Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
FASEB J ; 36(3): e22170, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35104011

RESUMEN

Chronic endoplasmic reticulum (ER) stress in hepatocytes plays a role in the pathogenesis of nonalcoholic fatty liver disease. Therefore, given the association between oxidative stress, mitochondrial dysfunction, and ER stress, our study investigated the role of NRF2-mediated SIRT3 activation in ER stress. SIRT3, a sirtuin, was predicted as the target of NRF2 based on bioinformatic analyses and animal experiments. Nrf2 abrogation diminished mitochondrial DNA content in hepatocytes with Ppargc1α and Cpt1a inhibition, whereas its overexpression enhanced oxygen consumption. Further, chromatin immunoprecipitation and luciferase reporter assays indicated that NRF2 induced SIRT3 through the antioxidant responsive element (ARE) sites comprising the -641 to -631 bp and -419 to -409 bp regions. In tunicamycin-induced ER stress conditions and liver injury animal models following ER stress, NRF2 levels were highly correlated with SIRT3. Nrf2 deficiency enhanced the tunicamycin-mediated induction of CHOP, which was attenuated by Sirt3 overexpression. Further, Sirt3 delivery to hepatocytes in Nrf2 knockout mice prevented tunicamycin from increasing mortality by decreasing ER stress. SIRT3 was upregulated in livers of patients with nonalcoholic liver diseases, whereas lower SIRT3 expression coincided with more severe disease conditions. Taken together, our findings indicated that NRF2-mediated SIRT3 induction protects hepatocytes from ER stress-induced injury, which may contribute to the inhibition of liver disease progression.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Hepatocitos/metabolismo , Hepatopatías/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Sustancias Protectoras/metabolismo , Sirtuina 3/metabolismo , Animales , Antioxidantes/metabolismo , Línea Celular , ADN Mitocondrial/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células HEK293 , Hepatocitos/efectos de los fármacos , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Factor de Transcripción CHOP/metabolismo , Tunicamicina/farmacología
2.
Hepatology ; 73(4): 1307-1326, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32557804

RESUMEN

BACKGROUND AND AIMS: Fat accumulation results from increased fat absorption and/or defective fat metabolism. Currently, the lipid-sensing nuclear receptor that controls fat utilization in hepatocytes is elusive. Liver X receptor alpha (LXRα) promotes accumulation of lipids through the induction of several lipogenic genes. However, its effect on lipid degradation is open for study. Here, we investigated the inhibitory role of LXRα in autophagy/lipophagy in hepatocytes and the underlying basis. APPROACH AND RESULTS: In LXRα knockout mice fed a high-fat diet, or cell models, LXRα activation suppressed the function of mitochondria by inhibiting autophagy/lipophagy and induced hepatic steatosis. Gene sets associated with "autophagy" were enriched in hepatic transcriptome data. Autophagy flux was markedly augmented in the LXRα knockout mouse liver and primary hepatocytes. Mechanistically, LXRα suppressed autophagy-related 4B cysteine peptidase (ATG4B) and Rab-8B, responsible for autophagosome and -lysosome formation, by inducing let-7a and microRNA (miR)-34a. Chromatin immunoprecipitation assay enabled us to find LXRα as a transcription factor of let-7a and miR-34a. Moreover, 3' untranslated region luciferase assay substantiated the direct inhibitory effects of let-7a and miR-34a on ATG4B and Rab-8B. Consistently, either LXRα activation or the let-7a/miR-34a transfection lowered mitochondrial oxygen consumption rate and mitochondrial transmembrane potential and increased fat levels. In obese animals or nonalcoholic fatty liver disease (NAFLD) patients, let-7a and miR-34a levels were elevated with simultaneous decreases in ATG4B and Rab-8B levels. CONCLUSIONS: LXRα inhibits autophagy in hepatocytes through down-regulating ATG4B and Rab-8B by transcriptionally activating microRNA let-7a-2 and microRNA 34a genes and suppresses mitochondrial biogenesis and fuel consumption. This highlights a function of LXRα that culminates in the progression of liver steatosis and steatohepatitis, and the identified targets may be applied for a therapeutic strategy in the treatment of NAFLD.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia/fisiología , Cisteína Endopeptidasas/metabolismo , Hepatocitos/metabolismo , Receptores X del Hígado/metabolismo , Mitocondrias/fisiología , Proteínas de Unión al GTP rab/metabolismo , Activación Metabólica , Animales , Autofagia/genética , Proteínas Relacionadas con la Autofagia/genética , Cisteína Endopeptidasas/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación hacia Abajo , Hígado Graso/etiología , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/fisiopatología , Células Hep G2/metabolismo , Células Hep G2/fisiología , Hepatocitos/fisiología , Humanos , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Hígado/fisiología , Hígado/fisiopatología , Receptores X del Hígado/genética , Receptores X del Hígado/fisiología , Ratones , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , MicroARNs/fisiología , Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Biogénesis de Organelos , Consumo de Oxígeno/genética , Consumo de Oxígeno/fisiología , Transcriptoma , Proteínas de Unión al GTP rab/genética
3.
FASEB J ; 33(7): 7953-7969, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30897343

RESUMEN

Long noncoding RNA (lncRNA) capable of controlling antioxidative capacity remains to be investigated. Nuclear factor erythroid-2-related factor 2 (Nrf2) is a central molecule for cellular defense that increases antioxidative capacity. We identified a novel lncRNA named Nrf2-activating lncRNA (Nrf2-lncRNA) transcribed from an upstream region of the microRNA 122 gene (MIR122). Nrf2-lncRNA existed in the cytoplasm, suggestive of its function as a competing endogenous RNA [ceRNA, microRNA (miRNA) sponge]. Nrf2-lncRNA served as a ceRNA for polo-like kinase (Plk) 2 and cyclin-dependent kinase inhibitor 1 (p21cip1) through binding of miRNA 128 and miRNA 224, inducing Plk2/Nrf2/p21cip1 complexation for Nrf2 activation in the cells under p53-activating conditions (i.e., DNA damage and serum deprivation). Nrf2-lncRNA expression was suppressed with the initiation of apoptosis, being a rheostat for cell fate determination. Nrf2-lncRNA levels correlated with the recurrence-free postsurgery survival rate of patients with hepatocellular carcinoma. Collectively, Nrf2-lncRNA promotes Plk2 and p21cip1 translation by competing for specific miRNAs and activating Nrf2 under surviving conditions from oxidative stress, implying that Nrf2-lncRNA serves as a fine-tuning rheostat for cell fate decision.-Joo, M. S., Shin, S.-B., Kim, E. J., Koo, J. H., Yim, H., Kim, S. G. Nrf2-lncRNA controls cell fate by modulating p53-dependent Nrf2 activation as an miRNA sponge for Plk2 and p21cip1.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Carcinoma Hepatocelular/sangre , Carcinoma Hepatocelular/mortalidad , Diferenciación Celular , Medio de Cultivo Libre de Suero , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Daño del ADN , Elementos de Facilitación Genéticos , Glutatión Transferasa/genética , Hepatocitos/metabolismo , Humanos , Neoplasias Hepáticas/sangre , Neoplasias Hepáticas/mortalidad , MicroARNs/genética , MicroARNs/metabolismo , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/genética , ARN Largo no Codificante/sangre , ARN Largo no Codificante/fisiología , ARN Neoplásico/sangre , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/metabolismo
4.
J Cell Mol Med ; 22(2): 849-860, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29077264

RESUMEN

The G12 family of G protein alpha subunits has been shown to participate in the regulation of various physiological processes. However, the role of Gα12 in bone physiology has not been well described. Here, by micro-CT analysis, we discovered that Gα12-knockout mice have an osteopetrotic phenotype. Histological examination showed lower osteoclast number in femoral tissue of Gα12-knockout mice compared to wild-type mice. Additionally, in vitro osteoclastic differentiation of precursor cells with receptor activator of nuclear factor-κB ligand (RANKL) showed that Gα12 deficiency decreased the number of osteoclast generated and the bone resorption activity. The induction of nuclear factor of activated T-cell c1 (NFATc1), the key transcription factor of osteoclastogenesis, and the activation of RhoA by RANKL was also significantly suppressed by Gα12 deficiency. We further found that the RANKL induction of NFATc1 was not dependent on RhoA signalling, while osteoclast precursor migration and bone resorption required RhoA in the Gα12-mediated regulation of osteoclasts. Therefore, Gα12 plays a role in differentiation through NFATc1 and in cell migration and resorption activity through RhoA during osteoclastogenesis.


Asunto(s)
Factores de Transcripción NFATC/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Eliminación de Gen , Humanos , Macrófagos/metabolismo , Masculino , Ratones Noqueados , Osteoclastos/citología , Osteoclastos/metabolismo , Osteogénesis , Osteopetrosis/patología , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al GTP rho/metabolismo
5.
Theranostics ; 14(5): 1841-1859, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38505605

RESUMEN

Rationale: The surge of severe liver damage underscores the necessity for identifying new targets and therapeutic agents. Endoplasmic reticulum (ER) stress induces ferroptosis with Gα12 overexpression. NF-κB essential modulator (NEMO) is a regulator of inflammation and necroptosis. Nonetheless, the regulatory basis of NEMO de novo synthesis and its impact on hepatocyte ferroptosis need to be established. This study investigated whether Nrf2 transcriptionally induces IKBKG (the NEMO gene) for ferroptosis inhibition and, if so, how NEMO induction protects hepatocytes against ER stress-induced ferroptosis. Methods: Experiments were conducted using human liver tissues, hepatocytes, and injury models, incorporating NEMO overexpression and Gα12 gene modulations. RNA sequencing, immunoblotting, immunohistochemistry, reporter assays, and mutation analyses were done. Results: NEMO downregulation connects closely to ER and oxidative stress, worsening liver damage via hepatocyte ferroptosis. NEMO overexpression protects hepatocytes from ferroptosis by promoting glutathione peroxidase 4 (GPX4) expression. This protective role extends to oxidative and ER stress. Similar shifts occur in nuclear factor erythroid-2-related factor-2 (Nrf2) expression alongside NEMO changes. Nrf2 is newly identified as an IKBKG (NEMO gene) transactivator. Gα12 changes, apart from Nrf2, impact NEMO expression, pointing to post-transcriptional control. Gα12 reduction lowers miR-125a, an inhibitor of NEMO, while overexpression has the opposite effect. NEMO also counters ER stress, which triggers Gα12 overexpression. Gα12's significance in NEMO-dependent hepatocyte survival is confirmed via ROCK1 inhibition, a Gα12 downstream kinase, and miR-125a. The verified alterations or associations within the targeted entities are validated in human liver specimens and datasets originating from livers subjected to exposure to other injurious agents. Conclusions: Hepatic injury prompted by ER stress leads to the suppression of NEMO, thereby facilitating ferroptosis through the inhibition of GPX4. IKBKG is transactivated by Nrf2 against Gα12 overexpression responsible for the increase of miR-125a, an unprecedented NEMO inhibitor, resulting in GPX4 induction. Accordingly, the induction of NEMO mitigates ferroptotic liver injury.


Asunto(s)
Ferroptosis , Hepatopatías , MicroARNs , Humanos , Estrés del Retículo Endoplásmico/genética , Ferroptosis/genética , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , MicroARNs/genética , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Quinasas Asociadas a rho
6.
Hepatol Commun ; 3(10): 1373-1387, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31592043

RESUMEN

Liver X receptor-alpha (LXRα) acts as a double-edged sword in different biological situations. Given the elusive role of LXRα in hepatocyte viability, this study investigated whether LXRα protects hepatocytes from injurious stimuli and the underlying basis. LXRα activation prevented hepatocyte apoptosis from CCl4 challenges in mice. Consistently, LXRα protected hepatocytes specifically from transforming growth factor-beta (TGF-ß), whereas LXRα deficiency aggravated TGF-ß-induced hepatocyte injury. In the Gene Expression Omnibus database analysis for LXR-/- mice, TGF-ß receptors were placed in the core network. Hierarchical clustering and correlation analyses enabled us to find cannabinoid receptor 2 (CB2) as a gene relevant to LXRα. In human fibrotic liver samples, both LXRα and CB2 were lower in patients with septal fibrosis and cirrhosis than those with portal fibrosis. LXRα transcriptionally induced CB2; CB2 then defended hepatocytes from TGF-ß. In a macrophage depletion model, JWH133 (a CB2 agonist) treatment prevented toxicant-induced liver injury. MicroRNA 27b (miR-27b) was identified as an inhibitor of ubiquitin-specific peptidase 4 (USP4), deubiquitylating TGF-ß receptor 1 (TßRI), downstream from CB2. Liver-specific overexpression of LXRα protected hepatocytes from injurious stimuli and attenuated hepatic inflammation and fibrosis. Conclusion: LXRα exerts a cytoprotective effect against TGF-ß by transcriptionally regulating the CB2 gene in hepatocytes, and CB2 then inhibits USP4-stabilizing TßRI through miR-27b. Our data provide targets for the treatment of acute liver injury.

7.
EBioMedicine ; 40: 488-503, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30638865

RESUMEN

BACKGROUND: The injured liver loses normal function, with concomitant decrease of key identity genes. Super-enhancers contribute to mammalian cell identity. Here, we identified core transcription factors (TFs) that are active in hepatocytes, using genome-wide analysis and hierarchical ordering of super-enhancer distribution. METHODS: Expression of core TFs was assessed in a cohort of patients with hepatitis or cirrhosis and animal models. Quantitative PCR, chromatin immunoprecipitation assays, and hydrodynamic gene delivery methods were used to assess gene regulation and hepatocyte viability. RNA-sequencing data were generated to investigate the role of LRH1 in hepatocyte protection from injury. RESULTS: Network analysis of super-enhancer-associated gene interactions and expression arrays for cohorts of patients with hepatitis and cirrhosis enabled us to identify a super-enhancer-associated network, and LRH1, HNF4α, PPARα, and RXRα as core TFs. In mouse models, expression of core TFs was robustly inhibited by single and multiple challenge(s) with liver toxicant. RNA-seq analysis revealed changes in expression in the super-enhancer-associated genes sensitively biased toward repression by intoxication. LRH1 gene delivery prevented the loss of hepatic super-enhancer-associated signaling circuitry in toxicant-challenged mice, and protected the liver from injury, indicating the role of LRH1 in hepatocyte identity and viability. In hepatocytes, overexpression of each core TF promoted induction of other TFs. CONCLUSION: Overall, this study identified LRH1-driven pathway as a circuitry responsible for hepatocyte identity by using cistromic analysis, improving our understanding of liver pathophysiology and identifying novel therapeutic targets.


Asunto(s)
Hepatocitos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis/genética , Supervivencia Celular/genética , Biología Computacional/métodos , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Ontología de Genes , Histonas/metabolismo , Humanos , Hígado/metabolismo , Hepatopatías/genética , Hepatopatías/metabolismo , Hepatopatías/patología , Masculino , Ratones , Unión Proteica , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Receptores Citoplasmáticos y Nucleares/química
8.
J Vis Exp ; (123)2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-28570513

RESUMEN

Protein phosphorylation at specific sites determines its conformation and interaction with other molecules. Thus, protein phosphorylation affects biological functions and characteristics of the cell. Currently, the most common method for discovering phosphorylation sites is by liquid chromatography/mass spectrometry (LC/MS) analysis, a rapid and sensitive method. However, relatively labile phosphate moieties are often released from phosphopeptides during the fragmentation step, which often yields false-negative signals. In such cases, a traditional in vitro kinase assay using site-directed mutants would be more accurate, but this method is laborious and time-consuming. Therefore, an alternative method using peptide competition may be advantageous. The consensus recognition motif of 5' adenosine monophosphate-activated protein kinase (AMPK) has been established1 and was validated using a positional scanning peptide library assay2. Thus, AMPK phosphorylation sites for a novel substrate could be predicted and confirmed by the peptide competition assays. In this report, we describe the detailed steps and procedures for the in vitro oligopeptide-competing kinase assay by illustrating AMPK-mediated nuclear factor erythroid 2-related factor 2 (Nrf2) phosphorylation. To authenticate the phosphorylation site, we carried out a sequential in vitro kinase assay using a site-specific mutant. Overall, the peptide competition assay provides a method to screen multiple potential phosphorylation sites and to identify sites for validation by the phosphorylation site mutants.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Oligopéptidos/metabolismo , Proteínas Quinasas Activadas por AMP/química , Sitios de Unión , Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Biblioteca de Péptidos , Fosforilación
9.
J Clin Invest ; 127(10): 3845-3860, 2017 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-28920922

