Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Search

Page Path
HOME > Search
32 "Fatty acid"
Filter
Filter
Article category
Keywords
Publication year
Authors
Funded articles
Original Articles
Basic Research
Article image
Extracellular Vimentin Alters Energy Metabolism And Induces Adipocyte Hypertrophy
Ji-Hae Park, Soyeon Kwon, Young Mi Park
Diabetes Metab J. 2024;48(2):215-230.   Published online September 26, 2023
DOI: https://doi.org/10.4093/dmj.2022.0332
  • 3,899 View
  • 299 Download
  • 3 Web of Science
  • 3 Crossref
AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
Previous studies have reported that oxidative stress contributes to obesity characterized by adipocyte hypertrophy. However, mechanism has not been studied extensively. In the current study, we evaluated role of extracellular vimentin secreted by oxidized low-density lipoprotein (oxLDL) in energy metabolism in adipocytes.
Methods
We treated 3T3-L1-derived adipocytes with oxLDL and measured vimentin which was secreted in the media. We evaluated changes in uptake of glucose and free fatty acid, expression of molecules functioning in energy metabolism, synthesis of adenosine triphosphate (ATP) and lactate, markers for endoplasmic reticulum (ER) stress and autophagy in adipocytes treated with recombinant vimentin.
Results
Adipocytes secreted vimentin in response to oxLDL. Microscopic evaluation revealed that vimentin treatment induced increase in adipocyte size and increase in sizes of intracellular lipid droplets with increased intracellular triglyceride. Adipocytes treated with vimentin showed increased uptake of glucose and free fatty acid with increased expression of plasma membrane glucose transporter type 1 (GLUT1), GLUT4, and CD36. Vimentin treatment increased transcription of GLUT1 and hypoxia-inducible factor 1α (Hif-1α) but decreased GLUT4 transcription. Adipose triglyceride lipase (ATGL), peroxisome proliferator-activated receptor γ (PPARγ), sterol regulatory element-binding protein 1 (SREBP1), diacylglycerol O-acyltransferase 1 (DGAT1) and 2 were decreased by vimentin treatment. Markers for ER stress were increased and autophagy was impaired in vimentin-treated adipocytes. No change was observed in synthesis of ATP and lactate in the adipocytes treated with vimentin.
Conclusion
We concluded that extracellular vimentin regulates expression of molecules in energy metabolism and promotes adipocyte hypertrophy. Our results show that vimentin functions in the interplay between oxidative stress and metabolism, suggesting a mechanism by which adipocyte hypertrophy is induced in oxidative stress.

Citations

Citations to this article as recorded by  
  • Novel secreted regulators of glucose and lipid metabolism in the development of metabolic diseases
    Lianna W. Wat, Katrin J. Svensson
    Diabetologia.2024;[Epub]     CrossRef
  • Mechanobiology in Metabolic Dysfunction-Associated Steatotic Liver Disease and Obesity
    Emily L. Rudolph, LiKang Chin
    Current Issues in Molecular Biology.2024; 46(7): 7134.     CrossRef
  • The Functions of SARS-CoV-2 Receptors in Diabetes-Related Severe COVID-19
    Adam Drzymała
    International Journal of Molecular Sciences.2024; 25(17): 9635.     CrossRef
Basic Research
Article image
Role of SUMO-Specific Protease 2 in Leptin-Induced Fatty Acid Metabolism in White Adipocytes
Praise Chanmee Kim, Ji Seon Lee, Sung Soo Chung, Kyong Soo Park
Diabetes Metab J. 2023;47(3):382-393.   Published online March 6, 2023
DOI: https://doi.org/10.4093/dmj.2022.0156
  • 4,300 View
  • 175 Download
  • 1 Web of Science
  • 1 Crossref
AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
Leptin is a 16-kDa fat-derived hormone with a primary role in controlling adipose tissue levels. Leptin increases fatty acid oxidation (FAO) acutely through adenosine monophosphate-activated protein kinase (AMPK) and on delay through the SUMO-specific protease 2 (SENP2)–peroxisome proliferator-activated receptor δ/γ (PPARδ/γ) pathway in skeletal muscle. Leptin also directly increases FAO and decreases lipogenesis in adipocytes; however, the mechanism behind these effects remains unknown. Here, we investigated the role of SENP2 in the regulation of fatty acid metabolism by leptin in adipocytes and white adipose tissues.
Methods
The effects of leptin mediated by SENP2 on fatty acid metabolism were tested by siRNA-mediated knockdown in 3T3-L1 adipocytes. The role of SENP2 was confirmed in vivo using adipocyte-specific Senp2 knockout (Senp2-aKO) mice. We revealed the molecular mechanism involved in the leptin-induced transcriptional regulation of carnitine palmitoyl transferase 1b (Cpt1b) and long-chain acyl-coenzyme A synthetase 1 (Acsl1) using transfection/reporter assays and chromatin immunoprecipitation.
Results
SENP2 mediated the increased expression of FAO-associated enzymes, CPT1b and ACSL1, which peaked 24 hours after leptin treatment in adipocytes. In contrast, leptin stimulated FAO through AMPK during the initial several hours after treatment. In white adipose tissues, FAO and mRNA levels of Cpt1b and Acsl1 were increased by 2-fold 24 hours after leptin injection in control mice but not in Senp2-aKO mice. Leptin increased PPARα binding to the Cpt1b and Acsl1 promoters in adipocytes through SENP2.
Conclusion
These results suggest that the SENP2-PPARα pathway plays an important role in leptin-induced FAO in white adipocytes.

Citations

Citations to this article as recorded by  
  • Intermittent cold stimulation affects energy metabolism and improves stress resistance in broiler heart
    Tingting Li, Haidong Wei, Shijie Zhang, Xiaotao Liu, Lu Xing, Yuanyuan Liu, Rixin Gong, Jianhong Li
    Poultry Science.2024; 103(1): 103190.     CrossRef
Complications
Fatty Acid-Binding Protein 4 in Patients with and without Diabetic Retinopathy
Ping Huang, Xiaoqin Zhao, Yi Sun, Xinlei Wang, Rong Ouyang, Yanqiu Jiang, Xiaoquan Zhang, Renyue Hu, Zhuqi Tang, Yunjuan Gu
Diabetes Metab J. 2022;46(4):640-649.   Published online April 28, 2022
DOI: https://doi.org/10.4093/dmj.2021.0195
  • 4,220 View
  • 204 Download
  • 2 Web of Science
  • 2 Crossref
AbstractAbstract PDFPubReader   ePub   
Background
Fatty acid-binding protein 4 (FABP4) has been demonstrated to be a predictor of early diabetic nephropathy. However, little is known about the relationship between FABP4 and diabetic retinopathy (DR). This study explored the value of FABP4 as a biomarker of DR in patients with type 2 diabetes mellitus (T2DM).
Methods
A total of 238 subjects were enrolled, including 20 healthy controls and 218 T2DM patients. Serum FABP4 levels were measured using a sandwich enzyme-linked immunosorbent assay. The grade of DR was determined using fundus fluorescence angiography. Based on the international classification of DR, all T2DM patients were classified into the following three subgroups: non-DR group, non-proliferative diabetic retinopathy (NPDR) group, and proliferative diabetic retinopathy (PDR) group. Multivariate logistic regression analyses were employed to assess the correlation between FABP4 levels and DR severity.
Results
FABP4 correlated positively with DR severity (r=0.225, P=0.001). Receiver operating characteristic curve analysis was used to assess the diagnostic potential of FABP4 in identifying DR, with an area under the curve of 0.624 (37% sensitivity, 83.6% specificity) and an optimum cut-off value of 76.4 μg/L. Multivariate logistic regression model including FABP4 as a categorized binary variable using the cut-off value of 76.4 μg/L showed that the concentration of FABP4 above the cut-off value increased the risk of NPDR (odds ratio [OR], 3.231; 95% confidence interval [CI], 1.574 to 6.632; P=0.001) and PDR (OR, 3.689; 95% CI, 1.306 to 10.424; P=0.014).
Conclusion
FABP4 may be used as a serum biomarker for the diagnosis of DR.

