Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Agric Food Chem ; 72(14): 7832-7844, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38544357

RESUMEN

Lycopene has been proven to alleviate nonalcoholic steatohepatitis (NASH), but the precise mechanisms are inadequately elucidated. In this study, we found a previously unknown regulatory effect of lycopene on the apoptosis signal-regulating kinase 1 (ASK1) signaling pathway in both in vivo and in vitro models. Lycopene supplementation (3 and 6 mg/kg/day) exhibited a significant reduction in lipid accumulation, inflammation, and fibrosis of the liver in mice fed with a high-fat/high-cholesterol diet or a methionine-choline-deficient diet. RNA sequencing uncovered that the mitogen-activated protein kinases signaling pathway, which is closely associated with inflammation and endoplasmic reticulum (ER) stress, was significantly downregulated by lycopene. Furthermore, we found lycopene ameliorated ER swelling and decreased the expression levels of ER stress markers (i.e., immunoglobulin heavy chain binding protein, C/EBP homologous protein, and X-box binding protein 1s). Especially, the inositol-requiring enzyme 1α involved in the ASK1 phosphorylation was inhibited by lycopene, resulting in the decline of the subsequent c-Jun N-terminal kinase (JNK) signaling cascade. ASK1 inhibitor DQOP-1 eliminated the lycopene-induced inhibition of the ASK1-JNK pathway in oleic acid and palmitic acid-induced HepG2 cells. Molecular docking further indicated hydrophobic interactions between lycopene and ASK1. Collectively, our research indicates that lycopene can alleviate ER stress and attenuate inflammation cascades and lipid accumulation by inhibiting the ASK1-JNK pathway.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Sistema de Señalización de MAP Quinasas/fisiología , Licopeno/metabolismo , MAP Quinasa Quinasa Quinasa 5/genética , MAP Quinasa Quinasa Quinasa 5/metabolismo , MAP Quinasa Quinasa Quinasa 5/farmacología , Simulación del Acoplamiento Molecular , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Inflamación/tratamiento farmacológico , Inflamación/genética , Estrés del Retículo Endoplásmico , Lípidos/farmacología , Apoptosis
2.
J Biochem Mol Toxicol ; 38(3): e23682, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38462752

RESUMEN

Claudin-6 (CLDN6) has been extensively studied in different tumors to date. However, in the case of nonsmall cell lung cancer (NSCLC), CLDN6 has a largely unknown role and molecular mechanism. We detected the expression of CLDN6 in NSCLC tissues and cells using reverse transcription-quantitative polymerase chain reaction (PCR) and western blot assays. A gain-of-function experiment was performed to evaluate the biological effects of CLDN6 on NSCLC cell behaviors. Methylation-specific PCR was utilized to detect the DNA methylation of CLDN6 gene promoter region. The interaction of CLDN6 and receptor interacting protein 1 (RIP1) was determined by coimmunoprecipitation assay. Furthermore, the modulation of CLDN6 on RIP1/apoptosis signal-regulating kinase 1 (ASK1)/c-Jun N-terminal kinase (JNK) axis was confirmed. The results showed that in NSCLC tissues and cells, CLDN6 expression level was declined, and was associated with a high level of DNA methylation. CLDN6 overexpression suppressed the viability, invasion, migration, and promoted cell apoptosis. Besides, the enhanced expression of CLDN6 reduced the glycolysis and the dysfunction of mitochondrial respiration of NSCLC cells. Mechanistic investigation confirmed that CLDN6 interacted with RIP1 and inhibited cellular biological function of NSCLC cells via RIP1/ASK1/JNK axis. Besides, CLDN6 overexpression inhibited tumor growth in vivo. In conclusion, CLDN6 inhibited NSCLC cell proliferation through inactivating aerobic glycolysis via the RIP1/ASK1/JNK axis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , MAP Quinasa Quinasa Quinasa 5/farmacología , Claudinas/genética , Claudinas/metabolismo , Línea Celular Tumoral , Apoptosis , Proliferación Celular
3.
Antiviral Res ; 220: 105736, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37863359

RESUMEN

Apoptosis signal-regulating kinase 1 (ASK1)/MAP3K5 is a stress response kinase that is activated by various stimuli. It is known as an upstream activator of p38- Mitogen-activated protein kinase (p38MAPK) and c-Jun N-terminal kinase (JNK) that are reactive oxygen species (ROS)-induced kinases. Accumulating evidence show that ROS accumulate in virus-infected cells. Here, we investigated the relationship between viruses and ASK1/p38MAPK or ASK1/JNK pathways. Our findings suggest that virus infection activates ASK1 related pathways. In parallel, ASK1 inhibition led to a remarkable reduction in the replication of a broad range of viruses including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), vaccinia virus (VV), vesicular stomatitis virus (VSV), Herpes Simplex Virus (HSV), and Human Immunodeficiency virus (HIV) in different human cell lines. Our work demonstrates the potential therapeutic use of Selonsertib, an ASK1 inhibitor, as a pan-antiviral drug in humans. Surprisingly, we observed differential effects of Selonsertib in in vitro and in vivo hamster models, suggesting caution in using rodent models to predict clinical and therapeutic outcomes in humans.


Asunto(s)
COVID-19 , Transducción de Señal , Humanos , ARN Viral , MAP Quinasa Quinasa Quinasa 5/metabolismo , MAP Quinasa Quinasa Quinasa 5/farmacología , Especies Reactivas de Oxígeno , Antivirales/farmacología , SARS-CoV-2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Apoptosis
4.
J Cell Biochem ; 124(3): 421-433, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36780445

RESUMEN

As one of the common and serious chronic complications of diabetes mellitus (DM), the related mechanism of diabetic retinopathy (DR) has not been fully understood. Müller cell reactive gliosis is one of the early pathophysiological features of DR. Therefore, exploring the manner to reduce diabetes-induced Müller cell damage is essential to delay DR. Thioredoxin 1 (Trx1), one of the ubiquitous redox enzymes, plays a vital role in redox homeostasis via protein-protein interactions, including apoptosis signal-regulating kinase 1 (ASK1). Previous studies have shown that upregulation of Trx by some drugs can attenuate endoplasmic reticulum stress (ERS) in DR, but the related mechanism was unclear. In this study, we used DM mouse and high glucose (HG)-cultured human Müller cells as models to clarify the effect of Trx1 on ERS and the underlying mechanism. The data showed that the diabetes-induced Müller cell damage was increased significantly. Moreover, the expression of ERS and reactive gliosis was also upregulated in diabetes in vivo and in vitro. However, it was reversed after Trx1 overexpression. Besides, ERS-related protein expression, reactive gliosis, and apoptosis were decreased after transfection with ASK1 small-interfering RNA in stable Trx1 overexpression Müller cells after HG treatment. Taken together, Trx1 could protect Müller cells from diabetes-induced damage, and the underlying mechanism was related to inhibited ERS via ASK1.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Ratones , Humanos , Animales , Células Ependimogliales/metabolismo , Gliosis , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , MAP Quinasa Quinasa Quinasa 5/genética , MAP Quinasa Quinasa Quinasa 5/farmacología , Retinopatía Diabética/genética , Apoptosis , Inflamación , Estrés del Retículo Endoplásmico
5.
Neurochem Res ; 47(12): 3829-3837, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36309631

RESUMEN

Selonsertib is a first-in-class apoptosis signal-regulating kinase 1 (ASK1) inhibitor in clinical trials for treating NASH and diabetic kidney disease due to its anti-inflammatory and anti-fibrotic activities. In the present study, we investigated the anti-neuroinflammatory effects and brain pharmacokinetic properties of selonsertib. It inhibited inflammatory cytokines and NO production by suppressing phosphorylated ASK1 in the LPS-stimulated microglial cell line, BV2 cells. Consistent with the in vitro results, selonsertib attenuated plasma and brain TNF-α levels in the LPS-induced murine neuroinflammation model. In vitro and in vivo pharmacokinetic studies of selonsertib were conducted in support of central nervous system (CNS) drug discovery. In both Caco-2 and MDR-MDCK cells, selonsertib exhibited a high efflux ratio, showing that it is a P-gp substrate. Selonsertib was rapidly and effectively absorbed into the systemic circulation after oral treatment, with a Tmax of 0.5 h and oral bioavailability of 74%. In comparison with high systemic exposure with Cmax of 16.2 µg/ml and AUC of 64 µg·h/mL following oral dosing of 10 mg/kg, the brain disposition of selonsertib was limited, with Cmax of 0.08 µg/g and Kp value of 0.004. This study demonstrates that selonsertib can be a therapeutic agent for neuroinflammatory diseases.


Asunto(s)
Lipopolisacáridos , MAP Quinasa Quinasa Quinasa 5 , Animales , Ratones , Encéfalo/metabolismo , Células CACO-2 , Lipopolisacáridos/farmacología , MAP Quinasa Quinasa Quinasa 5/metabolismo , MAP Quinasa Quinasa Quinasa 5/farmacología , Microglía/metabolismo
6.
Int J Biochem Cell Biol ; 151: 106276, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35953014

RESUMEN

Osteoporosis is a common bone disease in the elderly with high morbidity and mortality. Previous studies have shown ROS-revulsive osteoblast apoptosis to be involved in the pathogenesis of osteoporosis. At present, a research hotspot exists on the topic of the ROS-targeted clinical treatment of osteoporosis. TC-G 1008, a potent and selective GPR39 agonist, exerts a conspicuous influence on a myriad of cellular processes, ranging from cellular redox status, to gene expression, to cell apoptosis. However, the underlying mechanism by which TC-G 1008 regulates osteoblast function under oxidative stress has not yet been elucidated. The purpose of this study was to investigate the effect and underlying mechanism of TC-G 1008 in the rescue of ROS-induced apoptosis by upregulating peroxiredoxin (Prx1). In this study, experimental results demonstrated that TC-G 1008 could activate GPR39, which then accelerated ROS obliteration and apoptosis inhibition in osteoblasts via Prx1 upregulation through the nuclear factor (erythroid-derived 2)-related factor 2 (Nrf2). Interestingly, being regarded as an 'information' molecule rather than an anti-oxidase molecule, Prx1 was shown to restrict the dissociation of the apoptosis signal-regulating kinase 1 (ASK1)/thioredoxin (Trx) under oxidative stress, which signified the activation of the ASK1 pathway, thereby resulting in the suppression of apoptosis. In summary, this study explores the double mechanism of TC-G 1008 in osteoblast apoptosis amelioration under oxidative stress through (i) ROS elimination and (ii) ASK1/Trx signal suppression, both of which contribute to increased Prx1 expression, and the results suggest that TC-G 1008 has great potential in the clinical treatment of osteoporosis.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Osteoporosis , Anciano , Apoptosis , Humanos , MAP Quinasa Quinasa Quinasa 5/genética , MAP Quinasa Quinasa Quinasa 5/metabolismo , MAP Quinasa Quinasa Quinasa 5/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Osteoblastos , Osteoporosis/metabolismo , Estrés Oxidativo , Peroxirredoxinas/metabolismo , Pirimidinas , Especies Reactivas de Oxígeno/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sulfonamidas , Tiorredoxinas/metabolismo , Regulación hacia Arriba
7.
Immunopharmacol Immunotoxicol ; 44(5): 732-745, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35815528

RESUMEN

OBJECTIVES: This study aimed to explore the underlying role and mechanism of LINC00313 in osteoarthritis (OA) progression. METHODS: CHON-001 chondrocytes were treated with interleukin (IL)-1ß to induce OA in vitro, and then transfected with LINC00313 overexpression plasmids (pcDNA-LINC00313) or small interfering RNA against tumor necrosis factor (TNF) receptor-associated factor 1 (si-TRAF1). Cell viability, apoptosis, levels of inflammatory cytokines tumor necrosis factor-α (TNF-α), IL-6 and IL-8, and expression of extracellular matrix (ECM) degradation related proteins in CHON-001 cells were determined. TRAF1 promoter methylation were was detected with methylation-specific polymerase chain reaction (MSP) assay. Furthermore, a c-Jun N-terminal kinase (JNK) signaling activator was used to confirm whether the apoptosis signal-regulating kinase 1 (ASK1)/JNK signaling pathway was involved in the function of LINC00313/TRAF1 axis in chondrocytes. In addition, an OA mouse model was established and lentivirus LINC00313 overexpression vector (Lv-LINC00313) was injected, and then inflammatory cytokine levels, ECM protein expression, and pathological changes in cartilage tissues were detected. RESULTS: LINC00313 was downregulated and TRAF1 was upregulated in OA cartilage tissues. LINC00313 overexpression or TRAF1 silencing attenuated IL-1ß-induced viability inhibition, apoptosis, inflammation and ECM degradation in CHON-001 cells. Moreover, LINC00313 inhibited TRAF1 expression through promoting DNA methyltransferase 1 (DNMT1) mediated promoter methylation. TRAF1 overexpression reversed the effects of LINC00313 on IL-1ß-induced chondrocyte injury. LINC00313 overexpression inhibited the ASK1/JNK signaling pathway, and JNK activator reversed the effect. In addition, Lv-LINC00313 treatment alleviated cartilage tissue damage and cartilage matrix degradation in OA mice. CONCLUSIONS: LINC00313 alleviated OA progression through inhibiting TRAF1 expression and the ASK1/JNK signaling pathway.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Osteoartritis , Animales , Apoptosis , ADN/metabolismo , ADN/farmacología , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Interleucina-8/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , MAP Quinasa Quinasa Quinasa 5/farmacología , Metilación , Metiltransferasas/metabolismo , Metiltransferasas/farmacología , Ratones , Osteoartritis/genética , Osteoartritis/metabolismo , ARN Interferente Pequeño , Factor 1 Asociado a Receptor de TNF/genética , Factor de Necrosis Tumoral alfa/metabolismo
8.
Sci Rep ; 11(1): 22009, 2021 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-34759307

RESUMEN

Recent studies have shown that adipose tissue is an immunological organ. While inflammation in energy-storing white adipose tissues has been the focus of intense research, the regulatory mechanisms of inflammation in heat-producing brown adipose tissues remain largely unknown. We previously identified apoptosis signal-regulating kinase 1 (ASK1) as a critical regulator of brown adipocyte maturation; the PKA-ASK1-p38 axis facilitates uncoupling protein 1 (UCP1) induction cell-autonomously. Here, we show that ASK1 suppresses an innate immune pathway and contributes to maintenance of brown adipocytes. We report a novel chemical pull-down method for endogenous kinases using analog sensitive kinase allele (ASKA) technology and identify an ASK1 interactor in brown adipocytes, receptor-interacting serine/threonine-protein kinase 2 (RIPK2). ASK1 disrupts the RIPK2 signaling complex and inhibits the NOD-RIPK2 pathway to downregulate the production of inflammatory cytokines. As a potential biological significance, an in vitro model for intercellular regulation suggests that ASK1 facilitates the expression of UCP1 through the suppression of inflammatory cytokine production. In parallel to our previous report on the PKA-ASK1-p38 axis, our work raises the possibility of an auxiliary role of ASK1 in brown adipocyte maintenance through neutralizing the thermogenesis-suppressive effect of the NOD-RIPK2 pathway.


Asunto(s)
Adipocitos Marrones/metabolismo , MAP Quinasa Quinasa Quinasa 5/farmacología , Proteínas Adaptadoras de Señalización NOD/efectos de los fármacos , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/efectos de los fármacos , Adipocitos Marrones/efectos de los fármacos , Adipocitos Blancos/metabolismo , Animales , Citocinas/análisis , Células HEK293 , Humanos , Inflamación/tratamiento farmacológico , Ratones , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Desacopladora 1/efectos de los fármacos
9.
Br J Cancer ; 103(9): 1380-90, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20877355

RESUMEN

BACKGROUND: Sustained p38(MAPK) phosphorylation upregulates p75 neurotrophin (p75(NTR)) and induces apoptosis in Ewing's sarcoma family of tumours (ESFT). As fenretinide induces ESFT death through sustained p38(MAPK) phosphorylation, we hypothesised that this may be effected through upregulation of death receptors (DRs) and that treatment of fenretinide plus DR ligands may enhance apoptosis. METHODS: DR expression was determined by flow cytometry. Trypan blue exclusion assays, caspase-8 flow cytometry and immunoblotting for Bid were used to measure cell death. RESULTS: Fenretinide upregulated cell surface expression of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors, FAS and p75(NTR), in an ASK1- and p38α-dependent manner. Cotreatment with fenretinide and DR ligands resulted in synergistic death compared with either agent alone; caspase-8 and Bid were cleaved in a time-dependent manner. Fenretinide did not increase DR expression in non-malignant cells. Furthermore, fenretinide, TRAIL or a combination of both agents was non-cytotoxic to non-malignant cells. Etoposide and actinomycin D increased expression of all DRs examined, whereas vincristine increased FAS alone. Only actinomycin D and TRAIL, and etoposide with TRAIL or FasL, enhanced death compared with either agent alone. CONCLUSION: The synergistic death observed with fenretinide and DR ligands suggests that this combination may be an attractive strategy for the treatment of ESFT.


Asunto(s)
Fenretinida/farmacología , MAP Quinasa Quinasa Quinasa 5/farmacología , Receptores de Muerte Celular/metabolismo , Sarcoma de Ewing/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Fosforilación , Sarcoma de Ewing/patología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Free Radic Biol Med ; 43(1): 39-47, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17561092

RESUMEN

Several recent studies have demonstrated that thioredoxin (Trx) is an important antiapoptotic/cytoprotective molecule. The present study was designed to determine whether Trx activity is altered in the aging heart in a way that may contribute to increased susceptibility to myocardial ischemia/reperfusion (MI/R). Compared to young animals, MI/R-induced cardiomyocyte apoptosis and infarct size were increased in aging animals (p<0.01). Trx activity was decreased in the aging heart before MI/R, and this difference was further amplified after MI/R. Trx expression was moderately increased and Trx nitration, a posttranslational modification that inhibits Trx activity, was increased in the aging heart. Moreover, Trx-aptosis-regulating kinase-1 (Trx-ASK1) complex formation was reduced and activity of p38 mitogen-activated protein kinase (MAPK) was increased. Treatment with FP15 (a peroxynitrite decomposition catalyst) reduced Trx nitration, increased Trx activity, restored Trx-ASK1 interaction, reduced P38 MAPK activity, attenuated caspase 3 activation, and reduced infarct size in aging animals (p<0.01). Our results demonstrated that Trx activity is decreased in the aging heart by posttranslational nitrative modification. Interventions that restore Trx activity in the aging heart may be novel therapies to attenuate MI/R injury in aging patients.


Asunto(s)
Envejecimiento/metabolismo , Daño por Reperfusión Miocárdica/etiología , Miocardio/metabolismo , Especies de Nitrógeno Reactivo/metabolismo , Tiorredoxinas/antagonistas & inhibidores , Animales , MAP Quinasa Quinasa Quinasa 5/farmacología , Masculino , Ratones , Ratones Endogámicos , Daño por Reperfusión Miocárdica/metabolismo , Ácido Peroxinitroso/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Especies de Nitrógeno Reactivo/antagonistas & inhibidores , Tiorredoxinas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Arterioscler Thromb Vasc Biol ; 27(8): 1857-63, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17561491

RESUMEN

OBJECTIVE: Oxidative stress contributes to the pathogenesis of many diseases, including atherosclerosis and sepsis. We have previously described a novel class of therapeutic compounds with antioxidant and antiinflammatory properties. However, at present, the intracellular targets of these compounds have not been identified. The purpose of this study was to elucidate the mechanism by which 2 structurally-related antioxidants (AGI-1067 and AGI-1095) inhibit LPS induction of tissue factor (TF) expression in human monocytic cells and endothelial cells. METHODS AND RESULTS: We found that succinobucol (AGI-1067) and AGI-1095 inhibited LPS induction of TF expression in both monocytic cells and endothelial cells. These compounds also reduced LPS induction of nuclear AP-1 and expression of Egr-1 without affecting nuclear translocation of NF-kappaB. Importantly, these antioxidants inhibited LPS activation of the redox-sensitive kinase, apoptosis signal-regulating kinase-1 (ASK1) and the mitogen-activated protein kinases (MAPKs) p38, ERK1/2, and JNK1/2. CONCLUSIONS: AGI-1067 and AGI-1095 inhibit TF gene expression in both monocytic cells and endothelial cells through a mechanism that involves the inhibition of the redox-sensitive MAP3K, ASK1. These compounds selectively reduce the activation/induction of MAPK, AP-1, and Egr-1 without affecting NF-kappaB nuclear translocation.


Asunto(s)
Antioxidantes/farmacología , Lipopolisacáridos/farmacología , MAP Quinasa Quinasa Quinasa 5/farmacología , Proteína Quinasa 1 Activada por Mitógenos/farmacología , Tromboplastina/metabolismo , Northern Blotting , Western Blotting , Comunicación Celular , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Activación Enzimática , Regulación de la Expresión Génica , Humanos , Monocitos/efectos de los fármacos , Monocitos/fisiología , Estrés Oxidativo , Probabilidad , ARN Mensajero/análisis , Sensibilidad y Especificidad , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...