RESUMEN

Skeletal muscle is a key organ in energy homeostasis owing to its high requirement for nutrients. Heterotrimeric G proteins converge signals from cell-surface receptors to potentiate or blunt responses against environmental changes. Here, we show that muscle-specific ablation of Gα13 in mice promotes reprogramming of myofibers to the oxidative type, with resultant increases in mitochondrial biogenesis and cellular respiration. Mechanistically, Gα13 and its downstream effector RhoA suppressed nuclear factor of activated T cells 1 (NFATc1), a chief regulator of myofiber conversion, by increasing Rho-associated kinase 2-mediated (Rock2-mediated) phosphorylation at Ser243. Ser243 phosphorylation of NFATc1 was reduced after exercise, but was higher in obese animals. Consequently, Gα13 ablation in muscles enhanced whole-body energy metabolism and increased insulin sensitivity, thus affording protection from diet-induced obesity and hepatic steatosis. Our results define Gα13 as a switch regulator of myofiber reprogramming, implying that modulations of Gα13 and its downstream effectors in skeletal muscle are a potential therapeutic approach to treating metabolic diseases.


Asunto(s)
Metabolismo Energético , Hígado Graso/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Miofibrillas/metabolismo , Obesidad/metabolismo , Animales , Hígado Graso/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Ratones , Ratones Noqueados , Miofibrillas/genética , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Obesidad/genética , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
10.
Mol Cell Biol ; 36(14): 1931-42, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27161318

RESUMEN

Nrf2 (nuclear factor erythroid 2-related factor 2) is an antioxidant transcription factor. AMP-activated protein kinase (AMPK) functions as a central regulator of cell survival in response to stressful stimuli. Nrf2 should be coordinated with the cell survival pathway controlled by AMPK, but so far the mechanistic connections remain undefined. This study investigated the role of AMPK in Nrf2 trafficking and its activity regulation. A subnetwork integrating neighbor molecules suggested direct interaction between AMPK and Nrf2. In cells, AMPK activation caused nuclear accumulation of Nrf2. In the in vitro kinase and peptide competition assays, AMPK phosphorylated Nrf2 at the Ser558 residue (Ser550 in mouse) located in the canonical nuclear export signal. Nrf2 with an S550A mutation failed to be accumulated in the nucleus after AMPK activation. Leptomycin B, a nuclear export inhibitor, did not enhance nuclear accumulation of wild-type Nrf2 (WT-Nrf2) activated by AMPK or a phospho-Ser550-mimetic Nrf2 mutant, corroborating the finding that AMPK facilitated nuclear accumulation of Nrf2, probably by inhibiting nuclear export. Activated glycogen synthase kinase 3ß (GSK3ß) diminished the basal nuclear level of Myc-S550A-Nrf2. Taking the data collectively, AMPK phosphorylates Nrf2 at the Ser550 residue, which, in conjunction with AMPK-mediated GSK3ß inhibition, promotes nuclear accumulation of Nrf2 for antioxidant response element (ARE)-driven gene transactivation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Núcleo Celular/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Serina/metabolismo , Transporte Activo de Núcleo Celular , Animales , Redes Reguladoras de Genes , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Ratones , Mutación , Factor 2 Relacionado con NF-E2/química , Factor 2 Relacionado con NF-E2/genética , Fosforilación , Serina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA