridine orange/ethidium bromide (AO/EB) and Hoechst staining, and the results showed an association with cell cycle arrest at the G2/M phase. Both cadmium-proline complexes intensely inhibited HDAC activity at 2 µM concentration. Interestingly, Cd[L-proline]2 was found to be a potent inhibitor for all HDAC isoforms, whereas Cd[D-proline]2 inhibited only HDAC1 and 2. HDACi are novel chemotherapeutic drugs that induce hyperacetylation of histones H3 and H4, counteracting the aberrant repression of genes, such as insulin-like growth factor-binding protein 3 (IGFBP-3), p53, and p21. ERK/MAPK signaling pathway resulted in the downregulation of the expression of matrix metalloproteinases 2 and 9 (MMP-2 and MMP-9), contributing to the inhibition of metastasis in A549 cells. Apoptosis induction was accompanied by the activation of death receptors and their ligands which recruit initiator caspase 8, decrease in mitochondrial membrane potential (ΔΨm), as well as increased Bax/Bcl2 ratio, followed by activation of caspases 9 and 3. Our finding suggests that Cd[L-proline]2 complex accelerates epigenetic rearrangement by HDAC inhibition, which may be the key mechanism for its anticancer activity.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos de Cadmio/química , Compuestos de Cadmio/farmacología , Epigénesis Genética , Histona Desacetilasa 1/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Prolina/química , Células A549 , Acetilación , Puntos de Control del Ciclo Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/química , Humanos , Isoenzimas
17.
Biochem J ; 473(12): 1821-30, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27099338

RESUMEN

Lysine acetylation is tightly coupled to the nutritional status of the cell, as the availability of its cofactor, acetyl-CoA, fluctuates with changing metabolic conditions. Recent studies have demonstrated that acetyl-CoA levels act as an indicator of cellular nourishment, and increased abundance of this metabolite can block the induction of cellular recycling programmes. In the present study we investigated the cross-talk between mitochondrial metabolic pathways, acetylation and autophagy, using chemical inducers of mitochondrial acetyl-CoA production. Treatment of cells with α-lipoic acid (αLA), a cofactor of the pyruvate dehydrogenase complex, led to the unexpected hyperacetylation of α-tubulin in the cytosol. This acetylation was blocked by pharmacological inhibition of mitochondrial citrate export (a source for mitochondria-derived acetyl-CoA in the cytosol), was dependent on the α-tubulin acetyltransferase (αTAT) and was coupled to a loss in function of the cytosolic histone deacetylase, HDAC6. We further demonstrate that αLA slows the flux of substrates through autophagy-related pathways, and severely limits the ability of cells to remove depolarized mitochondria through PTEN-associated kinase 1 (PINK1)-mediated mitophagy.


Asunto(s)
Mitocondrias/metabolismo , Ácido Tióctico/farmacología , Tubulina (Proteína)/metabolismo , Acetilcoenzima A/metabolismo , Acetilación/efectos de los fármacos , Acetiltransferasas/metabolismo , Animales , Autofagia/efectos de los fármacos , Células COS , Chlorocebus aethiops , Células Hep G2 , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Microscopía Confocal , Mitocondrias/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
18.
Proc Natl Acad Sci U S A ; 111(46): E4920-8, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25368158

RESUMEN

In cardiomyocytes, calcium is known to control gene expression at the level of transcription, whereas its role in regulating alternative splicing has not been explored. Here we report that, in mouse primary or embryonic stem cell-derived cardiomyocytes, increased calcium levels induce robust and reversible skipping of several alternative exons from endogenously expressed genes. Interestingly, we demonstrate a calcium-mediated splicing regulatory mechanism that depends on changes of histone modifications. Specifically, the regulation occurs through changes in calcium-responsive kinase activities that lead to alterations in histone modifications and subsequent changes in the transcriptional elongation rate and exon skipping. We demonstrate that increased intracellular calcium levels lead to histone hyperacetylation along the body of the genes containing calcium-responsive alternative exons by disrupting the histone deacetylase-to-histone acetyltransferase balance in the nucleus. Consequently, the RNA polymerase II elongation rate increases significantly on those genes, resulting in skipping of the alternative exons. These studies reveal a mechanism by which calcium-level changes in cardiomyocytes impact on the output of gene expression through altering alternative pre-mRNA splicing patterns.


Asunto(s)
Empalme Alternativo , Señalización del Calcio/fisiología , Histona Desacetilasas/fisiología , Histonas/metabolismo , Miocitos Cardíacos/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Acetilación , Empalme Alternativo/efectos de los fármacos , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Exones , Regulación de la Expresión Génica/fisiología , Genes de Neurofibromatosis 1 , Ratones , Miocitos Cardíacos/efectos de los fármacos , Neurofibromina 1/biosíntesis , Neurofibromina 1/genética , Cloruro de Potasio/farmacología , Inhibidores de Proteínas Quinasas/farmacología , ARN Polimerasa II/metabolismo , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Canales Catiónicos TRPP/fisiología , Elongación de la Transcripción Genética
19.
Biochim Biophys Acta ; 1850(12): 2530-43, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26432600

RESUMEN

BACKGROUND: Current treatment options for cachexia, which impairs disease prognosis, are limited. Muscle-enriched microRNAs and protein acetylation are involved in muscle wasting including lung cancer (LC) cachexia. Poly(ADP-ribose) polymerases (PARP) are involved in muscle metabolism. We hypothesized that muscle-enriched microRNA, protein hyperacetylation, and expression levels of myogenic transcription factors (MTFs) and downstream targets, muscle loss and function improve in LC cachectic Parp-1(−/−) and Parp-2(−/−) mice. METHODS: Body and muscle weights, grip strength, muscle phenotype, muscle-enriched microRNAs (miR-1, -133, -206, and -486), protein acetylation, acetylated levels of FoxO1, FoxO3, and PGC-1α, histone deacetylases (HDACs) including SIRT1, MTFs, and downstream targets (α-actin, PGC-1α, and creatine kinase) were evaluated in diaphragm and gastrocnemius of LC (LP07 adenocarcinoma) wild type (WT), Parp-1(−/−) and Parp-2−/− mice. RESULTS: Compared to WT cachectic animals, in both respiratory and limb muscles of Parp-1(−/−) and Parp-2(−/−) cachectic mice: downregulation of muscle-specific microRNAs was counterbalanced especially in gastrocnemius of Parp-1(−/−) mice; increased protein acetylation was attenuated (improvement in HDAC3, SIRT-1, and acetylated FoxO3 levels in both muscles, acetylated FoxO1 levels in the diaphragm); reduced MTFs and creatine kinase levels were mitigated; body and muscle weights, strength, and muscle fiber sizes improved, while tumor weight and growth decreased. CONCLUSIONS: These molecular findings may explain the improvements seen in body and muscle weights, limb muscle force and fiber sizes in both Parp-1(−/−) and Parp-2(−/−) cachectic mice. GENERAL SIGNIFICANCE: PARP-1 and -2 play a role in cancer-induced cachexia, thus selective pharmacological inhibition of PARP-1 and -2 may be of interest in clinical settings.


Asunto(s)
Caquexia/metabolismo , Neoplasias Pulmonares/metabolismo , MicroARNs/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Acetilación , Animales , Caquexia/genética , Neoplasias Pulmonares/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética
20.
Biochem Biophys Res Commun ; 463(4): 832-8, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26056001

RESUMEN

Steatosis is the earliest and most common disease of the liver due to chronic ethanol consumption, and stems from alterations in the function of transcription factors related to lipid metabolism. Protein acetylation at the lysine residue (Kac) is known to have diverse functions in cell metabolism. Recent studies showed that ethanol exposure induces global protein hyperacetylation by reducing the deacetylase activities of SIRT1 and SIRT3. Although global acetylome analyses have revealed the involvement of a variety of lysine acetylation sites, the exact sites directly regulated by ethanol exposure are unknown. In this study, to elucidate the exact hyperacetylation sites that contribute to SIRT1 and SIRT3 downregulation, we identified and quantified a total of 1285 Kac sites and 686 Kac proteins in AML-12 cells after ethanol treatment (100 mM) for 3 days. All quantified Kac sites were divided into four quantiles: Q1 (0-15%), Q2 (15-50%), Q3 (50-85%), and Q4 (85-100%). Q4 had 192 Kac sites indicating ethanol-induced hyperacetylation. Using the Motif-x program, the [LXKL], [KH], and [KW] motifs were included in the Q4 category, where [KW] was a specific residue for SIRT3. We also performed gene ontology term and KEGG pathway enrichment analyses. Hyperacetylation sites were significantly enriched in biosynthetic processes and ATPase activities within the biological process and molecular function categories, respectively. In conclusion, ethanol regulates the acetylation of proteins in a variety of metabolic pathways mediated by SIRT1 and SIRT3. As a result, ethanol stimulates increased de novo fatty acid synthesis in hepatocytes.


Asunto(s)
Etanol/farmacología , Hígado Graso/inducido químicamente , Hepatocitos/efectos de los fármacos , Histona Acetiltransferasas/metabolismo , Acetilación , Animales , Línea Celular , Hígado Graso/fisiopatología , Ratones , Espectrometría de Masas en Tándem
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda