Skip Navigation
Skip to contents

Endocrinol Metab : Endocrinology and Metabolism

clarivate
OPEN ACCESS
SEARCH
Search

Search

Page Path
HOME > Search
8 "Jinmi Lee"
Filter
Filter
Article type
Keywords
Publication year
Authors
Funded articles
Original Articles
Diabetes, obesity and metabolism
Docosahexanoic Acid Attenuates Palmitate-Induced Apoptosis by Autophagy Upregulation via GPR120/mTOR Axis in Insulin-Secreting Cells
Seok-Woo Hong, Jinmi Lee, Sun Joon Moon, Hyemi Kwon, Se Eun Park, Eun-Jung Rhee, Won-Young Lee
Endocrinol Metab. 2024;39(2):353-363.   Published online January 23, 2024
DOI: https://doi.org/10.3803/EnM.2023.1809
  • 901 View
  • 40 Download
AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
Polyunsaturated fatty acids (PUFAs) reportedly have protective effects on pancreatic β-cells; however, the underlying mechanisms are unknown.
Methods
To investigate the cellular mechanism of PUFA-induced cell protection, mouse insulinoma 6 (MIN6) cells were cultured with palmitic acid (PA) and/or docosahexaenoic acid (DHA), and alterations in cellular signaling and apoptosis were examined.
Results
DHA treatment remarkably repressed caspase-3 cleavage and terminal deoxynucleotidyl transferase-mediated UTP nick end labeling (TUNEL)-positive red dot signals in PA-treated MIN6 cells, with upregulation of autophagy, an increase in microtubule- associated protein 1-light chain 3 (LC3)-II, autophagy-related 5 (Atg5), and decreased p62. Upstream factors involved in autophagy regulation (Beclin-1, unc51 like autophagy activating kinase 1 [ULK1], phosphorylated mammalian target of rapamycin [mTOR], and protein kinase B) were also altered by DHA treatment. DHA specifically induced phosphorylation on S2448 in mTOR; however, phosphorylation on S2481 decreased. The role of G protein-coupled receptor 120 (GPR120) in the effect of DHA was demonstrated using a GPR120 agonist and antagonist. Additional treatment with AH7614, a GPR120 antagonist, significantly attenuated DHA-induced autophagy and protection. Taken together, DHA-induced autophagy activation with protection against PA-induced apoptosis mediated by the GPR120/mTOR axis.
Conclusion
These findings indicate that DHA has therapeutic effects on PA-induced pancreatic β-cells, and that the cellular mechanism of β-cell protection by DHA may be a new research target with potential pharmacotherapeutic implications in β-cell protection.
Close layer
Diabetes, obesity and metabolism
Inhibition of Sodium-Glucose Cotransporter-2 during Serum Deprivation Increases Hepatic Gluconeogenesis via the AMPK/AKT/FOXO Signaling Pathway
Jinmi Lee, Seok-Woo Hong, Min-Jeong Kim, Yu-Mi Lim, Sun Joon Moon, Hyemi Kwon, Se Eun Park, Eun-Jung Rhee, Won-Young Lee
Endocrinol Metab. 2024;39(1):98-108.   Published online January 3, 2024
DOI: https://doi.org/10.3803/EnM.2023.1786
  • 1,401 View
  • 80 Download
AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
Sodium-dependent glucose cotransporter 2 (SGLT2) mediates glucose reabsorption in the renal proximal tubules, and SGLT2 inhibitors are used as therapeutic agents for treating type 2 diabetes mellitus. This study aimed to elucidate the effects and mechanisms of SGLT2 inhibition on hepatic glucose metabolism in both serum deprivation and serum supplementation states.
Methods
Huh7 cells were treated with the SGLT2 inhibitors empagliflozin and dapagliflozin to examine the effect of SGLT2 on hepatic glucose uptake. To examine the modulation of glucose metabolism by SGLT2 inhibition under serum deprivation and serum supplementation conditions, HepG2 cells were transfected with SGLT2 small interfering RNA (siRNA), cultured in serum-free Dulbecco’s modified Eagle’s medium for 16 hours, and then cultured in media supplemented with or without 10% fetal bovine serum for 8 hours.
Results
SGLT2 inhibitors dose-dependently decreased hepatic glucose uptake. Serum deprivation increased the expression levels of the gluconeogenesis genes peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1α), glucose 6-phosphatase (G6pase), and phosphoenolpyruvate carboxykinase (PEPCK), and their expression levels during serum deprivation were further increased in cells transfected with SGLT2 siRNA. SGLT2 inhibition by siRNA during serum deprivation induces nuclear localization of the transcription factor forkhead box class O 1 (FOXO1), decreases nuclear phosphorylated-AKT (p-AKT), and p-FOXO1 protein expression, and increases phosphorylated-adenosine monophosphate-activated protein kinase (p-AMPK) protein expression. However, treatment with the AMPK inhibitor, compound C, reversed the reduction in the protein expression levels of nuclear p- AKT and p-FOXO1 and decreased the protein expression levels of p-AMPK and PEPCK in cells transfected with SGLT2 siRNA during serum deprivation.
Conclusion
These data show that SGLT2 mediates glucose uptake in hepatocytes and that SGLT2 inhibition during serum deprivation increases gluconeogenesis via the AMPK/AKT/FOXO1 signaling pathway.
Close layer
Diabetes, Obesity and Metabolism
Dulaglutide Ameliorates Palmitic Acid-Induced Hepatic Steatosis by Activating FAM3A Signaling Pathway
Jinmi Lee, Seok-Woo Hong, Min-Jeong Kim, Sun Joon Moon, Hyemi Kwon, Se Eun Park, Eun-Jung Rhee, Won-Young Lee
Endocrinol Metab. 2022;37(1):74-83.   Published online February 9, 2022
DOI: https://doi.org/10.3803/EnM.2021.1293
  • 4,884 View
  • 235 Download
  • 5 Web of Science
  • 5 Crossref
AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
Dulaglutide, a long-acting glucagon-like peptide-1 receptor agonist (GLP-1RA), has been shown to reduce body weight and liver fat content in patients with type 2 diabetes. Family with sequence similarity 3 member A (FAM3A) plays a vital role in regulating glucose and lipid metabolism. The aim of this study was to determine the mechanisms by which dulaglutide protects against hepatic steatosis in HepG2 cells treated with palmitic acid (PA).
Methods
HepG2 cells were pretreated with 400 μM PA for 24 hours, followed by treatment with or without 100 nM dulaglutide for 24 hours. Hepatic lipid accumulation was determined using Oil red O staining and triglyceride (TG) assay, and the expression of lipid metabolism-associated factor was analyzed using quantitative real time polymerase chain reaction and Western blotting.
Results
Dulaglutide significantly decreased hepatic lipid accumulation and reduced the expression of genes associated with lipid droplet binding proteins, de novo lipogenesis, and TG synthesis in PA-treated HepG2 cells. Dulaglutide also increased the expression of proteins associated with lipolysis and fatty acid oxidation and FAM3A in PA-treated cells. However, exendin-(9-39), a GLP-1R antagonist, reversed the expression of FAM3A, and fatty acid oxidation-associated factors increased due to dulaglutide. In addition, inhibition of FAM3A by siRNA attenuated the reducing effect of dulaglutide on TG content and its increasing effect on regulation of fatty acid oxidation.
Conclusion
These results suggest that dulaglutide could be used therapeutically for improving nonalcoholic fatty liver disease, and its effect could be mediated in part via upregulation of FAM3A expression through a GLP-1R-dependent pathway.

Citations

Citations to this article as recorded by  
  • GLP-1/GLP-1RAs: New Options for the Drug Treatment of NAFLD
    Haoran Jiang, Linquan Zang
    Current Pharmaceutical Design.2024; 30(2): 100.     CrossRef
  • GLP-1 Receptor Agonists in Non-Alcoholic Fatty Liver Disease: Current Evidence and Future Perspectives
    Riccardo Nevola, Raffaella Epifani, Simona Imbriani, Giovanni Tortorella, Concetta Aprea, Raffaele Galiero, Luca Rinaldi, Raffaele Marfella, Ferdinando Carlo Sasso
    International Journal of Molecular Sciences.2023; 24(2): 1703.     CrossRef
  • FAM3A mediates the phenotypic switch of human aortic smooth muscle cells stimulated with oxidised low-density lipoprotein by influencing the PI3K-AKT pathway
    Lei Yang, Baoshun Du, Shitao Zhang, Maode Wang
    In Vitro Cellular & Developmental Biology - Animal.2023; 59(6): 431.     CrossRef
  • ATP Secretion and Metabolism in Regulating Pancreatic Beta Cell Functions and Hepatic Glycolipid Metabolism
    Jing Li, Han Yan, Rui Xiang, Weili Yang, Jingjing Ye, Ruili Yin, Jichun Yang, Yujing Chi
    Frontiers in Physiology.2022;[Epub]     CrossRef
  • Targeted therapeutics and novel signaling pathways in non-alcohol-associated fatty liver/steatohepatitis (NAFL/NASH)
    Xiaohan Xu, Kyle L. Poulsen, Lijuan Wu, Shan Liu, Tatsunori Miyata, Qiaoling Song, Qingda Wei, Chenyang Zhao, Chunhua Lin, Jinbo Yang
    Signal Transduction and Targeted Therapy.2022;[Epub]     CrossRef
Close layer
Endocrine Research
Clusterin Protects Lipotoxicity-Induced Apoptosis via Upregulation of Autophagy in Insulin-Secreting Cells
Seok-Woo Hong, Jinmi Lee, Min Jeong Kim, Sun Joon Moon, Hyemi Kwon, Se Eun Park, Eun-Jung Rhee, Won-Young Lee
Endocrinol Metab. 2020;35(4):943-953.   Published online December 2, 2020
DOI: https://doi.org/10.3803/EnM.2020.768
  • 5,662 View
  • 135 Download
  • 4 Web of Science
  • 6 Crossref
AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
There is a great need to discover factors that could protect pancreatic β-cells from apoptosis and thus prevent diabetes mellitus. Clusterin (CLU), a chaperone protein, plays an important role in cell protection in numerous cells and is involved in various cellular mechanisms, including autophagy. In the present study, we investigated the protective role of CLU through autophagy regulation in pancreatic β-cells.
Methods
To identify the protective role of CLU, mouse insulinoma 6 (MIN6) cells were incubated with CLU and/or free fatty acid (FFA) palmitate, and cellular apoptosis and autophagy were examined.
Results
Treatment with CLU remarkably upregulated microtubule-associated protein 1-light chain 3 (LC3)-II conversion in a doseand time-dependent manner with a significant increase in the autophagy-related 3 (Atg3) gene expression level, which is a mediator of LC3-II conversion. Moreover, co-immunoprecipitation and fluorescence microscopy experiments showed that the molecular interaction of LC3 with Atg3 and p62 was markedly increased by CLU. Stimulation of LC3-II conversion by CLU persisted in lipotoxic conditions, and FFA-induced apoptosis and dysfunction were simultaneously improved by CLU treatment. Finally, inhibition of LC3-II conversion by Atg3 gene knockdown markedly attenuated the cytoprotective effect of CLU.
Conclusion
Taken together, these findings suggest that CLU protects pancreatic β-cells against lipotoxicity-induced apoptosis via autophagy stimulation mediated by facilitating LC3-II conversion. Thus, CLU has therapeutic effects on FFA-induced pancreatic β-cell dysfunction.

Citations

Citations to this article as recorded by  
  • Exercise as a non-pharmacological intervention to protect pancreatic beta cells in individuals with type 1 and type 2 diabetes
    Alexandra Coomans de Brachène, Corentin Scoubeau, Anyïshai E. Musuaya, Jose Maria Costa-Junior, Angela Castela, Julie Carpentier, Vitalie Faoro, Malgorzata Klass, Miriam Cnop, Decio L. Eizirik
    Diabetologia.2023; 66(3): 450.     CrossRef
  • Apolipoprotein J Attenuates Vascular Restenosis by Promoting Autophagy and Inhibiting the Proliferation and Migration of Vascular Smooth Muscle Cells
    Ning Yang, Bo Dong, Yanqiu Song, Yang Li, Lu Kou, Qin Qin
    Journal of Cardiovascular Translational Research.2022; 15(5): 1086.     CrossRef
  • Targets for rescue from fatty acid-induced lipotoxicity in pancreatic beta cells
    Seok-Woo Hong, Won-Young Lee
    Cardiovascular Prevention and Pharmacotherapy.2022; 4(2): 57.     CrossRef
  • Co-regulators of autophagy and the cell cycle in HFD − As treated mice
    Marzieh Zeinvand-Lorestani, Mohammad Javad Khodayar, Ali Teimoori, Najmaldin Saki, Akram Ahangarpour, Ali Ranjbar, Hamed Zeinvand-Lorestani
    Journal of Trace Elements and Minerals.2022; 2: 100018.     CrossRef
  • Targeting pancreatic β cells for diabetes treatment
    Chirag Jain, Ansarullah, Sara Bilekova, Heiko Lickert
    Nature Metabolism.2022; 4(9): 1097.     CrossRef
  • Mechanisms of Beta-Cell Apoptosis in Type 2 Diabetes-Prone Situations and Potential Protection by GLP-1-Based Therapies
    Safia Costes, Gyslaine Bertrand, Magalie A. Ravier
    International Journal of Molecular Sciences.2021; 22(10): 5303.     CrossRef
Close layer
Endocrine Research
Deficiency of Sphingosine-1-Phosphate Reduces the Expression of Prohibitin and Causes β-Cell Impairment via Mitochondrial Dysregulation
Seok-Woo Hong, Jinmi Lee, Hyemi Kwon, Se Eun Park, Eun-Jung Rhee, Cheol-Young Park, Ki-Won Oh, Sung-Woo Park, Won-Young Lee
Endocrinol Metab. 2018;33(3):403-412.   Published online September 18, 2018
DOI: https://doi.org/10.3803/EnM.2018.33.3.403
  • 4,204 View
  • 50 Download
  • 16 Web of Science
  • 16 Crossref
AbstractAbstract PDFPubReader   ePub   
Background

Emerging evidence suggests that sphingolipids may be involved in type 2 diabetes. However, the exact signaling defect through which disordered sphingolipid metabolism induces β-cell dysfunction remains unknown. The current study demonstrated that sphingosine-1-phosphate (S1P), the product of sphingosine kinase (SphK), is an essential factor for maintaining β-cell function and survival via regulation of mitochondrial action, as mediated by prohibitin (PHB).

Methods

We examined β-cell function and viability, as measured by mitochondrial function, in mouse insulinoma 6 (MIN6) cells in response to manipulation of cellular S1P and PHB levels.

Results

Lack of S1P induced by sphingosine kinase inhibitor (SphKi) treatment caused β-cell dysfunction and apoptosis, with repression of mitochondrial function shown by decreases in cellular adenosine triphosphate content, the oxygen consumption rate, the expression of oxidative phosphorylation complexes, the mitochondrial membrane potential, and the expression of key regulators of mitochondrial dynamics (mitochondrial dynamin-like GTPase [OPA1] and mitofusin 1 [MFN1]). Supplementation of S1P led to the recovery of mitochondrial function and greatly improved β-cell function and viability. Knockdown of SphK2 using small interfering RNA induced mitochondrial dysfunction, decreased glucose-stimulated insulin secretion (GSIS), and reduced the expression of PHB, an essential regulator of mitochondrial metabolism. PHB deficiency significantly reduced GSIS and induced mitochondrial dysfunction, and co-treatment with S1P did not reverse these trends.

Conclusion

Altogether, these data suggest that S1P is an essential factor in the maintenance of β-cell function and survival through its regulation of mitochondrial action and PHB expression.

Citations

Citations to this article as recorded by  
  • Mitochondrial Cristae Morphology Reflecting Metabolism, Superoxide Formation, Redox Homeostasis, and Pathology
    Petr Ježek, Martin Jabůrek, Blanka Holendová, Hana Engstová, Andrea Dlasková
    Antioxidants & Redox Signaling.2023; 39(10-12): 635.     CrossRef
  • Sphingolipids in mitochondria—from function to disease
    Maryam Jamil, Lauren Ashley Cowart
    Frontiers in Cell and Developmental Biology.2023;[Epub]     CrossRef
  • Sphingosine‐1‐phosphate in mitochondrial function and metabolic diseases
    Meng Duan, Pan Gao, Sheng‐xi Chen, Petr Novák, Kai Yin, Xiao Zhu
    Obesity Reviews.2022;[Epub]     CrossRef
  • Involvement of miR‐27a‐3p in diabetic nephropathy via affecting renal fibrosis, mitochondrial dysfunction, and endoplasmic reticulum stress
    Lina Wu, Qingzhu Wang, Feng Guo, Xiaojun Ma, Jiao Wang, Yanyan Zhao, Yushan Yan, Guijun Qin
    Journal of Cellular Physiology.2021; 236(2): 1454.     CrossRef
  • Sphingosine‐1‐phosphate in acute exercise and training
    Katarzyna Hodun, Adrian Chabowski, Marcin Baranowski
    Scandinavian Journal of Medicine & Science in Sports.2021; 31(5): 945.     CrossRef
  • The Ethyl Acetate Extract From Celastrus orbiculatus Promotes Apoptosis of Gastric Cancer Cells Through Mitochondria Regulation by PHB
    Lide Tao, Zixin Yin, Tengyang Ni, Zewen Chu, Shihua Hao, Zeyu Wang, Masataka Sunagawa, Haibo Wang, Yanqing Liu
    Frontiers in Pharmacology.2021;[Epub]     CrossRef
  • Sphingosine 1-phosphate Stimulates Insulin Secretion and Improves Cell Survival by Blocking Voltage-dependent K+ Channels in β Cells
    Zhihong Liu, Huanhuan Yang, Linping Zhi, Huan Xue, Zhihong Lu, Yanli Zhao, Lijuan Cui, Tao Liu, Shouan Ren, Peifeng He, Yunfeng Liu, Yi Zhang
    Frontiers in Pharmacology.2021;[Epub]     CrossRef
  • Sphingosine-1 Phosphate Lyase Regulates Sensitivity of Pancreatic Beta-Cells to Lipotoxicity
    Yadi Tang, Thomas Plötz, Markus H. Gräler, Ewa Gurgul-Convey
    International Journal of Molecular Sciences.2021; 22(19): 10893.     CrossRef
  • Sphingolipids and Mitochondrial Dynamic
    Lais Brigliadori Fugio, Fernanda B. Coeli-Lacchini, Andréia Machado Leopoldino
    Cells.2020; 9(3): 581.     CrossRef
  • Diminished Sphingolipid Metabolism, a Hallmark of Future Type 2 Diabetes Pathogenesis, Is Linked to Pancreatic β Cell Dysfunction
    Saifur R. Khan, Yousef Manialawy, Andreea Obersterescu, Brian J. Cox, Erica P. Gunderson, Michael B. Wheeler
    iScience.2020; 23(10): 101566.     CrossRef
  • Neuronal Metabolism and Neuroprotection: Neuroprotective Effect of Fingolimod on Menadione-Induced Mitochondrial Damage
    Antonio Gil, Elisa Martín-Montañez, Nadia Valverde, Estrella Lara, Federica Boraldi, Silvia Claros, Silvana-Yanina Romero-Zerbo, Oscar Fernández, Jose Pavia, Maria Garcia-Fernandez
    Cells.2020; 10(1): 34.     CrossRef
  • WITHDRAWN: Ceramide and Sphingosine 1-Phosphate in adipose dysfunction
    Zijian Fang, Susan Pyne, Nigel J. Pyne
    Progress in Lipid Research.2019; : 100991.     CrossRef
  • Dynamic of mitochondrial network, cristae, and mitochondrial nucleoids in pancreatic β-cells
    Petr Ježek, Andrea Dlasková
    Mitochondrion.2019; 49: 245.     CrossRef
  • Sphingosine kinase 1 overexpression induces MFN2 fragmentation and alters mitochondrial matrix Ca2+ handling in HeLa cells
    I. Pulli, C. Löf, T. Blom, M.Y. Asghar, T. Lassila, N. Bäck, K.-L. Lin, J.H. Nyström, K. Kemppainen, D.M. Toivola, E. Dufour, A. Sanz, H.M. Cooper, J.B. Parys, K. Törnquist
    Biochimica et Biophysica Acta (BBA) - Molecular Cell Research.2019; 1866(9): 1475.     CrossRef
  • Ceramide and sphingosine 1-phosphate in adipose dysfunction
    Zijian Fang, Susan Pyne, Nigel J. Pyne
    Progress in Lipid Research.2019; 74: 145.     CrossRef
  • S1P/S1P Receptor Signaling in Neuromuscolar Disorders
    Elisabetta Meacci, Mercedes Garcia-Gil
    International Journal of Molecular Sciences.2019; 20(24): 6364.     CrossRef
Close layer
Diabetes
Pioglitazone Attenuates Palmitate-Induced Inflammation and Endoplasmic Reticulum Stress in Pancreatic β-Cells
Seok-Woo Hong, Jinmi Lee, Jung Hwan Cho, Hyemi Kwon, Se Eun Park, Eun-Jung Rhee, Cheol-Young Park, Ki-Won Oh, Sung-Woo Park, Won-Young Lee
Endocrinol Metab. 2018;33(1):105-113.   Published online March 21, 2018
DOI: https://doi.org/10.3803/EnM.2018.33.1.105
  • 6,245 View
  • 96 Download
  • 19 Web of Science
  • 23 Crossref
AbstractAbstract PDFPubReader   ePub   
Background

The nuclear receptor peroxisome proliferator-activator gamma (PPARγ) is a useful therapeutic target for obesity and diabetes, but its role in protecting β-cell function and viability is unclear.

Methods

To identify the potential functions of PPARγ in β-cells, we treated mouse insulinoma 6 (MIN6) cells with the PPARγ agonist pioglitazone in conditions of lipotoxicity, endoplasmic reticulum (ER) stress, and inflammation.

Results

Palmitate-treated cells incubated with pioglitazone exhibited significant improvements in glucose-stimulated insulin secretion and the repression of apoptosis, as shown by decreased caspase-3 cleavage and poly (adenosine diphosphate [ADP]-ribose) polymerase activity. Pioglitazone also reversed the palmitate-induced expression of inflammatory cytokines (tumor necrosis factor α, interleukin 6 [IL-6], and IL-1β) and ER stress markers (phosphor-eukaryotic translation initiation factor 2α, glucose-regulated protein 78 [GRP78], cleaved-activating transcription factor 6 [ATF6], and C/EBP homologous protein [CHOP]), and pioglitazone significantly attenuated inflammation and ER stress in lipopolysaccharide- or tunicamycin-treated MIN6 cells. The protective effect of pioglitazone was also tested in pancreatic islets from high-fat-fed KK-Ay mice administered 0.02% (wt/wt) pioglitazone or vehicle for 6 weeks. Pioglitazone remarkably reduced the expression of ATF6α, GRP78, and monocyte chemoattractant protein-1, prevented α-cell infiltration into the pancreatic islets, and upregulated glucose transporter 2 (Glut2) expression in β-cells. Moreover, the preservation of β-cells by pioglitazone was accompanied by a significant reduction of blood glucose levels.

Conclusion

Altogether, these results support the proposal that PPARγ agonists not only suppress insulin resistance, but also prevent β-cell impairment via protection against ER stress and inflammation. The activation of PPARγ might be a new therapeutic approach for improving β-cell survival and insulin secretion in patients with diabetes mellitus

Citations

Citations to this article as recorded by  
  • Nr1h4 and Thrb ameliorate ER stress and provide protection in the MPTP mouse model of Parkinson’s
    Nancy Ahuja, Shalini Gupta, Rashmi Arora, Ella Bhagyaraj, Drishti Tiwari, Sumit Kumar, Pawan Gupta
    Life Science Alliance.2024; 7(7): e202302416.     CrossRef
  • Prosthetic vascular grafts engineered to combat calcification: Progress and future directions
    Taylor K. Brown, Sara Alharbi, Karen J. Ho, Bin Jiang
    Biotechnology and Bioengineering.2023; 120(4): 953.     CrossRef
  • Obesity, diabetes mellitus, and cardiometabolic risk: An Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) 2023
    Harold Edward Bays, Shagun Bindlish, Tiffany Lowe Clayton
    Obesity Pillars.2023; 5: 100056.     CrossRef
  • Metformin promotes osteogenic differentiation and prevents hyperglycaemia-induced osteoporosis by suppressing PPARγ expression
    Lifeng Zheng, Ximei Shen, Yun Xie, Hong Lian, Sunjie Yan, Shizhong Wang
    Acta Biochimica et Biophysica Sinica.2023; 55(3): 394.     CrossRef
  • Peroxisome proliferator-activated receptors as targets to treat metabolic diseases: Focus on the adipose tissue, liver, and pancreas
    Henrique Souza-Tavares, Carolline Santos Miranda, Isabela Macedo Lopes Vasques-Monteiro, Cristian Sandoval, Daiana Araujo Santana-Oliveira, Flavia Maria Silva-Veiga, Aline Fernandes-da-Silva, Vanessa Souza-Mello
    World Journal of Gastroenterology.2023; 29(26): 4136.     CrossRef
  • Nicotinamide N-methyltransferase upregulation contributes to palmitate-elicited peroxisome proliferator-activated receptor transactivation in hepatocytes
    Qing Song, Jun Wang, Alexandra Griffiths, Samuel Man Lee, Iredia D. Iyamu, Rong Huang, Jose Cordoba-Chacon, Zhenyuan Song
    American Journal of Physiology-Cell Physiology.2023; 325(1): C29.     CrossRef
  • The global perspective on peroxisome proliferator-activated receptor γ (PPARγ) in ectopic fat deposition: A review
    Yanhao Qiu, Mailin Gan, Xingyu Wang, Tianci Liao, Qiuyang Chen, Yuhang Lei, Lei Chen, Jinyong Wang, Ye Zhao, Lili Niu, Yan Wang, Shunhua Zhang, Li Zhu, Linyuan Shen
    International Journal of Biological Macromolecules.2023; 253: 127042.     CrossRef
  • Chemical inducer of regucalcin attenuates lipopolysaccharide‐induced inflammatory responses in pancreatic MIN6 β‐cells and RAW264.7 macrophages
    Tomiyasu Murata, Kazunori Hashimoto, Susumu Kohno, Chiaki Takahashi, Masayoshi Yamaguchi, Chihiro Ito, Itoigawa Masataka, Roji Kojima, Kiyomi Hikita, Norio Kaneda
    FEBS Open Bio.2022; 12(1): 175.     CrossRef
  • Targets for rescue from fatty acid-induced lipotoxicity in pancreatic beta cells
    Seok-Woo Hong, Won-Young Lee
    Cardiovascular Prevention and Pharmacotherapy.2022; 4(2): 57.     CrossRef
  • Analysis of changes in the proteomic profile of porcine corpus luteum during different stages of the oestrous cycle: effects of PPAR gamma ligands
    Zuzanna Kunicka, Karol Mierzejewski, Aleksandra Kurzyńska, Robert Stryiński, Jesús Mateos, Mónica Carrera, Monika Golubska, Iwona Bogacka, Xiaolong Wang
    Reproduction, Fertility and Development.2022; 34(11): 776.     CrossRef
  • Activation of PPARγ Protects Obese Mice from Acute Lung Injury by Inhibiting Endoplasmic Reticulum Stress and Promoting Mitochondrial Biogenesis
    Yin Tang, Ke Wei, Ling Liu, Jingyue Ma, Siqi Wu, Wenjing Tang, Stéphane Mandard
    PPAR Research.2022; 2022: 1.     CrossRef
  • Effect of Pioglitazone on endoplasmic reticulum stress regarding in situ perfusion rat model
    Vivien Telek, Luca Erlitz, Ibitamuno Caleb, Tibor Nagy, Mónika Vecsernyés, Bálint Balogh, György Sétáló, Péter Hardi, Gábor Jancsó, Ildikó Takács
    Clinical Hemorheology and Microcirculation.2021; 79(2): 311.     CrossRef
  • Inflammation in Metabolic Diseases and Insulin Resistance
    Won-Young Lee
    Cardiovascular Prevention and Pharmacotherapy.2021; 3(2): 31.     CrossRef
  • Current Status of Endoplasmic Reticulum Stress in Type II Diabetes
    Sagir Mustapha, Mustapha Mohammed, Ahmad Khusairi Azemi, Abubakar Ibrahim Jatau, Aishatu Shehu, Lukman Mustapha, Ibrahim Muazzamu Aliyu, Rabi’u Nuhu Danraka, Abdulbasit Amin, Auwal Adam Bala, Wan Amir Nizam Wan Ahmad, Aida Hanum Ghulam Rasool, Mohd Rais M
    Molecules.2021; 26(14): 4362.     CrossRef
  • JunD Regulates Pancreatic β-Cells Function by Altering Lipid Accumulation
    Kexin Wang, Yixin Cui, Peng Lin, Zhina Yao, Yu Sun
    Frontiers in Endocrinology.2021;[Epub]     CrossRef
  • Pioglitazone even at low dosage improves NAFLD in type 2 diabetes: clinical and pathophysiological insights from a subgroup of the TOSCA.IT randomised trial
    Giuseppe Della Pepa, Marco Russo, Marilena Vitale, Fabrizia Carli, Claudia Vetrani, Maria Masulli, Gabriele Riccardi, Olga Vaccaro, Amalia Gastaldelli, Angela A. Rivellese, Lutgarda Bozzetto
    Diabetes Research and Clinical Practice.2021; 178: 108984.     CrossRef
  • Radioprotective Effect of Pioglitazone Against Genotoxicity Induced by Ionizing Radiation in Healthy Human Lymphocytes
    Roya Kazemi, Seyed J. Hosseinimehr
    Cardiovascular & Hematological Agents in Medicinal Chemistry .2021; 19(1): 72.     CrossRef
  • Recent Insights Into Mechanisms of β-Cell Lipo- and Glucolipotoxicity in Type 2 Diabetes
    Maria Lytrivi, Anne-Laure Castell, Vincent Poitout, Miriam Cnop
    Journal of Molecular Biology.2020; 432(5): 1514.     CrossRef
  • Artemisinin and dihydroartemisinin promote β-cell apoptosis induced by palmitate via enhancing ER stress
    Ke Chen, Hu Hua, Ziyang Zhu, Tong Wu, Zhanjun Jia, Qianqi Liu
    Apoptosis.2020; 25(3-4): 192.     CrossRef
  • Mechanisms of impaired pancreatic β‑cell function in high‑fat diet‑induced obese mice: The role of endoplasmic reticulum stress
    Xiaoqing Yi, Xuan Cai, Sisi Wang, Yanfeng Xiao
    Molecular Medicine Reports.2020;[Epub]     CrossRef
  • Docosahexaenoic and Eicosapentaenoic Acids Prevent Altered-Muc2 Secretion Induced by Palmitic Acid by Alleviating Endoplasmic Reticulum Stress in LS174T Goblet Cells
    Quentin Escoula, Sandrine Bellenger, Michel Narce, Jérôme Bellenger
    Nutrients.2019; 11(9): 2179.     CrossRef
  • PPAR-γ agonist, pioglitazone, reduced oxidative and endoplasmic reticulum stress associated with L-NAME-induced hypertension in rats
    Eman Soliman, Shereen F. Behairy, Nabila N. El-maraghy, Shimaa M. Elshazly
    Life Sciences.2019; 239: 117047.     CrossRef
  • Changes of MODY signal pathway genes in the endoplasmic reticulum stress in INS-1-3 cells
    Yanan Dong, Shirui Li, Wenhui Zhao, Yanlei Wang, Tingting Ge, Jianzhong Xiao, Yukun Li, Herve Le Stunff
    PLOS ONE.2018; 13(6): e0198614.     CrossRef
Close layer
Obesity and Metabolism
Exendin-4 Inhibits the Expression of SEPP1 and Fetuin-A via Improvement of Palmitic Acid-Induced Endoplasmic Reticulum Stress by AMPK
Jinmi Lee, Seok-Woo Hong, Se Eun Park, Eun-Jung Rhee, Cheol-Young Park, Ki-Won Oh, Sung-Woo Park, Won-Young Lee
Endocrinol Metab. 2015;30(2):177-184.   Published online June 30, 2015
DOI: https://doi.org/10.3803/EnM.2015.30.2.177
  • 4,511 View
  • 45 Download
  • 12 Web of Science
  • 10 Crossref
AbstractAbstract PDFPubReader   
Background

Selenoprotein P (SEPP1) and fetuin-A, both circulating liver-derived glycoproteins, are novel biomarkers for insulin resistance and nonalcoholic fatty liver disease. However, the effect of exendin-4 (Ex-4), a glucagon-like peptide-1 receptor agonist, on the expression of hepatokines, SEPP1, and fetuin-A, is unknown.

Methods

The human hepatoma cell line HepG2 was treated with palmitic acid (PA; 0.4 mM) and tunicamycin (tuni; 2ug/ml) with or without exendin-4 (100 nM) for 24 hours. The change in expression of PA-induced SEPP1, fetuin-A, and endoplasmic reticulum (ER) stress markers by exendin-4 treatment were evaluated using quantitative real-time reverse transcription polymerase chain reaction and Western blotting. Transfection of cells with AMP-activated protein kinase (AMPK) small interfering RNA (siRNA) was performed to establish the effect of exendin-4-mediated AMPK in the regulation of SEPP1 and fetuin-A expression.

Results

Exendin-4 reduced the expression of SEPP1, fetuin-A, and ER stress markers including PKR-like ER kinase, inositol-requiring kinase 1α, activating transcription factor 6, and C/EBP homologous protein in HepG2 cells. Exendin-4 also reduced the expression of SEPP1 and fetuin-A in cells treated with tunicamycin, an ER stress inducer. In cells treated with the AMPK activator 5-aminoidazole-4-carboxamide ribonucleotide (AICAR), the expression of hepatic SEPP1 and fetuin-A were negatively related by AMPK, which is the target of exendin-4. In addition, exendin-4 treatment did not decrease SEPP1 and fetuin-A expression in cells transfected with AMPK siRNA.

Conclusion

These data suggest that exendin-4 can attenuate the expression of hepatic SEPP1 and fetuin-A via improvement of PA-induced ER stress by AMPK.

Citations

Citations to this article as recorded by  
  • Maternal Organic Selenium Supplementation Relieves Intestinal Endoplasmic Reticulum Stress in Piglets by Enhancing the Expression of Glutathione Peroxidase 4 and Selenoprotein S
    Dajiang Ding, Daolin Mou, Heng Zhu, Xuemei Jiang, Lianqiang Che, Zhengfeng Fang, Shengyu Xu, Yan Lin, Yong Zhuo, Jian Li, Chao Huang, Yuanfeng Zou, Lixia Li, De Wu, Bin Feng
    Frontiers in Nutrition.2022;[Epub]     CrossRef
  • Alliin, capsaicin, and gingerol attenuate endoplasmic reticulum stress-induced hepatic steatosis in HepG2 cells and C57BL/6N mice
    Ye-Rang Yun, Ji-Eun Lee
    Journal of Functional Foods.2022; 95: 105186.     CrossRef
  • PNPLA3 I148M is involved in the variability in anti-NAFLD response to exenatide
    Yunzhi Chen, Xuemei Yan, Xiao Xu, Shuhua Yuan, Fen Xu, Hua Liang
    Endocrine.2020; 70(3): 517.     CrossRef
  • Green tea extracts reduce leukocyte cell–Derived chemotaxin 2 and selenoprotein P levels in the livers of C57BL/6J mice fed a high-fat diet
    Shintaro Onishi, Hidefumi Kitazawa, Shinichi Meguro, Ichiro Tokimitsu
    Bioscience, Biotechnology, and Biochemistry.2018; 82(9): 1568.     CrossRef
  • Melatonin improves insulin resistance and hepatic steatosis through attenuation of alpha‐2‐HS‐glycoprotein
    Jee‐In Heo, Dae Wui Yoon, Ji Hee Yu, Nam Hoon Kim, Hye Jin Yoo, Ji A. Seo, Sin Gon Kim, Kyung Mook Choi, Sei Hyun Baik, Dong Seop Choi, Nan Hee Kim
    Journal of Pineal Research.2018;[Epub]     CrossRef
  • Palmitic acid induces ceramide accumulation, mitochondrial protein hyperacetylation, and mitochondrial dysfunction in porcine oocytes†
    Nobuhiko Itami, Koumei Shirasuna, Takehito Kuwayama, Hisataka Iwata
    Biology of Reproduction.2018; 98(5): 644.     CrossRef
  • Astragaloside IV attenuates free fatty acid-induced ER stress and lipid accumulation in hepatocytes via AMPK activation
    Bing Zhou, Dan-li Zhou, Xiao-hong Wei, Rong-yu Zhong, Jie Xu, Liao Sun
    Acta Pharmacologica Sinica.2017; 38(7): 998.     CrossRef
  • New Potential Targets of Glucagon-Like Peptide 1 Receptor Agonists in Pancreatic β-Cells and Hepatocytes
    Won-Young Lee
    Endocrinology and Metabolism.2017; 32(1): 1.     CrossRef
  • Selenoprotein P neutralizes lipopolysaccharide and participates in hepatic cell endoplasmic reticulum stress response
    Yongzhong Zhao, Shuvojit Banerjee, Ping Huang, Xinning Wang, Candece L. Gladson, Warren D. Heston, Charles B. Foster
    FEBS Letters.2016; 590(24): 4519.     CrossRef
  • Novel phenotypes of prediabetes?
    Hans-Ulrich Häring
    Diabetologia.2016; 59(9): 1806.     CrossRef
Close layer
Obesity and Metabolism
Activation of AMP-Activated Protein Kinase Attenuates Tumor Necrosis Factor-α-Induced Lipolysis via Protection of Perilipin in 3T3-L1 Adipocytes
Seok-Woo Hong, Jinmi Lee, Se Eun Park, Eun-Jung Rhee, Cheol-Young Park, Ki-Won Oh, Sung-Woo Park, Won-Young Lee
Endocrinol Metab. 2014;29(4):553-560.   Published online December 29, 2014
DOI: https://doi.org/10.3803/EnM.2014.29.4.553
  • 3,406 View
  • 26 Download
  • 12 Web of Science
  • 10 Crossref
AbstractAbstract PDFPubReader   
Background

Tumor necrosis factor (TNF)-α and AMP-activated protein kinase (AMPK) are known to stimulate and repress lipolysis in adipocytes, respectively; however, the mechanisms regulating these processes have not been completely elucidated.

Methods

The key factors and mechanism of action of TNF-α and AMPK in lipolysis were investigated by evaluating perilipin expression and activity of protein kinase RNA-like endoplasmic reticulum kinase (PERK)/eukaryotic initiation factor 2 α (eIF2α) by Western blot and an immunofluorescence assay in 24-hour TNF-α-treated 3T3-L1 adipocytes with artificial manipulation of AMPK activation.

Results

Enhancement of AMPK activity by the addition of activator minoimidazole carboxamide ribonucleotide (AICAR) suppressed TNF-α-induced lipolysis, whereas the addition of compound C, an inhibitor of AMPK phosphorylation, enhanced lipolysis. Perilipin, a lipid droplet-associated protein, was decreased by TNF-α and recovered following treatment with AICAR, showing a correlation with the antilipolytic effect of AICAR. Significant activation of PERK/eIF2α, a component of the unfolded protein response signaling pathway, was observed in TNF-α or vesicle-treated 3T3-L1 adipocytes. The antilipolytic effect and recovery of perilipin expression by AICAR in TNF-α-treated 3T3-L1 adipocytes were significantly diminished by treatment with 2-aminopurine, a specific inhibitor of eIF2α.

Conclusion

These data indicated that AICAR-induced AMPK activation attenuates TNF-α-induced lipolysis via preservation of perilipin in 3T3-L1 adipocytes. In addition, PERK/eIF2α activity is a novel mechanism of the anti-lipolytic effect of AICAR.

Citations

Citations to this article as recorded by  
  • Dysregulation of Lipid Droplet Protein Expression in Adipose Tissues and Association with Metabolic Risk Factors in Adult Females with Obesity and Type 2 Diabetes
    Chan Yoon Park, Donguk Kim, Min Kyeong Seo, Jimin Kim, Han Choe, Jong-Hyeok Kim, Joon Pio Hong, Yeon Ji Lee, Yoonseok Heo, Hwa Jung Kim, Hye Soon Park, Yeon Jin Jang
    The Journal of Nutrition.2023; 153(3): 691.     CrossRef
  • Tschimganidine reduces lipid accumulation through AMPK activation and alleviates high-fat diet-induced metabolic diseases
    Min-Seon Hwang, Jung-Hwan Baek, Jun-Kyu Song, In Hye Lee, Kyung-Hee Chun
    BMB Reports.2023; 56(4): 246.     CrossRef
  • Acetate stimulates lipogenesis via AMPKα signaling in rabbit adipose-derived stem cells
    Lei Liu, Chunyan Fu, Yongxu Liu, Fuchang Li
    General and Comparative Endocrinology.2021; 303: 113715.     CrossRef
  • Docosahexaenoic acid-enriched phospholipids and eicosapentaenoic acid-enriched phospholipids inhibit tumor necrosis factor-alpha-induced lipolysis in 3T3-L1 adipocytes by activating sirtuin 1 pathways
    Yu-Hong Yang, Yi-Ming Hao, Xiao-Fang Liu, Xiang Gao, Bao-Zhen Wang, Koretaro Takahashi, Lei Du
    Food & Function.2021; 12(11): 4783.     CrossRef
  • Role of the AMPK/ACC Signaling Pathway in TRPP2-Mediated Head and Neck Cancer Cell Proliferation
    Kun Li, Lei Chen, Zhangying Lin, Junwei Zhu, Yang Fang, Juan Du, Bing Shen, Kaile Wu, Yehai Liu, Gianmarco Saponaro
    BioMed Research International.2020; 2020: 1.     CrossRef
  • GLUT12 and adipose tissue: Expression, regulation and its relation with obesity in mice
    Eva Gil‐Iturbe, José Miguel Arbones‐Mainar, María J. Moreno‐Aliaga, María Pilar Lostao
    Acta Physiologica.2019;[Epub]     CrossRef
  • Bilobalide Suppresses Adipogenesis in 3T3-L1 Adipocytes via the AMPK Signaling Pathway
    Su Bu, Chun Ying Yuan, Quan Xue, Ying Chen, Fuliang Cao
    Molecules.2019; 24(19): 3503.     CrossRef
  • Sulforaphane induces adipocyte browning and promotes glucose and lipid utilization
    Hui Q. Zhang, Shi Y. Chen, An S. Wang, An J. Yao, Jian F. Fu, Jin S. Zhao, Fen Chen, Zu Q. Zou, Xiao H. Zhang, Yu J. Shan, Yong P. Bao
    Molecular Nutrition & Food Research.2016; 60(10): 2185.     CrossRef
  • Fyn phosphorylates AMPK to inhibit AMPK activity and AMP-dependent activation of autophagy
    Eijiro Yamada, Shuichi Okada, Claire C. Bastie, Manu Vatish, Yasuyo Nakajima, Ryo Shibusawa, Atsushi Ozawa, Jeffrey E. Pessin, Masanobu Yamada
    Oncotarget.2016; 7(46): 74612.     CrossRef
  • Articles in 'Endocrinology and Metabolism' in 2014
    Won-Young Lee
    Endocrinology and Metabolism.2015; 30(1): 47.     CrossRef
Close layer

Endocrinol Metab : Endocrinology and Metabolism