Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Exp Mol Med ; 55(8): 1720-1733, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37524868

RESUMEN

Autophagy functions in cellular quality control and metabolic regulation. Dysregulation of autophagy is one of the major pathogenic factors contributing to the progression of nonalcoholic fatty liver disease (NAFLD). Autophagy is involved in the breakdown of intracellular lipids and the maintenance of healthy mitochondria in NAFLD. However, the mechanisms underlying autophagy dysregulation in NAFLD remain unclear. Here, we demonstrate that the hepatic expression level of Thrap3 was significantly increased in NAFLD conditions. Liver-specific Thrap3 knockout improved lipid accumulation and metabolic properties in a high-fat diet (HFD)-induced NAFLD model. Furthermore, Thrap3 deficiency enhanced autophagy and mitochondrial function. Interestingly, Thrap3 knockout increased the cytosolic translocation of AMPK from the nucleus and enhanced its activation through physical interaction. The translocation of AMPK was regulated by direct binding with AMPK and the C-terminal domain of Thrap3. Our results indicate a role for Thrap3 in NAFLD progression and suggest that Thrap3 is a potential target for NAFLD treatment.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia/genética , Dieta Alta en Grasa/efectos adversos , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Factores de Transcripción/metabolismo , Humanos , Células Hep G2
2.
Elife ; 102021 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-34964438

RESUMEN

Background: Non-alcoholic fatty liver disease (NAFLD) is characterized by excessive lipid accumulation and imbalances in lipid metabolism in the liver. Although nuclear receptors (NRs) play a crucial role in hepatic lipid metabolism, the underlying mechanisms of NR regulation in NAFLD remain largely unclear. Methods: Using network analysis and RNA-seq to determine the correlation between NRs and microRNA in human NAFLD patients, we revealed that MIR20B specifically targets PPARA. MIR20B mimic and anti-MIR20B were administered to human HepG2 and Huh-7 cells and mouse primary hepatocytes as well as high-fat diet (HFD)- or methionine-deficient diet (MCD)-fed mice to verify the specific function of MIR20B in NAFLD. We tested the inhibition of the therapeutic effect of a PPARα agonist, fenofibrate, by Mir20b and the synergic effect of combination of fenofibrate with anti-Mir20b in NAFLD mouse model. Results: We revealed that MIR20B specifically targets PPARA through miRNA regulatory network analysis of nuclear receptor genes in NAFLD. The expression of MIR20B was upregulated in free fatty acid (FA)-treated hepatocytes and the livers of both obesity-induced mice and NAFLD patients. Overexpression of MIR20B significantly increased hepatic lipid accumulation and triglyceride levels. Furthermore, MIR20B significantly reduced FA oxidation and mitochondrial biogenesis by targeting PPARA. In Mir20b-introduced mice, the effect of fenofibrate to ameliorate hepatic steatosis was significantly suppressed. Finally, inhibition of Mir20b significantly increased FA oxidation and uptake, resulting in improved insulin sensitivity and a decrease in NAFLD progression. Moreover, combination of fenofibrate and anti-Mir20b exhibited the synergic effect on improvement of NAFLD in MCD-fed mice. Conclusions: Taken together, our results demonstrate that the novel MIR20B targets PPARA, plays a significant role in hepatic lipid metabolism, and present an opportunity for the development of novel therapeutics for NAFLD. Funding: This research was funded by Korea Mouse Phenotyping Project (2016M3A9D5A01952411), the National Research Foundation of Korea (NRF) grant funded by the Korea government (2020R1F1A1061267, 2018R1A5A1024340, NRF-2021R1I1A2041463, 2020R1I1A1A01074940, 2016M3C9A394589324), and the Future-leading Project Research Fund (1.210034.01) of UNIST.


Asunto(s)
Fenofibrato/farmacología , Hipolipemiantes/farmacología , Metabolismo de los Lípidos , MicroARNs/genética , Enfermedad del Hígado Graso no Alcohólico/genética , PPAR alfa/genética , Animales , Femenino , Humanos , Masculino , Ratones , MicroARNs/metabolismo , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , PPAR alfa/metabolismo
3.
J Ethnopharmacol ; 278: 114238, 2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-34048878

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Vernicia fordii (Hemsl.) Airy Shaw (V. fordii) is also known as the tung tree and its leaves and fruit are used as an oriental treatment for dyspepsia, edema, and skin diseases, which are known as diabetic complications. AIM OF THE STUDY: In this study, we aimed to investigate the methanolic extract (VF5) of the leaves of V. fordii as an insulin secretagogue and its probable mechanism and verify the effect in HFD-fed mice. MATERIALS AND METHODS: The insulin secretagogue activity of different doses of VF5 (0.1, 0.3 and 1.0 µg/ml) was assessed using in vitro insulin secretion assay and confirmed the anti-diabetic effect in mice fed HFD for 4 weeks with different doses of VF5 (10, 20 and 50 mg/kg oral) for another 6 weeks. Glbenclamide (30 mg/kg, oral) was used as positive control drug. The possible mechanisms were evaluated by using Gö6983 (10 µM), U73122 (10 µM) and nifedipine (10 µM). The major constituents of VF5 were analyzed by UPLC-QToF-MS and 1H and 13C NMR spectroscopy. RESULTS: UPLC-QToF-MS and NMR spectroscopy analysis indicated that one of the main active components of VF5 was tigliane-diterpene esters. VF5 functioned as an insulin secretagogue and enhanced mitochondria respiration and insulin homeostasis. We confirmed that VF5 preserved the ß-cell and reduced the ß-cell expansion which caused by metabolic stress under HFD. The antidiabetic role of VF5 in HFD fed mice was assessed by glucose tolerance test (GTT) and insulin tolerance test (ITT), fasting plasma insulin level, fasting blood glucose level, AKT signal in peripheral tissue in the absence of toxic effects. Mechanistically, insulinotropic effect of VF5 was mediated by activation of PKCα via intracellular Ca2+ influx and enhanced mitochondria function. CONCLUSION: VF5 exhibits potent insulin secretagogue function and improves insulin sensitivity and protection of pancreatic ß-cells from metabolic stress without toxicity. Taken together, our study suggests that VF5 could be potentially used for treating diabetes and metabolic diseases through improving ß-cell function.


Asunto(s)
Aleurites/química , Diabetes Mellitus Experimental/tratamiento farmacológico , Secreción de Insulina/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Diabetes Mellitus Experimental/fisiopatología , Relación Dosis-Respuesta a Droga , Prueba de Tolerancia a la Glucosa , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/aislamiento & purificación , Hipoglucemiantes/farmacología , Resistencia a la Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Extractos Vegetales/administración & dosificación , Extractos Vegetales/efectos adversos , Estrés Fisiológico/efectos de los fármacos
4.
Exp Mol Med ; 52(6): 940-950, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32504039

RESUMEN

The endoplasmic reticulum (ER) stress response is an adaptive mechanism that is activated upon disruption of ER homeostasis and protects the cells against certain harmful environmental stimuli. However, critical and prolonged cell stress triggers cell death. In this study, we demonstrate that Flightless-1 (FliI) regulates ER stress-induced apoptosis in colon cancer cells by modulating Ca2+ homeostasis. FliI was highly expressed in both colon cell lines and colorectal cancer mouse models. In a mouse xenograft model using CT26 mouse colorectal cancer cells, tumor formation was slowed due to elevated levels of apoptosis in FliI-knockdown (FliI-KD) cells. FliI-KD cells treated with ER stress inducers, thapsigargin (TG), and tunicamycin exhibited activation of the unfolded protein response (UPR) and induction of UPR-related gene expression, which eventually triggered apoptosis. FliI-KD increased the intracellular Ca2+ concentration, and this upregulation was caused by accelerated ER-to-cytosolic efflux of Ca2+. The increase in intracellular Ca2+ concentration was significantly blocked by dantrolene and tetracaine, inhibitors of ryanodine receptors (RyRs). Dantrolene inhibited TG-induced ER stress and decreased the rate of apoptosis in FliI-KD CT26 cells. Finally, we found that knockdown of FliI decreased the levels of sorcin and ER Ca2+ and that TG-induced ER stress was recovered by overexpression of sorcin in FliI-KD cells. Taken together, these results suggest that FliI regulates sorcin expression, which modulates Ca2+ homeostasis in the ER through RyRs. Our findings reveal a novel mechanism by which FliI influences Ca2+ homeostasis and cell survival during ER stress.


Asunto(s)
Calcio/metabolismo , Neoplasias Colorrectales/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Proteínas de Microfilamentos/metabolismo , Transactivadores/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Línea Celular Tumoral , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Neoplasias Colorrectales/genética , Estrés del Retículo Endoplásmico/genética , Humanos , Immunoblotting , Masculino , Ratones , Proteínas de Microfilamentos/genética , Transactivadores/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nutrients ; 12(3)2020 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-32183397

RESUMEN

: The chronic low-grade inflammation in adipose tissue plays a causal role in obesity-induced insulin resistance and its associated pathophysiological consequences. In this study, we investigated the effects of extracts of Broussonetia papyrifera root bark (PRE) and its bioactive components on inflammation and insulin sensitivity. PRE inhibited TNF-α-induced NF-κB transcriptional activity in the NF-κB luciferase assay and pro-inflammatory genes' expression by blocking phosphorylation of IκB and NF-κB in 3T3-L1 adipocytes, which were mediated by activating AMPK. Ten-week-high fat diet (HFD)-fed C57BL6 male mice treated with PRE had improved glucose intolerance and decreased inflammation in adipose tissue, as indicated by reductions in NF-κB phosphorylation and pro-inflammatory genes' expression. Furthermore, PRE activated AMP-activated protein kinase (AMPK) and reduced lipogenic genes' expression in both adipose tissue and liver. Finally, we identified broussoflavonol B (BF) and kazinol J (KJ) as bioactive constituents to suppress pro-inflammatory responses via activating AMPK in 3T3-L1 adipocytes. Taken together, these results indicate the therapeutic potential of PRE, especially BF or KJ, in metabolic diseases such as obesity and type 2 diabetes.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Tejido Adiposo/metabolismo , Antiinflamatorios , Broussonetia/química , Resistencia a la Insulina , Corteza de la Planta/química , Extractos Vegetales , Raíces de Plantas/química , Transducción de Señal/efectos de los fármacos , Células 3T3-L1 , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Activación Enzimática/efectos de los fármacos , Células HEK293 , Humanos , Masculino , Ratones , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Obesidad/patología , Extractos Vegetales/química , Extractos Vegetales/farmacología , Células RAW 264.7
6.
Cells ; 9(2)2020 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-32024237

RESUMEN

Peroxisome proliferator-activated receptor γ (PPARγ) is a master regulator of adipose tissue biology. In obesity, phosphorylation of PPARγ at Ser273 (pSer273) by cyclin-dependent kinase 5 (CDK5)/extracellular signal-regulated kinase (ERK) orchestrates diabetic gene reprogramming via dysregulation of specific gene expression. Although many recent studies have focused on the development of non-classical agonist drugs that inhibit the phosphorylation of PPARγ at Ser273, the molecular mechanism of PPARγ dephosphorylation at Ser273 is not well characterized. Here, we report that protein phosphatase Mg2+/Mn2+-dependent 1A (PPM1A) is a novel PPARγ phosphatase that directly dephosphorylates Ser273 and restores diabetic gene expression which is dysregulated by pSer273. The expression of PPM1A significantly decreases in two models of insulin resistance: diet-induced obese (DIO) mice and db/db mice, in which it negatively correlates with pSer273. Transcriptomic analysis using microarray and genotype-tissue expression (GTEx) data in humans shows positive correlations between PPM1A and most of the genes that are dysregulated by pSer273. These findings suggest that PPM1A dephosphorylates PPARγ at Ser273 and represents a potential target for the treatment of obesity-linked metabolic disorders.


Asunto(s)
Diabetes Mellitus/genética , PPAR gamma/metabolismo , Proteína Fosfatasa 2C/metabolismo , Serina/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Regulación de la Expresión Génica , Células HEK293 , Humanos , Resistencia a la Insulina/genética , Ratones , Obesidad/genética , Fosforilación , Unión Proteica , Proteína Fosfatasa 2C/genética
7.
Exp Mol Med ; 50(10): 1-11, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30323259

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-dependent transcription factor that regulates adipocyte differentiation and glucose homeostasis. The transcriptional activity of PPARγ is regulated not only by ligands but also by post-translational modifications (PTMs). In this study, we demonstrate that a novel E3 ligase of PPARγ, tripartite motif-containing 25 (TRIM25), directly induced the ubiquitination of PPARγ, leading to its proteasome-dependent degradation. During adipocyte differentiation, both TRIM25 mRNA and protein expression significantly decreased and negatively correlated with the expression of PPARγ. The stable expression of TRIM25 reduced PPARγ protein levels and suppressed adipocyte differentiation in 3T3-L1 cells. In contrast, the specific knockdown of TRIM25 increased PPARγ protein levels and stimulated adipocyte differentiation. Furthermore, TRIM25-knockout mouse embryonic fibroblasts (MEFs) exhibited an increased adipocyte differentiation capability compared with wild-type MEFs. Taken together, these data indicate that TRIM25 is a novel E3 ubiquitin ligase of PPARγ and that TRIM25 is a novel target for PPARγ-associated metabolic diseases.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Diferenciación Celular , PPAR gamma/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética , Células 3T3-L1 , Animales , Diferenciación Celular/genética , Células HEK293 , Humanos , Ratones , Unión Proteica , Estabilidad Proteica , Proteolisis , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
8.
Diabetes ; 65(4): 829-39, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26740599

RESUMEN

Blocking phosphorylation of peroxisome proliferator-activated receptor (PPAR)γ at Ser(273) is one of the key mechanisms for antidiabetes drugs to target PPARγ. Using high-throughput phosphorylation screening, we here describe that Gleevec blocks cyclin-dependent kinase 5-mediated PPARγ phosphorylation devoid of classical agonism as a PPARγ antagonist ligand. In high fat-fed mice, Gleevec improved insulin sensitivity without causing severe side effects associated with other PPARγ-targeting drugs. Furthermore, Gleevec reduces lipogenic and gluconeogenic gene expression in liver and ameliorates inflammation in adipose tissues. Interestingly, Gleevec increases browning of white adipose tissue and energy expenditure. Taken together, the results indicate that Gleevec exhibits greater beneficial effects on both glucose/lipid metabolism and energy homeostasis by blocking PPARγ phosphorylation. These data illustrate that Gleevec could be a novel therapeutic agent for use in insulin resistance and type 2 diabetes.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Mesilato de Imatinib/farmacología , Resistencia a la Insulina , PPAR gamma/antagonistas & inhibidores , Células 3T3-L1 , Tejido Adiposo Pardo/fisiología , Tejido Adiposo Blanco/fisiología , Animales , Células Cultivadas , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
9.
Chem Sci ; 7(8): 5523-5529, 2016 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-30034693

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-regulated transcription factor that plays crucial roles in adipogenesis, lipid metabolism, and glucose homeostasis. Several PPARγ ligands possess anti-diabetic activity and they commonly inhibit the phosphorylation of PPARγ at serine 273 (Ser273). The recently reported PPARγ ligand SR1664, which selectively blocks the phosphorylation of PPARγ without classical agonism, has potent anti-diabetic activity, indicating that the inhibition of Ser273 phosphorylation is sufficient to provoke anti-diabetic effects. In this study, we revealed the X-ray structure of PPARγ co-crystallized with SR1664 bound to the alternate binding site of PPARγ and confirmed that the alternate site binding of SR1664 blocks the phosphorylation of Ser273. Furthermore, using covalent inhibitors as chemical tools, we demonstrated that the inhibition of phosphorylation is attributed to the occupation of a specific site which is a hydrophobic region between helix 3 and ß3-ß4 at the binding pocket of PPARγ. In high-fat diet-induced obese mice, we confirmed the anti-diabetic activity of our covalent inhibitor SB1453 that was designed to bind at the specific site in PPARγ for blocking the phosphorylation of Ser273. Lastly, the target selectivity of SB1453 was demonstrated by fluorescence-based visualization of target proteins complexed with the covalent probe 11 containing a bioorthogonal functional group.

10.
Diabetologia ; 58(12): 2867-76, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26342595

RESUMEN

AIMS/HYPOTHESIS: O-GlcNAcylation plays a role as a metabolic sensor regulating cellular signalling, transcription and metabolism. Transcription factors and signalling pathways related to metabolism are modulated by N-acetyl-glucosamine (O-GlcNAc) modification. Aberrant regulation of O-GlcNAcylation is closely linked to insulin resistance, type 2 diabetes and obesity. Current evidence shows that increased O-GlcNAcylation negatively regulates insulin signalling, which is associated with insulin resistance and type 2 diabetes. Here, we aimed to evaluate the effects of Oga (also known as Mgea5) haploinsufficiency, which causes hyper-O-GlcNAcylation, on metabolism. METHODS: We examined whether Oga(+/-) mice developed insulin resistance. Metabolic variables were determined including body weight, glucose and insulin tolerance, metabolic rate and thermogenesis. RESULTS: Oga deficiency does not affect insulin signalling even at hyper-O-GlcNAc levels. Oga(+/-) mice are lean with reduced fat mass and improved glucose tolerance. Furthermore, Oga(+/-) mice resist high-fat diet-induced obesity with ameliorated hepatic steatosis and improved glucose metabolism. Oga haploinsufficiency potentiates energy expenditure through the enhancement of brown adipocyte differentiation from the stromal vascular fraction of subcutaneous white adipose tissue (WAT). CONCLUSIONS/INTERPRETATION: Our observations suggest that O-GlcNAcase (OGA) is essential for energy metabolism via regulation of the thermogenic WAT program.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Metabolismo Energético/genética , Obesidad/genética , beta-N-Acetilhexosaminidasas/genética , Acetilglucosamina/metabolismo , Adipocitos Marrones/metabolismo , Adipocitos Marrones/patología , Animales , Glucemia/metabolismo , Peso Corporal/genética , Diferenciación Celular , Diabetes Mellitus Tipo 2/genética , Intolerancia a la Glucosa/genética , Resistencia a la Insulina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Termogénesis/genética
11.
Cell Signal ; 27(3): 614-20, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25479590

RESUMEN

Peroxisome proliferator-activated receptor γ (PPARγ) is a member of the nuclear receptor family and plays key roles in glucose and lipid metabolism. Its transcriptional control of target genes is mediated by ligand-dependent recruitment of coactivators. In this study, we demonstrate that a novel transcriptional modulator of PPARγ, Flightless-I (FLII) binds directly to and suppresses the transcriptional activity of PPARγ. The LXXLL motif within the leucine-rich repeat (LRR) domain of FLII interacts directly with the DNA-binding domain of PPARγ. Interestingly, in the presence of PPARγ ligands, such as rosiglitazone and SR1664, this interaction was abolished in vitro. When FLII was overexpressed, both the transcriptional activity of PPARγ and adipogenesis were suppressed significantly, whereas specific knockdown of FLII reversed these effects. Furthermore, DNA occupancy of PPARγ on its target gene promoters was enhanced by FLII knockdown, and the interaction between PPARγ and retinoid X receptor α (RXRα) was blocked by FLII. Together, these findings strongly suggest that FLII functions in PPARγ activation as a molecular switch to repress transcriptional activity by interrupting formation of the PPARγ/RXRα complex, and FLII may serve as a novel therapeutic target in the treatment of adiposity-related metabolic syndromes.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , PPAR gamma/metabolismo , Receptor alfa X Retinoide/metabolismo , Células 3T3-L1 , Adipogénesis/efectos de los fármacos , Secuencias de Aminoácidos , Animales , Proteínas Portadoras , Proteínas del Citoesqueleto/antagonistas & inhibidores , Proteínas del Citoesqueleto/genética , Dimerización , Células HEK293 , Humanos , Ratones , Proteínas de Microfilamentos , PPAR gamma/genética , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Receptor alfa X Retinoide/genética , Rosiglitazona , Tiazolidinedionas/farmacología , Transactivadores , Transcripción Genética/efectos de los fármacos
12.
Genes Dev ; 28(21): 2361-9, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25316675

RESUMEN

Phosphorylation of peroxisome proliferator-activated receptor γ (PPARγ) at Ser273 by cyclin-dependent kinase 5 (CDK5) in adipose tissue stimulates insulin resistance, but the underlying molecular mechanisms are unclear. We show here that Thrap3 (thyroid hormone receptor-associated protein 3) can directly interact with PPARγ when it is phosphorylated at Ser273, and this interaction controls the diabetic gene programming mediated by the phosphorylation of PPARγ. Knockdown of Thrap3 restores most of the genes dysregulated by CDK5 action on PPARγ in cultured adipocytes. Importantly, reduced expression of Thrap3 in fat tissue by antisense oligonucleotides (ASOs) regulates a specific set of genes, including the key adipokines adiponectin and adipsin, and effectively improves hyperglycemia and insulin resistance in high-fat-fed mice without affecting body weight. These data indicate that Thrap3 plays a crucial role in controlling diabetic gene programming and may provide opportunities for the development of new therapeutics for obesity and type 2 diabetes.


Asunto(s)
Proteínas de Unión al ADN/genética , Diabetes Mellitus Tipo 2/genética , PPAR gamma/metabolismo , Factores de Transcripción/genética , Células 3T3 , Adipoquinas/genética , Animales , Células Cultivadas , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Fosfoserina/metabolismo , Unión Proteica
13.
J Biol Chem ; 289(38): 26618-26629, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25100724

RESUMEN

Thiazolidinedione class of anti-diabetic drugs which are known as peroxisome proliferator-activated receptor γ (PPARγ) ligands have been used to treat metabolic disorders, but thiazolidinediones can also cause several severe side effects, including congestive heart failure, fluid retention, and weight gain. In this study, we describe a novel synthetic PPARγ ligand UNIST HYUNDAI Compound 1 (UHC1) that binds tightly to PPARγ without the classical agonism and which blocks cyclin-dependent kinase 5 (CDK5)-mediated PPARγ phosphorylation. We modified the non-agonist PPARγ ligand SR1664 chemically to improve its solubility and then developed a novel PPARγ ligand, UHC1. According to our docking simulation, UHC1 occupied the ligand-binding site of PPARγ with a higher docking score than SR1664. In addition, UHC1 more potently blocked CDK5-mediated PPARγ phosphorylation at Ser-273. Surprisingly, UHC1 treatment effectively ameliorated the inflammatory response both in vitro and in high-fat diet-fed mice. Furthermore, UHC1 treatment dramatically improved insulin sensitivity in high-fat diet-fed mice without causing fluid retention and weight gain. Taken together, compared with SR1664, UHC1 exhibited greater beneficial effects on glucose and lipid metabolism by blocking CDK5-mediated PPARγ phosphorylation, and these data indicate that UHC1 could be a novel therapeutic agent for use in type 2 diabetes and related metabolic disorders.


Asunto(s)
Benzoatos/farmacología , Quinasa 5 Dependiente de la Ciclina/fisiología , Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/farmacología , Indoles/farmacología , PPAR gamma/metabolismo , Células 3T3-L1 , Adipogénesis/efectos de los fármacos , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Benzoatos/química , Sitios de Unión , Compuestos de Bifenilo/química , Compuestos de Bifenilo/farmacología , Evaluación Preclínica de Medicamentos , Células HEK293 , Humanos , Enlace de Hidrógeno , Hipoglucemiantes/química , Indoles/química , Mediadores de Inflamación/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , PPAR gamma/agonistas , PPAR gamma/química , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Ratas Sprague-Dawley
14.
BMB Rep ; 47(11): 599-608, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25154720

RESUMEN

As the prevalence of obesity has increased explosively over the last several decades, associated metabolic disorders, including type 2 diabetes, dyslipidemia, hypertension, and cardiovascular diseases, have been also increased. Thus, new strategies for preventing and treating them are needed. The nuclear peroxisome proliferator-activated receptors (PPARs) are involved fundamentally in regulating energy homeostasis; thus, they have been considered attractive drug targets for addressing metabolic disorders. Among the PPARs, PPARγ is a master regulator of gene expression for metabolism, inflammation, and other pathways in many cell types, especially adipocytes. It is a physiological receptor of the potent anti-diabetic drugs of the thiazolidinediones (TZDs) class, including rosiglitazone (Avandia). However, TZDs have undesirable and severe side effects, such as weight gain, fluid retention, and cardiovascular dysfunction. Recently, many reports have suggested that PPARγ could be modulated by post-translational modifications (PTMs), and modulation of PTM has been considered as novel approaches for treating metabolic disorders with fewer side effects than the TZDs. In this review, we discuss how PTM of PPARγ may be regulated and issues to be considered in making novel anti-diabetic drugs that can modulate the PTM of PPARγ.


Asunto(s)
Enfermedades Metabólicas/tratamiento farmacológico , PPAR gamma/agonistas , Tiazolidinedionas/uso terapéutico , Fármacos Antidiuréticos/uso terapéutico , Metabolismo Energético , Humanos , Resistencia a la Insulina , Enfermedades Metabólicas/metabolismo , Obesidad/tratamiento farmacológico , PPAR gamma/metabolismo , Procesamiento Proteico-Postraduccional , Tiazolidinedionas/efectos adversos
15.
Zebrafish ; 11(2): 122-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24588364

RESUMEN

Peroxisome proliferator-activated receptor γ (PPARγ) is a target of antidiabetic drugs. However, many PPARγ activators, including rosiglitazone, show unwanted side effects, such as weight gain. The KR-62980 [1-(trans-methylimino-N-oxy)-3-phenyl-6-(3-phenylpropoxy)-1H-indene-2-carboxylic acid ethyl ester], a novel partial agonist of PPARγ, is a new compound for diabetes with antihyperglycemic activity and weak antiadipogenic activity. This study was performed to elucidate the mechanism of the weak adipogenesis induced by KR-62980 despite its being a PPARγ agonist in zebrafish. We elucidated the mechanism of KR-62980 in lipid metabolism using adipocytes and zebrafish. Since NADPH is a critical cofactor in fat metabolism, we investigated effect of KR-62980 on NADPH-producing enzymes such as cytosolic NADP(+) isocitrate dehydrogenase (cICDH). We found that the mRNA expression of cICDH was significantly decreased by KR-62980 in 3T3-L1 cells. KR-62980 inhibited lipase activity and lipid metabolism in zebrafish. Further, KR-62980 substantially suppressed cICDH in adipocytes and zebrafish. These results suggest that cICDH may be one of the targets of KR-62980 responsible for weight gain and adipogenesis.


Asunto(s)
Expresión Génica/efectos de los fármacos , Indenos/farmacología , Isocitrato Deshidrogenasa/genética , Metabolismo de los Lípidos/efectos de los fármacos , Morfolinas/farmacología , Pez Cebra/genética , Pez Cebra/metabolismo , Células 3T3-L1 , Animales , Línea Celular , Isocitrato Deshidrogenasa/metabolismo , Ratones , PPAR gamma/agonistas , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Mensajero/metabolismo
16.
J Toxicol Sci ; 38(5): 697-702, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24025786

RESUMEN

Sodium dodecyl sulfate (SDS) and sodium dodecyl benzenesulfonate (SDBS) are widely used anionic surfactants in household, industrial, and institutional cleaners. Although there are many reports of their toxic effects, few studies have focused on the pharmacological properties of these surfactants. Peroxisome proliferator-activated receptor (PPAR) γ is a transcriptional factor belonging to the nuclear receptor superfamily. The ligands of PPARγ regulate its transcriptional activity and modulate many biological functions, including adipocyte differentiation and lipid metabolism. In this study, we investigated the ligand activities of SDS and SDBS for nuclear receptors using time-resolved fluorescence resonance energy transfer-based coactivator recruitment assays. SDS and SDBS showed selective ligand activities for PPARγ and these ligand activities were eliminated by a PPARγ antagonist. SDS and SDBS also promoted adipocyte differentiation accompanied by upregulation of adipocyte-specific gene expression in 3T3-L1 preadipocytes. These findings reveal novel actions of anionic alkyl surfactants as PPARγ ligands.


Asunto(s)
Bencenosulfonatos/farmacología , PPAR gamma/fisiología , Dodecil Sulfato de Sodio/farmacología , Tensoactivos/farmacología , Células 3T3/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Anilidas/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Regulación de la Expresión Génica , Ligandos , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , PPAR gamma/agonistas , PPAR gamma/antagonistas & inhibidores , Regulación hacia Arriba
17.
J Nutr Biochem ; 24(1): 156-62, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22898571

RESUMEN

Nobiletin (NOB) is a polymethoxylated flavone present in citrus fruits and has been reported to have antitumor and anti-inflammatory effects. However, little is known about the effects of NOB on obesity and insulin resistance. In this study, we examined the effects of NOB on obesity and insulin resistance, and the underlying mechanisms, in high-fat diet (HFD)-induced obese mice. Obese mice were fed a HFD for 8 weeks and then treated without (HFD control group) or with NOB at 10 or 100mg/kg. NOB decreased body weight gain, white adipose tissue (WAT) weight and plasma triglyceride. Plasma glucose levels tended to decrease compared with the HFD group and improved plasma adiponectin levels and glucose tolerance. Furthermore, NOB altered the expression levels of several lipid metabolism-related and adipokine genes. NOB increased the mRNA expression of peroxisome proliferator-activated receptor (PPAR)-γ, sterol regulatory element-binding protein-1c, fatty acid synthase, stearoyl-CoA desaturase-1, PPAR-α, carnitine palmitoyltransferase-1, uncoupling protein-2 and adiponectin, and decreased the mRNA expression of tumor necrosis factor-α and monocyte chemoattractant protein-1 in WAT. NOB also up-regulated glucose transporter-4 protein expression and Akt phosphorylation and suppressed IκBα degradation in WAT. Taken together, these results suggest that NOB improves adiposity, dyslipidemia, hyperglycemia and insulin resistance. These effects may be elicited by regulating the expression of lipid metabolism-related and adipokine genes, and by regulating the expression of inflammatory makers and activity of the insulin signaling pathway.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Flavonas/farmacología , Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Obesidad/tratamiento farmacológico , Adipoquinas/genética , Adiponectina/genética , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Ingestión de Alimentos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Insulina/metabolismo , Metabolismo de los Lípidos/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Tamaño de los Órganos/efectos de los fármacos , PPAR gamma/genética , Triglicéridos/sangre , Triglicéridos/metabolismo , Aumento de Peso/efectos de los fármacos
18.
J Nat Med ; 67(2): 320-6, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22791412

RESUMEN

Flos Magnoliae (FM) is a commonly used Chinese medicinal herb for symptomatic relief of allergic rhinitis, sinusitis and headache. Although several FM species have been used as substitutes or adulterants for clinical use, possible differences in their pharmacological actions have not been reported. To confirm the effects of FM on skeletal muscle glucose metabolism, we tested the effects of several compounds isolated from FM on glucose uptake by L6 myotubes. We found that fargesin, a component of FM, dose-dependently stimulated glucose consumption in L6 myotubes, which was accompanied by enhanced glucose transporter (GLUT)-4 translocation to the cell surface. Fargesin-stimulated glucose uptake was blocked by wortmannin, a phosphatidylinositol-3 kinase (PI3 K) inhibitor. Fargesin stimulated Akt phosphorylation, a key component in the insulin signaling pathway, which was completely inhibited by wortmannin. Here, we demonstrated that fargesin, a bioactive component of Flos Magnoliae, increases basal glucose uptake and GLUT4 translocation in L6 myotubes by activating the PI3 K-Akt pathway.


Asunto(s)
Benzodioxoles/farmacología , Transporte Biológico/efectos de los fármacos , Glucosa/metabolismo , Lignanos/farmacología , Magnolia/química , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Animales , Transportador de Glucosa de Tipo 4/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
19.
Biofactors ; 38(4): 300-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22674784

RESUMEN

This study examined the effects of fargesin, a neolignan isolated from Magnolia plants, on obesity and insulin resistance and the possible mechanisms involved in these effects in 3T3-L1 adipocytes and high-fat diet (HFD)-induced obese mice. Fargesin promoted the glucose uptake in 3T3-L1 adipocytes. In HFD-induced obese mice, fargesin decreased the body weight gain, white adipose tissue (WAT), and plasma triglyceride, non-esterified fatty acid and glucose levels, and improved the glucose tolerance. Fargesin increased glucose transporter 4 (GLUT4) protein expression and phosphorylation of Akt, AMP-activated protein kinase (AMPK), and acetyl-CoA carboxylase (ACC) in both 3T3-L1 adipocytes and WAT of HFD-induced obese mice. Fargesin also decreased the mRNA expression levels of fatty acid oxidation-related genes, such as peroxisome proliferator-activated receptor α (PPARα), carnitine palmitoyltransferase-1 (CPT-1), uncoupling protein-2 (UCP-2) and leptin in WAT. Taken together, the present findings suggest that fargesin improves dyslipidemia and hyperglycemia by activating Akt and AMPK in WAT.


Asunto(s)
Benzodioxoles/farmacología , Dieta Alta en Grasa/efectos adversos , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Hipolipemiantes/farmacología , Lignanos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Obesidad/metabolismo , Células 3T3-L1 , Acetil-CoA Carboxilasa/metabolismo , Adenilato Quinasa/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Benzodioxoles/uso terapéutico , Glucemia , Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 4/metabolismo , Hipoglucemiantes/uso terapéutico , Hipolipemiantes/uso terapéutico , Lignanos/uso terapéutico , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/tratamiento farmacológico , Obesidad/etiología , Tamaño de los Órganos/efectos de los fármacos , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , Aumento de Peso/efectos de los fármacos
20.
Biofactors ; 38(5): 372-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22674833

RESUMEN

Honokiol and magnolol, ingredients of Magnolia officinalis, which is used in traditional Chinese and Japanese medicines, have been reported to have antioxidant, anticancer, and antiangiogenic effects. Effects of these compounds on glucose metabolism in adipocytes have also been reported. However, their effects on skeletal muscle glucose uptake and the underlying molecular mechanisms are still unknown. Here, we investigated the direct effects and signaling pathways activated by honokiol and magnolol in skeletal muscle cells using L6 myotubes. We found that honokiol and magnolol dose-dependently acutely stimulated glucose uptake without synergistic effects of combined administration in L6 myotubes. Treatment with honokiol and magnolol also stimulated glucose transporter-4 translocation to the cell surface. Honokiol- and magnolol-stimulated glucose uptake was blocked by the phosphatidylinositol-3 kinase inhibitor, wortmannin. Both honokiol and magnolol stimulated Akt phosphorylation, a key element in the insulin signaling pathway, which was completely inhibited by wortmannin. These results suggest that honokiol and magnolol might have beneficial effects on glucose metabolism by activating the insulin signaling pathway.


Asunto(s)
Compuestos de Bifenilo/farmacología , Glucosa/metabolismo , Lignanos/farmacología , Fibras Musculares Esqueléticas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Androstadienos/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/metabolismo , Magnolia/química , Fibras Musculares Esqueléticas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Plantas Medicinales/química , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/agonistas , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Transducción de Señal/efectos de los fármacos , Wortmanina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...