Citations

Citations to this article as recorded by  
  • Circulating AFABP, FGF21, and PEDF Levels as Prognostic Biomarkers of Sight-threatening Diabetic Retinopathy
    Chi-Ho Lee, David Tak-Wai Lui, Chloe Yu-Yan Cheung, Carol Ho-Yi Fong, Michele Mae-Ann Yuen, Yu-Cho Woo, Wing-Sun Chow, Ian Yat-Hin Wong, Aimin Xu, Karen Siu-Ling Lam
    The Journal of Clinical Endocrinology & Metabolism.2023; 108(9): e799.     CrossRef
  • A Prediction Model for Sight-Threatening Diabetic Retinopathy Based on Plasma Adipokines among Patients with Mild Diabetic Retinopathy
    Yaxin An, Bin Cao, Kun Li, Yongsong Xu, Wenying Zhao, Dong Zhao, Jing Ke, Takayuki Masaki
    Journal of Diabetes Research.2023; 2023: 1.     CrossRef
Metabolic Risk/Epidemiology
Article image
Postprandial Free Fatty Acids at Mid-Pregnancy Increase the Risk of Large-for-Gestational-Age Newborns in Women with Gestational Diabetes Mellitus
So-Yeon Kim, Young Shin Song, Soo-Kyung Kim, Yong-Wook Cho, Kyung-Soo Kim
Diabetes Metab J. 2022;46(1):140-148.   Published online August 9, 2021
DOI: https://doi.org/10.4093/dmj.2021.0023
  • 5,765 View
  • 168 Download
  • 3 Web of Science
  • 3 Crossref
Graphical AbstractGraphical Abstract AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
To investigate the association between free fatty acid (FFA) level at mid-pregnancy and large-for-gestational-age (LGA) newborns in women with gestational diabetes mellitus (GDM).
Methods
We enrolled 710 pregnant women diagnosed with GDM from February 2009 to October 2016. GDM was diagnosed by a ‘two-step’ approach with Carpenter and Coustan criteria. We measured plasma lipid profiles including fasting and 2-hour postprandial FFA (2h-FFA) levels at mid-pregnancy. LGA was defined if birthweights of newborns were above the 90th percentile for their gestational age.
Results
Mean age of pregnant women in this study was 33.1 years. Mean pre-pregnancy body mass index (BMI) was 22.4 kg/m2. The prevalence of LGA was 8.3% (n=59). Levels of 2h-FFA were higher in women who delivered LGA newborns than in those who delivered non-LGA newborns (416.7 μEq/L vs. 352.5 μEq/L, P=0.006). However, fasting FFA was not significantly different between the two groups. The prevalence of delivering LGA newborns was increased with increasing tertile of 2h-FFA (T1, 4.3%; T2, 9.8%; T3, 10.7%; P for trend <0.05). After adjustment for maternal age, pre-pregnancy BMI, and fasting plasma glucose, the highest tertile of 2h-FFA was 2.38 times (95% confidence interval, 1.11 to 5.13) more likely to have LGA newborns than the lowest tertile. However, there was no significant difference between groups according to fasting FFA tertiles.
Conclusion
In women with GDM, a high 2h-FFA level (but not fasting FFA) at mid-pregnancy is associated with an increasing risk of delivering LGA newborns.

Citations

Citations to this article as recorded by  
  • Advances in free fatty acid profiles in gestational diabetes mellitus
    Haoyi Du, Danyang Li, Laura Monjowa Molive, Na Wu
    Journal of Translational Medicine.2024;[Epub]     CrossRef
  • Modulation of gut microbiota and lipid metabolism in rats fed high-fat diets by Ganoderma lucidum triterpenoids
    Aijun Tong, Weihao Wu, Zhengxin Chen, Jiahui Wen, Ruibo Jia, Bin Liu, Hui Cao, Chao Zhao
    Current Research in Food Science.2023; 6: 100427.     CrossRef
  • Fetal Abdominal Obesity Detected at 24 to 28 Weeks of Gestation Persists until Delivery Despite Management of Gestational Diabetes Mellitus (Diabetes Metab J 2021;45:547-57)
    Wonjin Kim, Soo Kyung Park, Yoo Lee Kim
    Diabetes & Metabolism Journal.2021; 45(6): 970.     CrossRef
Pathophysiology
Article image
Distinct Dose-Dependent Association of Free Fatty Acids with Diabetes Development in Nonalcoholic Fatty Liver Disease Patients
Fuxi Li, Junzhao Ye, Yanhong Sun, Yansong Lin, Tingfeng Wu, Congxiang Shao, Qianqian Ma, Xianhua Liao, Shiting Feng, Bihui Zhong
Diabetes Metab J. 2021;45(3):417-429.   Published online March 15, 2021
DOI: https://doi.org/10.4093/dmj.2020.0039
  • 6,761 View
  • 162 Download
  • 9 Web of Science
  • 8 Crossref
Graphical AbstractGraphical Abstract AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
Excessive delivery of free fatty acids (FFAs) to the liver promotes steatosis and insulin resistance (IR), with IR defined as reduced glucose uptake, glycogen synthesis and anti-lipolysis stimulated by normal insulin levels. Whether the associations between FFAs and diabetes development differ between patients with and without nonalcoholic fatty liver disease (NAFLD) remains unclear.
Methods
Consecutive subjects (2,220 NAFLD subjects and 1,790 non-NAFLD subjects according to ultrasound imaging) were enrolled from the First Affiliated Hospital of Sun Yat-sen University between 2009 and 2019. The homeostasis model assessment of insulin resistance (HOMA-IR) was calculated.
Results
There was an approximate J-shaped relationship between FFA levels and HOMA-IR in the NAFLD group. Higher FFA concentration quartiles were associated with higher risks of IR (odds ratio [OR], 9.24; 95% confidence interval [CI], 6.43 to 13.36), prediabetes (OR, 10.48; 95% CI, 5.66 to 19.39), and type 2 diabetes mellitus (T2DM; OR, 19.43; 95% CI, 12.75 to 29.81) in the NAFLD group but not in the non-NAFLD group. The cut-off points for the FFA levels increased in a stepwise manner in discriminating IR, prediabetes and T2DM (573, 697, and 715 μmol/L) in the NAFLD group but not in non-NAFLD individuals.
Conclusion
A distinct dose-dependent relationship of FFA levels was found with IR, prediabetes and T2DM in NAFLD patients. Screening serum FFA levels in NAFLD patients would be valuable in preventing diabetes development.

Citations

Citations to this article as recorded by  
  • Mortality in metabolic dysfunction-associated steatotic liver disease: A nationwide population-based cohort study
    Eugene Han, Byung-Wan Lee, Eun Seok Kang, Bong-Soo Cha, Sang Hoon Ahn, Yong-ho Lee, Seung Up Kim
    Metabolism.2024; 152: 155789.     CrossRef
  • Global epidemiology of type 2 diabetes in patients with NAFLD or MAFLD: a systematic review and meta-analysis
    Limin Cao, Yu An, Huiyuan Liu, Jinguo Jiang, Wenqi Liu, Yuhan Zhou, Mengyuan Shi, Wei Dai, Yanling Lv, Yuhong Zhao, Yanhui Lu, Liangkai Chen, Yang Xia
    BMC Medicine.2024;[Epub]     CrossRef
  • Nutrition, health benefits, and processing of sand rice (Agriophyllum squarrosum): Comparisons with quinoa and buckwheat
    Xiaofan Yang, Wenting Fu, Liuyang Xiao, Zhaojun Wei, Lihong Han
    Food Science & Nutrition.2024;[Epub]     CrossRef
  • Metabolic Dysfunction-Associated Fatty Liver Disease and Mortality: A Population-Based Cohort Study
    Kyung-Soo Kim, Sangmo Hong, Hong-Yup Ahn, Cheol-Young Park
    Diabetes & Metabolism Journal.2023; 47(2): 220.     CrossRef
  • Experimental model and novel therapeutic targets for non-alcoholic fatty liver disease development
    Yujin Jin, Kyung-Sun Heo
    The Korean Journal of Physiology & Pharmacology.2023; 27(4): 299.     CrossRef
  • Triglyceride and glucose index is a simple and easy‐to‐calculate marker associated with nonalcoholic fatty liver disease
    Kyung‐Soo Kim, Sangmo Hong, Hong‐Yup Ahn, Cheol‐Young Park
    Obesity.2022; 30(6): 1279.     CrossRef
  • Mongolian medicine in treating type 2 diabetes mellitus combined with nonalcoholic fatty liver disease via FXR/LXR-mediated P2X7R/NLRP3/NF-κB pathway activation
    Shuyin Bao, Xiuzhi Wang, Qianqian Ma, Chengxi Wei, Jixing Nan, Wuliji Ao
    Chinese Herbal Medicines.2022; 14(3): 367.     CrossRef
  • Triglyceride Glucose-Waist Circumference Is Superior to the Homeostasis Model Assessment of Insulin Resistance in Identifying Nonalcoholic Fatty Liver Disease in Healthy Subjects
    Hwi Seung Kim, Yun Kyung Cho, Eun Hee Kim, Min Jung Lee, Chang Hee Jung, Joong-Yeol Park, Hong-Kyu Kim, Woo Je Lee
    Journal of Clinical Medicine.2021; 11(1): 41.     CrossRef
Review
Basic Research
The Role of CD36 in Type 2 Diabetes Mellitus: β-Cell Dysfunction and Beyond
Jun Sung Moon, Udayakumar Karunakaran, Elumalai Suma, Seung Min Chung, Kyu Chang Won
Diabetes Metab J. 2020;44(2):222-233.   Published online April 23, 2020
DOI: https://doi.org/10.4093/dmj.2020.0053
  • 8,593 View
  • 186 Download
  • 18 Web of Science
  • 17 Crossref
AbstractAbstract PDFPubReader   

Impaired β-cell function is the key pathophysiology of type 2 diabetes mellitus, and chronic exposure of nutrient excess could lead to this tragedy. For preserving β-cell function, it is essential to understand the cause and mechanisms about the progression of β-cells failure. Glucotoxicity, lipotoxicity, and glucolipotoxicity have been suggested to be a major cause of β-cell dysfunction for decades, but not yet fully understood. Fatty acid translocase cluster determinant 36 (CD36), which is part of the free fatty acid (FFA) transporter system, has been identified in several tissues such as muscle, liver, and insulin-producing cells. Several studies have reported that induction of CD36 increases uptake of FFA in several cells, suggesting the functional interplay between glucose and FFA in terms of insulin secretion and oxidative metabolism. However, we do not currently know the regulating mechanism and physiological role of CD36 on glucolipotoxicity in pancreatic β-cells. Also, the downstream and upstream targets of CD36 related signaling have not been defined. In the present review, we will focus on the expression and function of CD36 related signaling in the pancreatic β-cells in response to hyperglycemia and hyperlipidemia (ceramide) along with the clinical studies on the association between CD36 and metabolic disorders.

Citations

Citations to this article as recorded by  
  • Nrf2 inhibition regulates intracellular lipid accumulation in mouse insulinoma cells and improves insulin secretory function
    Alpana Mukhuty, Samanwita Mandal, Chandrani Fouzder, Snehasis Das, Dipanjan Chattopadhyay, Tanmay Majumdar, Rakesh Kundu
    Molecular and Cellular Endocrinology.2024; 581: 112112.     CrossRef
  • CD36 gene variant rs1761667(G/A) as a biomarker in obese type 2 diabetes mellitus cases
    Ashwin Kumar Shukla, Amreen Shamsad, Atar Singh Kushwah, Shalini Singh, Kauser Usman, Monisha Banerjee
    Egyptian Journal of Medical Human Genetics.2024;[Epub]     CrossRef
  • CD36 regulates macrophage and endothelial cell activation and multinucleate giant cell formation in anti neutrophil cytoplasm antibody vasculitis
    Xiang Zhang, Catherine King, Alexander Dowell, Paul Moss, Lorraine Harper, Dimitrios Chanouzas, Xiong-zhong Ruan, Alan David Salama
    Clinical Immunology.2024; 260: 109914.     CrossRef
  • Identification and validation of oxidative stress-related genes in sepsis-induced myopathy
    Ning Zhang, Dan Huang, Xiang Li, JinXia Yan, Qi Yan, WeiXing Ge, Jun Zhou
    Medicine.2024; 103(18): e37933.     CrossRef
  • The association of soluble cluster of differentiation 36 with metabolic diseases: A potential biomarker and therapeutic target
    Yun Li, Yaxi Chen, Xiong Z. Ruan
    Pediatric Discovery.2023;[Epub]     CrossRef
  • The role of candidate transport proteins in β‐cell long‐chain fatty acid uptake: Where are we now?
    Christina Clavelo‐Farrow, Patricia Thomas
    Diabetic Medicine.2023;[Epub]     CrossRef
  • SARS-CoV-2 in the pancreas and the impaired islet function in COVID-19 patients
    Ningfei Ji, Mingshun Zhang, Liang Ren, Yunyun Wang, Bicheng Hu, Jie Xiang, Yingyun Gong, Chaojie Wu, Guoqiang Qu, Wenqiu Ding, Zhiqiang Yin, Shan Li, Zhengxia Wang, Lianzheng Zhou, Xueqin Chen, Yuan Ma, Jinhai Tang, Yun Liu, Liang Liu, Mao Huang
    Emerging Microbes & Infections.2022; 11(1): 1115.     CrossRef
  • Is imaging-based muscle quantity associated with risk of diabetes? A meta-analysis of cohort studies
    Shanhu Qiu, Xue Cai, Yang Yuan, Bo Xie, Zilin Sun, Tongzhi Wu
    Diabetes Research and Clinical Practice.2022; 189: 109939.     CrossRef
  • Lipotoxicity in a Vicious Cycle of Pancreatic Beta Cell Exhaustion
    Vladimir Grubelnik, Jan Zmazek, Matej Završnik, Marko Marhl
    Biomedicines.2022; 10(7): 1627.     CrossRef
  • Association of cluster determinant 36, scavenger receptor class B type 1, and major facilitator superfamily domain containing the 2a genetic polymorphism with serum lipid profile in aging population with type 2 diabetes mellitus
    Xixiang Wang, Xiaojun Ma, Jingjing Xu, Yujie Guo, Shaobo Zhou, Huiyan Yu, Linhong Yuan
    Frontiers in Nutrition.2022;[Epub]     CrossRef
  • CD36-Fatty Acid-Mediated Metastasis via the Bidirectional Interactions of Cancer Cells and Macrophages
    Noorzaileen Eileena Zaidi, Nur Aima Hafiza Shazali, Thean-Chor Leow, Mohd Azuraidi Osman, Kamariah Ibrahim, Wan-Hee Cheng, Kok-Song Lai, Nik Mohd Afizan Nik Abd Rahman
    Cells.2022; 11(22): 3556.     CrossRef
  • The Past and Present Lives of the Intraocular Transmembrane Protein CD36
    Rucui Yang, Qingping Liu, Mingzhi Zhang
    Cells.2022; 12(1): 171.     CrossRef
  • Implicating the effect of ketogenic diet as a preventive measure to obesity and diabetes mellitus
    Sachin Kumar, Tapan Behl, Monika Sachdeva, Aayush Sehgal, Shilpa Kumari, Arun Kumar, Gagandeep Kaur, Harlokesh Narayan Yadav, Simona Bungau
    Life Sciences.2021; 264: 118661.     CrossRef
  • Contribution of rs3211938 polymorphism at CD36 to glucose levels, oxidized low-density lipoproteins, insulin resistance, and body mass index in Mexican mestizos with type-2 diabetes from western Mexico
    Beatriz Teresita Martín-Márquez, Flavio Sandoval-Garcia, Mónica Vazquez-Del Mercado, Erika-Aurora Martínez-García, Fernanda-Isadora Corona-Meraz, Ana-Lilia Fletes-Rayas, Soraya-Amalí Zavaleta-Muñiz
    Nutrición Hospitalaria.2021;[Epub]     CrossRef
  • Investigating the association of CD36 gene polymorphisms (rs1761667 and rs1527483) with T2DM and dyslipidemia: Statistical analysis, machine learning based prediction, and meta-analysis
    Ma’mon M. Hatmal, Walhan Alshaer, Ismail S. Mahmoud, Mohammad A. I. Al-Hatamleh, Hamzeh J. Al-Ameer, Omar Abuyaman, Malek Zihlif, Rohimah Mohamud, Mais Darras, Mohammad Al Shhab, Rand Abu-Raideh, Hilweh Ismail, Ali Al-Hamadi, Ali Abdelhay, Kanhaiya Singh
    PLOS ONE.2021; 16(10): e0257857.     CrossRef
  • Misregulation of Wnt Signaling Pathways at the Plasma Membrane in Brain and Metabolic Diseases
    Mustafa Karabicici, Yagmur Azbazdar, Evin Iscan, Gunes Ozhan
    Membranes.2021; 11(11): 844.     CrossRef
  • CD36 Signal Transduction in Metabolic Diseases: Novel Insights and Therapeutic Targeting
    Udayakumar Karunakaran, Suma Elumalai, Jun-Sung Moon, Kyu-Chang Won
    Cells.2021; 10(7): 1833.     CrossRef
Original Articles
Obesity and Metabolic Syndrome
PF-04620110, a Potent Antidiabetic Agent, Suppresses Fatty Acid-Induced NLRP3 Inflammasome Activation in Macrophages
Seung Il Jo, Jung Hwan Bae, Seong Jin Kim, Jong Min Lee, Ji Hun Jeong, Jong-Seok Moon
Diabetes Metab J. 2019;43(5):683-699.   Published online October 24, 2019
DOI: https://doi.org/10.4093/dmj.2019.0112
  • 6,080 View
  • 70 Download
  • 3 Web of Science
  • 2 Crossref
AbstractAbstract PDFSupplementary MaterialPubReader   
Background

Chronic inflammation has been linked to insulin resistance and type 2 diabetes mellitus (T2DM). High-fat diet (HFD)-derived fatty acid is associated with the activation of chronic inflammation in T2DM. PF-04620110, which is currently in phase 1 clinical trials as a selective acyl-CoA:diacylglycerol acyltransferase-1 (DGAT1) inhibitor, is a potent anti-diabetic agent that may be important for the regulation of chronic inflammation in T2DM. However, the mechanisms by which PF-04620110 regulates fatty acid-induced chronic inflammation remain unclear.

Methods

PF-04620110 was used in vitro and in vivo. DGAT1-targeting gRNAs were used for deletion of mouse DGAT1 via CRISPR ribonucleoprotein (RNP) system. The activation of NLRP3 inflammasome was measured by immunoblot or cytokine analysis in vitro and in vivo.

Results

Here we show that PF-04620110 suppressed fatty acid-induced nucleotide-binding domain, leucine-rich-repeat-containing receptor (NLR), pyrin-domain-containing 3 (NLRP3) inflammasome activation in macrophages. In contrast, PF-04620110 did not change the activation of the NLR family, CARD-domain-containing 4 (NLRC4), or the absent in melanoma 2 (AIM2) inflammasomes. Moreover, PF-04620110 inhibited K+ efflux and the NLRP3 inflammasome complex formation, which are required for NLRP3 inflammasome activation. PF-04620110 reduced the production of interleukin 1β (IL-1β) and IL-18 and blood glucose levels in the plasma of mice fed HFD. Furthermore, genetic inhibition of DGAT1 suppressed fatty acid-induced NLRP3 inflammasome activation.

Conclusion

Our results suggest that PF-04620110 suppresses fatty acid-induced NLRP3 inflammasome activation.

Citations

Citations to this article as recorded by  
  • Drug Targeting of Acyltransferases in the Triacylglyceride and 1-O-AcylCeramide Biosynthetic Pathways
    Maria Hernandez-Corbacho, Daniel Canals
    Molecular Pharmacology.2024; 105(3): 166.     CrossRef
  • Possible therapeutic targets for NLRP3 inflammasome-induced breast cancer
    Xixi Wang, Junyi Lin, Zhe Wang, Zhi Li, Minghua Wang
    Discover Oncology.2023;[Epub]     CrossRef
Drug/Regimen
Efficacy and Safety of Omega-3 Fatty Acids in Patients Treated with Statins for Residual Hypertriglyceridemia: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial
Ji Eun Jun, In-Kyung Jeong, Jae Myung Yu, Sung Rae Kim, In Kye Lee, Kyung-Ah Han, Sung Hee Choi, Soo-Kyung Kim, Hyeong Kyu Park, Ji-Oh Mok, Yong-ho Lee, Hyuk-Sang Kwon, So Hun Kim, Ho-Cheol Kang, Sang Ah Lee, Chang Beom Lee, Kyung Mook Choi, Sung-Ho Her, Won Yong Shin, Mi-Seung Shin, Hyo-Suk Ahn, Seung Ho Kang, Jin-Man Cho, Sang-Ho Jo, Tae-Joon Cha, Seok Yeon Kim, Kyung Heon Won, Dong-Bin Kim, Jae Hyuk Lee, Moon-Kyu Lee
Diabetes Metab J. 2020;44(1):78-90.   Published online June 20, 2019
DOI: https://doi.org/10.4093/dmj.2018.0265
  • 10,430 View
  • 218 Download
  • 7 Web of Science
  • 8 Crossref
AbstractAbstract PDFSupplementary MaterialPubReader   
Background

Cardiovascular risk remains increased despite optimal low density lipoprotein cholesterol (LDL-C) level induced by intensive statin therapy. Therefore, recent guidelines recommend non-high density lipoprotein cholesterol (non-HDL-C) as a secondary target for preventing cardiovascular events. The aim of this study was to assess the efficacy and tolerability of omega-3 fatty acids (OM3-FAs) in combination with atorvastatin compared to atorvastatin alone in patients with mixed dyslipidemia.

Methods

This randomized, double-blind, placebo-controlled, parallel-group, and phase III multicenter study included adults with fasting triglyceride (TG) levels ≥200 and <500 mg/dL and LDL-C levels <110 mg/dL. Eligible subjects were randomized to ATOMEGA (OM3-FAs 4,000 mg plus atorvastatin calcium 20 mg) or atorvastatin 20 mg plus placebo groups. The primary efficacy endpoints were the percent changes in TG and non-HDL-C levels from baseline at the end of treatment.

Results

After 8 weeks of treatment, the percent changes from baseline in TG (−29.8% vs. 3.6%, P<0.001) and non-HDL-C (−10.1% vs. 4.9%, P<0.001) levels were significantly greater in the ATOMEGA group (n=97) than in the atorvastatin group (n=103). Moreover, the proportion of total subjects reaching TG target of <200 mg/dL in the ATOMEGA group was significantly higher than that in the atorvastatin group (62.9% vs. 22.3%, P<0.001). The incidence of adverse events did not differ between the two groups.

Conclusion

The addition of OM3-FAs to atorvastatin improved TG and non-HDL-C levels to a significant extent compared to atorvastatin alone in subjects with residual hypertriglyceridemia.

Citations

Citations to this article as recorded by  
  • Current trends in solving the problem of residual cardiovascular risk
    N. Yu. Obedkova, A. A. Guslyakova, G. S. Mal, E. G. Obedkov
    Meditsinskiy sovet = Medical Council.2024; (6): 155.     CrossRef
  • Association Between Omega‐3 Fatty Acid Intake and Dyslipidemia: A Continuous Dose–Response Meta‐Analysis of Randomized Controlled Trials
    Tianjiao Wang, Xin Zhang, Na Zhou, Yuxuan Shen, Biao Li, Bingshu E. Chen, Xinzhi Li
    Journal of the American Heart Association.2023;[Epub]     CrossRef
  • Nutraceutical support in the prevention and treatment of cardiovascular diseases
    E. V. Gracheva, E. A. Starovoytova, E. S. Kulikov, N. A. Kirillova, S. V. Fedosenko, M. A. Balaganskaya, D. V. Kromka
    Rational Pharmacotherapy in Cardiology.2023; 19(3): 298.     CrossRef
  • Effect of coadministration of omega-3 fatty acids with glimepiride on glycemic control, lipid profile, irisin, and sirtuin-1 in type 2 diabetes mellitus patients: a randomized controlled trial
    Rehab H. Werida, Aalaa Ramzy, Youssri Nassief Ebrahim, Maged Wasfy Helmy
    BMC Endocrine Disorders.2023;[Epub]     CrossRef
  • The Effect of Dietary Interventions on Hypertriglyceridemia: From Public Health to Molecular Nutrition Evidence
    Karla Paulina Luna-Castillo, Xochitl Citlalli Olivares-Ochoa, Rocío Guadalupe Hernández-Ruiz, Iris Monserrat Llamas-Covarrubias, Saraí Citlalic Rodríguez-Reyes, Alejandra Betancourt-Núñez, Barbara Vizmanos, Erika Martínez-López, José Francisco Muñoz-Valle
    Nutrients.2022; 14(5): 1104.     CrossRef
  • The effect of omega-3 fatty acids and its combination with statins on lipid profile in patients with hypertriglyceridemia: A systematic review and meta-analysis of randomized controlled trials
    Yunjiao Yang, Wen Deng, Yanmei Wang, Tongyi Li, Yiding Chen, Cong Long, Qing Wen, Yue Wu, Qiu Chen
    Frontiers in Nutrition.2022;[Epub]     CrossRef
  • Comparison of the Efficacy and Safety of Atorvastatin 40 mg/ω-3 Fatty Acids 4 g Fixed-dose Combination and Atorvastatin 40 mg Monotherapy in Hypertriglyceridemic Patients who Poorly Respond to Atorvastatin 40 mg Monotherapy: An 8-week, Multicenter, Random
    Jong Shin Woo, Soon Jun Hong, Dong Hoon Cha, Kee Sik Kim, Moo Hyun Kim, Jun-Won Lee, Myung Ho Jeong, Jin-Ok Jeong, Jun-Hee Lee, Doo Soo Jeon, Eun Joo Cho, Soon Kil Kim, Jun Kwan, Chang Gyu Park, Hae Young Lee, Taek Jong Hong, Jinho Shin, Ho Joong Youn, Do
    Clinical Therapeutics.2021; 43(8): 1419.     CrossRef
  • All-Cause Mortality and Cardiovascular Death between Statins and Omega-3 Supplementation: A Meta-Analysis and Network Meta-Analysis from 55 Randomized Controlled Trials
    Jeongseon Kim, Tung Hoang, Ji-Myung Kim, So Young Bu, Jeong-Hwa Choi, Eunju Park, Seung-Min Lee, Eunmi Park, Ji Yeon Min, In Seok Lee, So Young Youn, Jee-Young Yeon
    Nutrients.2020; 12(10): 3203.     CrossRef
Clinical Diabetes & Therapeutics
Comparison of the Efficacy of Rosuvastatin Monotherapy 20 mg with Rosuvastatin 5 mg and Ezetimibe 10 mg Combination Therapy on Lipid Parameters in Patients with Type 2 Diabetes Mellitus
You-Cheol Hwang, Ji Eun Jun, In-Kyung Jeong, Kyu Jeung Ahn, Ho Yeon Chung
Diabetes Metab J. 2019;43(5):582-589.   Published online January 16, 2019
DOI: https://doi.org/10.4093/dmj.2018.0124
  • 7,959 View
  • 210 Download
  • 15 Web of Science
  • 13 Crossref
AbstractAbstract PDFPubReader   
Background

The apolipoprotein B/A1 (apoB/A1) ratio is a stronger predictor of future cardiovascular disease than is the level of conventional lipids. Statin and ezetimibe combination therapy have shown additional cardioprotective effects over statin monotherapy.

Methods

This was a single-center, randomized, open-label, active-controlled study in Korea. A total of 36 patients with type 2 diabetes mellitus were randomized to either rosuvastatin monotherapy (20 mg/day, n=20) or rosuvastatin/ezetimibe (5 mg/10 mg/day, n=16) combination therapy for 6 weeks.

Results

After the 6-week treatment, low density lipoprotein cholesterol (LDL-C) and apoB reduction were comparable between the two groups (−94.3±15.4 and −62.0±20.9 mg/dL in the rosuvastatin group, −89.9±22.7 and −66.8±21.6 mg/dL in the rosuvastatin/ezetimibe group, P=0.54 and P=0.86, respectively). In addition, change in apoB/A1 ratio (−0.44±0.16 in the rosuvastatin group and −0.47±0.25 in the rosuvastatin/ezetimibe group, P=0.58) did not differ between the two groups. On the other hand, triglyceride and free fatty acid (FFA) reductions were greater in the rosuvastatin/ezetimibe group than in the rosuvastatin group (−10.5 mg/dL [interquartile range (IQR), −37.5 to 29.5] and 0.0 µEq/L [IQR, −136.8 to 146.0] in the rosuvastatin group, −49.5 mg/dL [IQR, −108.5 to −27.5] and −170.5 µEq/L [IQR, −353.0 to 0.8] in the rosuvastatin/ezetimibe group, P=0.010 and P=0.049, respectively). Both treatments were generally well tolerated, and there were no differences in muscle or liver enzyme elevation.

Conclusion

A 6-week combination therapy of low-dose rosuvastatin and ezetimibe showed LDL-C, apoB, and apoB/A1 ratio reduction comparable to that of high-dose rosuvastatin monotherapy in patients with type 2 diabetes mellitus. Triglyceride and FFA reductions were greater with the combination therapy than with rosuvastatin monotherapy.

Citations

Citations to this article as recorded by  
  • Moderate-Intensity Rosuvastatin/Ezetimibe Combination versus Quadruple-Dose Rosuvastatin Monotherapy: A Meta-Analysis and Systemic Review
    Yura Kang, Jung Mi Park, Sang-Hak Lee
    Yonsei Medical Journal.2024; 65(1): 19.     CrossRef
  • A Comparison of Rosuvastatin Monotherapy and Rosuvastatin Plus Ezetimibe Combination Therapy in Patients With Type 2 Diabetes: A Meta-Analysis of Randomized Controlled Trials
    Samuel K Dadzie, Godfrey Tabowei, Mandeep Kaur, Saeed Ahmed, Aayushi Thakur, Khaldoun Khreis, Monika Bai, Adil Amin
    Cureus.2024;[Epub]     CrossRef
  • Combination Therapy of Ezetimibe and Rosuvastatin for Dyslipidemia: Current Insights
    Maya R Chilbert, Dylan VanDuyn, Sara Salah, Collin M Clark, Qing Ma
    Drug Design, Development and Therapy.2022; Volume 16: 2177.     CrossRef
  • Ezetimibe and diabetes mellitus:a new strategy for lowering cholesterol
    V.A. Serhiyenko, A.A. Serhiyenko
    INTERNATIONAL JOURNAL OF ENDOCRINOLOGY (Ukraine).2022; 18(5): 302.     CrossRef
  • The Effect of Rosuvastatin on Plasma/Serum Levels of High-Sensitivity C-Reactive Protein, Interleukin-6, and D-Dimer in People Living with Human Immunodeficiency Virus: A Systematic Review and Meta-Analysis
    Akililu Alemu Ashuro, Yin-Guang Fan, Yuan-Sheng Fu, Dong-Sheng Di, Napoleon Bellua Sam, Hai-Feng Pan, Dong-Qing Ye
    AIDS Research and Human Retroviruses.2021; 37(11): 821.     CrossRef
  • Comparison of the Efficacy and Safety of Rosuvastatin/Ezetimibe Combination Therapy and Rosuvastatin Monotherapy on Lipoprotein in Patients With Type 2 Diabetes: Multicenter Randomized Controlled Study
    Jiwoo Lee, You-Cheol Hwang, Woo Je Lee, Jong Chul Won, Kee-Ho Song, Cheol-Young Park, Kyu Jeung Ahn, Joong-Yeol Park
    Diabetes Therapy.2020; 11(4): 859.     CrossRef
  • Comparison of Renal Effects of Ezetimibe–Statin Combination versus Statin Monotherapy: A Propensity-Score-Matched Analysis
    Jaehyun Bae, Namki Hong, Byung-Wan Lee, Eun Seok Kang, Bong-Soo Cha, Yong-ho Lee
    Journal of Clinical Medicine.2020; 9(3): 798.     CrossRef
  • Combined use of rosuvastatin and ezetimibe improves hepatic steatosis in patients with dyslipidemia
    Won Dong Lee, Beom Kyung Kim, Jun Yong Park, Do Young Kim, Sang Hoon Ahn, Kwang-Hyub Han, Seung Up Kim
    European Journal of Gastroenterology & Hepatology.2020; 32(12): 1538.     CrossRef
  • Influence of rosuvastatin dose on total fatty acids and free fatty acids in plasma
    Cristian I. Ciucanu, Sonia Olariu, Daliborca C. Vlad, Victor Dumitraşcu
    Medicine.2020; 99(48): e23356.     CrossRef
  • The effect of switching from statin-monotherapy to statin/ezetimibe combination therapy on lipid profiles in patients with type 2 diabetes and dyslipidemia: a multicenter open-label study (EUCLID)
    Mitsuhide Takeshita, Atsushi Tanaka, Atsushi Kawaguchi, Keiko Sato, Shigeru Toyoda, Teruo Inoue, Koichi Node
    Vascular Failure.2020; 4(1): 22.     CrossRef
  • Response: Comparison of the Efficacy of Rosuvastatin Monotherapy 20 mg with Rosuvastatin 5 mg and Ezetimibe 10 mg Combination Therapy on Lipid Parameters in Patients with Type 2 Diabetes Mellitus (Diabetes Metab J 2019;43:582–9)
    You-Cheol Hwang
    Diabetes & Metabolism Journal.2019; 43(6): 915.     CrossRef
  • Letter: Comparison of the Efficacy of Rosuvastatin Monotherapy 20 mg with Rosuvastatin 5 mg and Ezetimibe 10 mg Combination Therapy on Lipid Parameters in Patients with Type 2 Diabetes Mellitus (Diabetes Metab J2019;43:582–9)
    Tae Seo Sohn
    Diabetes & Metabolism Journal.2019; 43(6): 909.     CrossRef
  • Changes in Plasma Free Fatty Acids Associated with Type-2 Diabetes
    Amélie I. S. Sobczak, Claudia A. Blindauer, Alan J. Stewart
    Nutrients.2019; 11(9): 2022.     CrossRef
Review
Clinical Diabetes & Therapeutics
Effects of Omega-3 Supplementation on Adipocytokines in Prediabetes and Type 2 Diabetes Mellitus: Systematic Review and Meta-Analysis of Randomized Controlled Trials
Tarik Becic, Christian Studenik
Diabetes Metab J. 2018;42(2):101-116.   Published online April 19, 2018
DOI: https://doi.org/10.4093/dmj.2018.42.2.101
  • 8,751 View
  • 66 Download
  • 16 Web of Science
  • 17 Crossref
AbstractAbstract PDFPubReader   
Background

The objective of this systematic review and meta-analysis was to determine the effects of omega-3 supplementation on adipocytokine levels in adult prediabetic and diabetic individuals.

Methods

We searched PubMed, Medline, EMBASE, Scopus, Web of Science, Google Scholar, Cochrane Trial Register, World Health Organization Clinical Trial Registry Platform, and Clinicaltrial.gov Registry from inception to August 1, 2017 for randomized controlled trials. Pooled effects of interventions were assessed as mean difference using random effects model. We conducted a sensitivity, publication bias and subgroup analysis.

Results

Fourteen studies individuals (n=685) were included in the meta-analysis. Omega-3 supplementation increased levels of adiponectin (0.48 µg/mL; 95% confidence interval [CI], 0.27 to 0.68; P<0.00001, n=10 trials), but effects disappeared after sensitivity analysis. Tumor necrosis factor α (TNF-α) levels were reduced (−1.71; 95% CI, −3.38 to −0.14; P=0.03, n=8 trials). Treatment duration shorter than 12 weeks was associated with greater reduction than longer treatment duration. Levels of other adipocytokines were not significantly affected. Publication bias could generally not be excluded.

Conclusion

Eicosapentaenoic acid and docosahexaenoic acid supplementation may increase adiponectin and reduce TNF-α levels in this population group. However, due to overall study heterogeneity and potential publication bias, a cautious interpretation is needed.

Citations

Citations to this article as recorded by  
  • The Effects of Omega-3 Fatty Acid Supplementation on the Lipid Profile and Cardiovascular Markers Following Downhill Running in Long-Distance Runners
    Marzena Jaworska, Szymon Siatkowski, Aleksandra Żebrowska
    Journal of Human Kinetics.2023; 89: 123.     CrossRef
  • Omega-3 supplementation in the treatment of polycystic ovary syndrome (PCOS) – a review of clinical trials and cohort
    Vitoria Melo, Thomas Silva, Thaissa Silva, Juliana Freitas, Joselita Sacramento, Mirian Vazquez, Edilene Araujo
    Endocrine Regulations.2022; 56(1): 66.     CrossRef
  • The effects of omega-3 fatty acids in type 2 diabetes: A systematic review and meta-analysis
    Yanan Xiao, Qifang Zhang, Xueling Liao, Ulf Elbelt, Karsten H. Weylandt
    Prostaglandins, Leukotrienes and Essential Fatty Acids.2022; 182: 102456.     CrossRef
  • Anti-inflammatory and antioxidant activity of astragalus polysaccharide in ulcerative colitis: A systematic review and meta-analysis of animal studies
    Heng-Chang Hu, Wei Zhang, Pei-Yu Xiong, Li Song, Bo Jia, Xing-Long Liu
    Frontiers in Pharmacology.2022;[Epub]     CrossRef
  • Eicosopenthaenoic acid: Gnawing at the perivascular adipose tissue
    Paolo Raggi, Arthur E. Stillman
    Atherosclerosis.2021; 316: 69.     CrossRef
  • The Effect of Omega-3 Fatty Acid Supplementation on Serum Adipocytokines, Lipid Profile and Biochemical Markers of Inflammation in Recreational Runners
    Aleksandra Żebrowska, Barbara Hall, Anna Stolecka-Warzecha, Arkadiusz Stanula, Ewa Sadowska-Krępa
    Nutrients.2021; 13(2): 456.     CrossRef
  • The Influence of Nutrition on Adiponectin—A Narrative Review
    Justyna Janiszewska, Joanna Ostrowska, Dorota Szostak-Węgierek
    Nutrients.2021; 13(5): 1394.     CrossRef
  • Adiponectin’s roles in lipid and glucose metabolism modulation in fish: Mechanisms and perspectives
    Renlei Ji, Xiang Xu, Giovanni M. Turchini, Kangsen Mai, Qinghui Ai
    Reviews in Aquaculture.2021; 13(4): 2305.     CrossRef
  • Omega-3 Fatty Acids and Vulnerability to Addiction: Reviewing Preclinical and Clinical Evidence
    Valerie L. Darcey, Katherine M. Serafine
    Current Pharmaceutical Design.2020; 26(20): 2385.     CrossRef
  • Therapeutic effects of different doses of prebiotic (isolated from Saccharomyces cerevisiae) in comparison to n-3 supplement on glycemic control, lipid profiles and immunological response in diabetic rats
    Janina de Sales Guilarducci, Breno Augusto Ribeiro Marcelino, Isaac Filipe Moreira Konig, Tamira Maria Orlando, Mary Suzan Varaschin, Luciano José Pereira
    Diabetology & Metabolic Syndrome.2020;[Epub]     CrossRef
  • Endothelial dysfunction in neuroprogressive disorders—causes and suggested treatments
    Gerwyn Morris, Basant K. Puri, Lisa Olive, Andre Carvalho, Michael Berk, Ken Walder, Lise Tuset Gustad, Michael Maes
    BMC Medicine.2020;[Epub]     CrossRef
  • Evaluation of walking exercise on glycemic control in patients with type 2 diabetes mellitus
    Hengchang Hu, Yuanhong Lei, Liping Yin, Xiaoqiong Luo
    Medicine.2020; 99(47): e22735.     CrossRef
  • Omega-3 polyunsaturated fatty acids in supporting pregnancy and fetal development: dosing considerations
    Olga A. Gromova, Ivan Iu. Torshin, Tatiana R. Grishina, Svetlana I. Maliavskaia
    Gynecology.2020; 22(5): 61.     CrossRef
  • A Significant Association Between Rhein and Diabetic Nephropathy in Animals: A Systematic Review and Meta-Analysis
    Heng-Chang Hu, Liu-Tao Zheng, Hai-Yan Yin, Yuan Tao, Xiao-Qiong Luo, Kai-Shan Wei, Li-Ping Yin
    Frontiers in Pharmacology.2019;[Epub]     CrossRef
  • Dietary n-3 polyunsaturated fatty acids, fish intake and healthy ageing
    Esther García-Esquinas, Rosario Ortolá, Jose Ramón Banegas, Esther Lopez-García, Fernando Rodríguez-Artalejo
    International Journal of Epidemiology.2019; 48(6): 1914.     CrossRef
  • Beneficial Effects of Adiponectin on Glucose and Lipid Metabolism and Atherosclerotic Progression: Mechanisms and Perspectives
    Hidekatsu Yanai, Hiroshi Yoshida
    International Journal of Molecular Sciences.2019; 20(5): 1190.     CrossRef
  • Examining the Potential of Developing and Implementing Use of Adiponectin-Targeted Therapeutics for Metabolic and Cardiovascular Diseases
    Ying Liu, Vivian Vu, Gary Sweeney
    Frontiers in Endocrinology.2019;[Epub]     CrossRef
Original Articles
Obesity and Metabolic Syndrome
In Vitro Effect of Fatty Acids Identified in the Plasma of Obese Adolescents on the Function of Pancreatic β-Cells
Claudia Velasquez, Juan Sebastian Vasquez, Norman Balcazar
Diabetes Metab J. 2017;41(4):303-315.   Published online May 24, 2017
DOI: https://doi.org/10.4093/dmj.2017.41.4.303
  • 4,352 View
  • 39 Download
  • 8 Web of Science
  • 6 Crossref
AbstractAbstract PDFPubReader   
Background

The increase in circulating free fatty acid (FFA) levels is a major factor that induces malfunction in pancreatic β-cells. We evaluated the effect of FFAs reconstituted according to the profile of circulating fatty acids found in obese adolescents on the viability and function of the murine insulinoma cell line (mouse insulinoma [MIN6]).

Methods

From fatty acids obtained commercially, plasma-FFA profiles of three different youth populations were reconstituted: obese with metabolic syndrome; obese without metabolic syndrome; and normal weight without metabolic syndrome. MIN6 cells were treated for 24 or 48 hours with the three FFA profiles, and glucose-stimulated insulin secretion, cell viability, mitochondrial function and antioxidant activity were evaluated.

Results

The high FFA content and high polyunsaturated ω6/ω3 ratio, present in plasma of obese adolescents with metabolic syndrome had a toxic effect on MIN6 cell viability and function, increasing oxidative stress and decreasing glucose-dependent insulin secretion.

Conclusion

These results could help to guide nutritional management of obese young individuals, encouraging the increase of ω-3-rich food consumption in order to reduce the likelihood of deterioration of β-cells and the possible development of type 2 diabetes mellitus.

Citations

Citations to this article as recorded by  
  • The reversible effects of free fatty acids on sulfonylurea-stimulated insulin secretion are related to the expression and dynamin-mediated endocytosis of KATP channels in pancreatic β cells
    Chenmin Wei, Zichen Zhang, Qi Fu, Yunqiang He, Tao Yang, Min Sun
    Endocrine Connections.2023;[Epub]     CrossRef
  • Preparation of fatty acid solutions exerts significant impact on experimental outcomes in cell culture models of lipotoxicity
    Axel Römer, Divya Rawat, Thomas Linn, Sebastian F Petry
    Biology Methods and Protocols.2022;[Epub]     CrossRef
  • Lipotoxic Impairment of Mitochondrial Function in β-Cells: A Review
    Axel Römer, Thomas Linn, Sebastian F. Petry
    Antioxidants.2021; 10(2): 293.     CrossRef
  • Contribution of Oxidative Stress and Impaired Biogenesis of Pancreatic β-Cells to Type 2 Diabetes
    Petr Ježek, Martin Jabůrek, Lydie Plecitá-Hlavatá
    Antioxidants & Redox Signaling.2019; 31(10): 722.     CrossRef
  • The role of polyunsaturated fatty acids (n-3 PUFAs) on the pancreatic β-cells and insulin action
    Habtamu Wondifraw Baynes, Seifu Mideksa, Sintayehu Ambachew
    Adipocyte.2018; : 1.     CrossRef
  • Fatty Acid-Stimulated Insulin Secretion vs. Lipotoxicity
    Petr Ježek, Martin Jabůrek, Blanka Holendová, Lydie Plecitá-Hlavatá
    Molecules.2018; 23(6): 1483.     CrossRef
Obesity and Metabolic Syndrome
Statins Increase Mitochondrial and Peroxisomal Fatty Acid Oxidation in the Liver and Prevent Non-Alcoholic Steatohepatitis in Mice
Han-Sol Park, Jung Eun Jang, Myoung Seok Ko, Sung Hoon Woo, Bum Joong Kim, Hyun Sik Kim, Hye Sun Park, In-Sun Park, Eun Hee Koh, Ki-Up Lee
Diabetes Metab J. 2016;40(5):376-385.   Published online April 5, 2016
DOI: https://doi.org/10.4093/dmj.2016.40.5.376
  • 7,140 View
  • 90 Download
  • 67 Web of Science
  • 128 Crossref
AbstractAbstract PDFPubReader   
Background

Non-alcoholic fatty liver disease is the most common form of chronic liver disease in industrialized countries. Recent studies have highlighted the association between peroxisomal dysfunction and hepatic steatosis. Peroxisomes are intracellular organelles that contribute to several crucial metabolic processes, such as facilitation of mitochondrial fatty acid oxidation (FAO) and removal of reactive oxygen species through catalase or plasmalogen synthesis. Statins are known to prevent hepatic steatosis and non-alcoholic steatohepatitis (NASH), but underlying mechanisms of this prevention are largely unknown.

Methods

Seven-week-old C57BL/6J mice were given normal chow or a methionine- and choline-deficient diet (MCDD) with or without various statins, fluvastatin, pravastatin, simvastatin, atorvastatin, and rosuvastatin (15 mg/kg/day), for 6 weeks. Histological lesions were analyzed by grading and staging systems of NASH. We also measured mitochondrial and peroxisomal FAO in the liver.

Results

Statin treatment prevented the development of MCDD-induced NASH. Both steatosis and inflammation or fibrosis grades were significantly improved by statins compared with MCDD-fed mice. Gene expression levels of peroxisomal proliferator-activated receptor α (PPARα) were decreased by MCDD and recovered by statin treatment. MCDD-induced suppression of mitochondrial and peroxisomal FAO was restored by statins. Each statin's effect on increasing FAO and improving NASH was independent on its effect of decreasing cholesterol levels.

Conclusion

Statins prevented NASH and increased mitochondrial and peroxisomal FAO via induction of PPARα. The ability to increase hepatic FAO is likely the major determinant of NASH prevention by statins. Improvement of peroxisomal function by statins may contribute to the prevention of NASH.

Citations

Citations to this article as recorded by  
  • Organelle stress and alterations in interorganelle crosstalk during liver fibrosis
    Saloni Sinha, Nora Hassan, Robert E. Schwartz
    Hepatology.2024; 79(2): 482.     CrossRef
  • Statin therapy: a potential adjuvant to immunotherapies in hepatocellular carcinoma
    Jiao Wang, Chengyu Liu, Ronghua Hu, Licheng Wu, Chuanzhou Li
    Frontiers in Pharmacology.2024;[Epub]     CrossRef
  • Recent Progress in Anti‐Tumor Nanodrugs Based on Tumor Microenvironment Redox Regulation
    Lan Yao, Xiang Zhu, Yunyi Shan, Liang Zhang, Jing Yao, Hui Xiong
    Small.2024;[Epub]     CrossRef
  • Exploration of the Key Genes Involved in Non-alcoholic Fatty Liver Disease and Possible MicroRNA Therapeutic Targets
    Ali Mahmoudi, Amin Jalili, Alexandra E. Butler, Seyed H. Aghaee-Bakhtiari, Tannaz Jamialahmadi, Amirhossein Sahebkar
    Journal of Clinical and Experimental Hepatology.2024; 14(4): 101365.     CrossRef
  • Inflammation drives pathogenesis of early intestinal failure-associated liver disease
    Scott C. Fligor, Savas T. Tsikis, Thomas I. Hirsch, Ashish Jain, Liang Sun, Shira Rockowitz, Kathleen M. Gura, Mark Puder
    Scientific Reports.2024;[Epub]     CrossRef
  • A Systematic Review of Statins for the Treatment of Nonalcoholic Steatohepatitis: Safety, Efficacy, and Mechanism of Action
    Shiqin Zhang, Xiaoling Ren, Bingzheng Zhang, Tian Lan, Bing Liu
    Molecules.2024; 29(8): 1859.     CrossRef
  • Statins in Chronic Liver Disease: Review of the Literature and Future Role
    Nguyen Pham, Jihane N. Benhammou
    Seminars in Liver Disease.2024; 44(02): 191.     CrossRef
  • Team players in the pathogenesis of metabolic dysfunctions-associated steatotic liver disease: The basis of development of pharmacotherapy
    Shahid Habib
    World Journal of Gastrointestinal Pathophysiology.2024;[Epub]     CrossRef
  • Effects of metformin and simvastatin treatment on ultrastructural features of liver macrophages in HFD mice
    Darko Ciric, Tamara Kravic-Stevovic, Vladimir Bumbasirevic, Sasa Petricevic, Sofija Jovanovic, Vladimir Trajkovic, Tamara Martinovic
    Ultrastructural Pathology.2023; 47(1): 1.     CrossRef
  • Statins for the Treatment of Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis: A Systematic Review and Meta-Analysis
    Weiwei Dai, Baohong Xu, Peng Li, Junhua Weng
    American Journal of Therapeutics.2023; 30(1): e17.     CrossRef
  • Atorvastatin Attenuates Diet-Induced Non-Alcoholic Steatohepatitis in APOE*3-Leiden Mice by Reducing Hepatic Inflammation
    José A. Inia, Geurt Stokman, Elsbet J. Pieterman, Martine C. Morrison, Aswin L. Menke, Lars Verschuren, Martien P. M. Caspers, Martin Giera, J. Wouter Jukema, Anita M. van den Hoek, Hans M. G. Princen
    International Journal of Molecular Sciences.2023; 24(9): 7818.     CrossRef
  • The interplay between nonalcoholic fatty liver disease and atherosclerotic cardiovascular disease
    Alexandra C. Finney, Sandeep Das, Dhananjay Kumar, M. Peyton McKinney, Bishuang Cai, Arif Yurdagul, Oren Rom
    Frontiers in Cardiovascular Medicine.2023;[Epub]     CrossRef
  • Serum metabolomic signatures of fatty acid oxidation defects differentiate host-response subphenotypes of acute respiratory distress syndrome
    Tomeka L. Suber, Stacy G. Wendell, Steven J. Mullett, Benjamin Zuchelkowski, William Bain, Georgios D. Kitsios, Bryan J. McVerry, Prabir Ray, Anuradha Ray, Rama K. Mallampalli, Yingze Zhang, Faraaz Shah, Seyed Mehdi Nouraie, Janet S. Lee
    Respiratory Research.2023;[Epub]     CrossRef
  • Statins on nonalcoholic fatty liver disease: A systematic review and meta-analysis of 14 RCTs
    Haiyan Zhou, Maeda Toshiyoshi,, Wenli Zhao, Ye Zhao, Yan Zhao,
    Medicine.2023; 102(26): e33981.     CrossRef
  • Atorvastatin rescues hyperhomocysteinemia-induced cognitive deficits and neuroinflammatory gene changes
    Erica M. Weekman, Sherika N. Johnson, Colin B. Rogers, Tiffany L. Sudduth, Kevin Xie, Qi Qiao, David W. Fardo, Teodoro Bottiglieri, Donna M. Wilcock
    Journal of Neuroinflammation.2023;[Epub]     CrossRef
  • Crosstalk between Lipids and Non-Alcoholic Fatty Liver Disease
    Divyavani Gowda, Chandra Shekhar, Siddabasave Gowda B. Gowda, Yifan Chen, Shu-Ping Hui
    Livers.2023; 3(4): 687.     CrossRef
  • Empagliflozin: Potential Protective Effects on Hepatocytes and Liver Outcomes in Streptozotocin -Diabetic Rats.
    Alia Khwaldeh, Nour Al-Sawalha, Shefa' Aljabali, Ziad Shraideh, Sokiyna Ababneh, Roba Bdeir
    Biomedical and Pharmacology Journal.2023; 16(4): 2123.     CrossRef
  • Is there a role of lipid-lowering therapies in the management of fatty liver disease?
    Ismini Tzanaki, Aris P Agouridis, Michael S Kostapanos
    World Journal of Hepatology.2022; 14(1): 119.     CrossRef
  • Bifidobacterium animalis subsp. lactis A6 Enhances Fatty Acid β-Oxidation of Adipose Tissue to Ameliorate the Development of Obesity in Mice
    Yanxiong Huo, Guoping Zhao, Jinwang Li, Ran Wang, Fazheng Ren, Yixuan Li, Xiaoyu Wang
    Nutrients.2022; 14(3): 598.     CrossRef
  • PharmOmics: A species- and tissue-specific drug signature database and gene-network-based drug repositioning tool
    Yen-Wei Chen, Graciel Diamante, Jessica Ding, Thien Xuan Nghiem, Jessica Yang, Sung-Min Ha, Peter Cohn, Douglas Arneson, Montgomery Blencowe, Jennifer Garcia, Nima Zaghari, Paul Patel, Xia Yang
    iScience.2022; 25(4): 104052.     CrossRef
  • Pleyotropic Effects of Statins Non-Alcoholic Fat Disease of the Liver Non-Alcoholic Steatohepatitis
    И.Р. Агабабян, Ш.Ш. Садыкова
    Рецепт.2022; (2): 194.     CrossRef
  • PPAR Alpha as a Metabolic Modulator of the Liver: Role in the Pathogenesis of Nonalcoholic Steatohepatitis (NASH)
    Simona Todisco, Anna Santarsiero, Paolo Convertini, Giulio De Stefano, Michele Gilio, Vito Iacobazzi, Vittoria Infantino
    Biology.2022; 11(5): 792.     CrossRef
  • Association between organochlorine pesticides and nonalcoholic fatty liver disease in the National Health and Nutrition Examination Survey 2003–2004
    Hyunji Sang, Kyu-Na Lee, Chang Hee Jung, Kyungdo Han, Eun Hee Koh
    Scientific Reports.2022;[Epub]     CrossRef
  • Integrating the contributions of mitochondrial oxidative metabolism to lipotoxicity and inflammation in NAFLD pathogenesis
    Curtis C. Hughey, Patrycja Puchalska, Peter A. Crawford
    Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids.2022; 1867(11): 159209.     CrossRef
  • Supplementation of Lycium barbarum Polysaccharide Combined with Aerobic Exercise Ameliorates High-Fat-Induced Nonalcoholic Steatohepatitis via AMPK/PPARα/PGC-1α Pathway
    Dou-Dou Li, Jia-Min Ma, Ming-Jing Li, Lu-Lu Gao, Yan-Na Fan, Yan-Nan Zhang, Xiu-Juan Tao, Jian-Jun Yang
    Nutrients.2022; 14(15): 3247.     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
  • Regulation of Hepatic Lipid and Glucose Metabolism by INSP3R1
    Rachel J. Perry
    Diabetes.2022; 71(9): 1834.     CrossRef
  • Effects of SLCO1B1 Genetic Variant on Metabolite Profile in Participants on Simvastatin Treatment
    Lilian Fernandes Silva, Rowmika Ravi, Jagadish Vangipurapu, Anniina Oravilahti, Markku Laakso
    Metabolites.2022; 12(12): 1159.     CrossRef
  • Acute and chronic effects of environmental realistic concentrations of simvastatin in danio rerio: evidences of oxidative alterations and endocrine disruptive activity
    D. Rebelo, A.T. Correia, B. Nunes
    Environmental Toxicology and Pharmacology.2021; 81: 103522.     CrossRef
  • Allopurinol ameliorates high fructose diet induced hepatic steatosis in diabetic rats through modulation of lipid metabolism, inflammation, and ER stress pathway
    In-Jin Cho, Da-Hee Oh, Jin Yoo, You-Cheol Hwang, Kyu Jeung Ahn, Ho-Yeon Chung, Soung Won Jeong, Ju-Young Moon, Sang-Ho Lee, Sung-Jig Lim, In-Kyung Jeong
    Scientific Reports.2021;[Epub]     CrossRef
  • Atorvastatin Modulates Bile Acid Homeostasis in Mice with Diet-Induced Nonalcoholic Steatohepatitis
    Hana Lastuvkova, Fatemeh Alaei Faradonbeh, Jolana Schreiberova, Milos Hroch, Jaroslav Mokry, Hana Faistova, Zuzana Nova, Radomír Hyspler, Ivone Cristina Igreja Sa, Petr Nachtigal, Alzbeta Stefela, Petr Pavek, Stanislav Micuda
    International Journal of Molecular Sciences.2021; 22(12): 6468.     CrossRef
  • PPARs as Metabolic Sensors and Therapeutic Targets in Liver Diseases
    Hugo Christian Monroy-Ramirez, Marina Galicia-Moreno, Ana Sandoval-Rodriguez, Alejandra Meza-Rios, Arturo Santos, Juan Armendariz-Borunda
    International Journal of Molecular Sciences.2021; 22(15): 8298.     CrossRef
  • Simvastatin affects the PPARα signaling pathway and causes oxidative stress and embryonic development interference in Mugilogobius abei
    Chao Wang, Tianli Tang, Yimeng Wang, Xiangping Nie, Kaibin Li
    Aquatic Toxicology.2021; 239: 105951.     CrossRef
  • Impact of statin use on the risk and prognosis of hepatocellular carcinoma: a meta-analysis
    Jianfeng Wang, Xiaogang Li
    European Journal of Gastroenterology & Hepatology.2021; 33(12): 1603.     CrossRef
  • Statins in Non-alcoholic Steatohepatitis
    Jose D. Torres-Peña, Laura Martín-Piedra, Francisco Fuentes-Jiménez
    Frontiers in Cardiovascular Medicine.2021;[Epub]     CrossRef
  • Comparison of the liver findings after simvastatin-treatment between Spontaneously Diabetic Torii-Leprfa (SDT fatty) rats and Sprague-Dawley rats
    Tadakazu Takahashi, Yusuke Suzuki, Naohito Yamada, Kaoru Toyoda, Keisuke Goda, Katsunori Ryoke, Chizuru Matsuura, Akio Kobayashi, Shoichiro Sugai, Kayoko Shimoi
    Fundamental Toxicological Sciences.2020; 7(1): 41.     CrossRef
  • Current and emerging pharmacotherapeutic interventions for the treatment of liver fibrosis
    Joeri Lambrecht, Leo A. van Grunsven, Frank Tacke
    Expert Opinion on Pharmacotherapy.2020; 21(13): 1637.     CrossRef
  • The Influence of Statins on the Aerobic Metabolism of Endothelial Cells
    Izabela Broniarek, Karolina Dominiak, Lukasz Galganski, Wieslawa Jarmuszkiewicz
    International Journal of Molecular Sciences.2020; 21(4): 1485.     CrossRef
  • NADPH Oxidase Inhibition in Fibrotic Pathologies
    Karen Bernard, Victor J. Thannickal
    Antioxidants & Redox Signaling.2020; 33(6): 455.     CrossRef
  • Pleiotropic Effects of Statins in the Light of Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis
    Farah Ahsan, Federico Oliveri, Harshit K Goud, Zainab Mehkari, Lubna Mohammed, Moiz Javed, Aldanah Althwanay, Ian H Rutkofsky
    Cureus.2020;[Epub]     CrossRef
  • Peroxisomal footprint in the pathogenesis of nonalcoholic steatohepatitis
    S.M. Touhidul Islam, Jeseong Won, Mushfiquddin Khan, Kenneth D. Chavin, Inderjit Singh
    Annals of Hepatology.2020; 19(5): 466.     CrossRef
  • Structural Basis for Activation of Human Sirtuin 6 by Fluvastatin
    Weijie You, Clemens Steegborn
    ACS Medicinal Chemistry Letters.2020; 11(11): 2285.     CrossRef
  • Specificity of transaminase activities in the prediction of drug-induced hepatotoxicity
    Akio Kobayashi, Yusuke Suzuki, Shoichiro Sugai
    The Journal of Toxicological Sciences.2020; 45(9): 515.     CrossRef
  • Disease-associated gut microbiome and metabolome changes in patients with chronic obstructive pulmonary disease
    Kate L. Bowerman, Saima Firdous Rehman, Annalicia Vaughan, Nancy Lachner, Kurtis F. Budden, Richard Y. Kim, David L. A. Wood, Shaan L. Gellatly, Shakti D. Shukla, Lisa G. Wood, Ian A. Yang, Peter A. Wark, Philip Hugenholtz, Philip M. Hansbro
    Nature Communications.2020;[Epub]     CrossRef
  • Simvastatin Reduces Hepatic Oxidative Stress and Endoplasmic Reticulum Stress in Nonalcoholic Steatohepatitis Experimental Model
    Graziella Rodrigues, Andrea Janz Moreira, Silvia Bona, Elizângela Schemitt, Cláudio Augusto Marroni, Fábio Cangeri Di Naso, Alexandre Simões Dias, Thienne Rocha Pires, Jaqueline Nascimento Picada, Norma Possa Marroni
    Oxidative Medicine and Cellular Longevity.2019; 2019: 1.     CrossRef
  • Impaired Peroxisomal Fitness in Obese Mice, a Vicious Cycle Exacerbating Adipocyte Dysfunction via Oxidative Stress
    Lingjuan Piao, Debra Dorotea, Songling Jiang, Eun Hee Koh, Goo Taeg Oh, Hunjoo Ha
    Antioxidants & Redox Signaling.2019; 31(18): 1339.     CrossRef
  • (5R)-5-hydroxytriptolide ameliorates liver lipid accumulation by suppressing lipid synthesis and promoting lipid oxidation in mice
    Yunxia Dong, Henglei Lu, Qiang Li, Xinming Qi, Yuanchao Li, Zean Zhang, Jing Chen, Jin Ren
    Life Sciences.2019; 232: 116644.     CrossRef
  • Simvastatin alleviates bone resorption in apical periodontitis possibly by inhibition of mitophagy‐related osteoblast apoptosis
    C.‐N. Yang, S.‐H. Kok, H.‐W. Wang, J. Z.‐C. Chang, E. H.‐H. Lai, C.‐T. Shun, H. Yang, M.‐H. Chen, C.‐Y. Hong, S.‐K. Lin
    International Endodontic Journal.2019; 52(5): 676.     CrossRef
  • Different Effects of Pravastatin on Preeclampsia-like Symptoms in Different Mouse Models
    Jing Huai, Zi Yang, Yan-Hong Yi, Guang-Jiao Wang
    Chinese Medical Journal.2018; 131(4): 461.     CrossRef
  • Ameliorating effects of D-47, a newly developed compound, on lipid metabolism in an animal model of familial hypercholesterolemia (WHHLMI rabbits)
    Shohei Tamura, Yui Koike, Hiroaki Takeda, Tomonari Koike, Yoshihiro Izumi, Ryosuke Nagasaka, Tetsuto Tsunoda, Motoo Tori, Kazuo Ogawa, Takeshi Bamba, Masashi Shiomi
    European Journal of Pharmacology.2018; 822: 147.     CrossRef
  • Fluvastatin activates sirtuin 6 to regulate sterol regulatory element-binding proteins and AMP-activated protein kinase in HepG2 cells
    Ji-Hye Kim, Jun Mi Lee, Jong-Hoon Kim, Kwang Rok Kim
    Biochemical and Biophysical Research Communications.2018; 503(3): 1415.     CrossRef
  • Statin use and the risk of hepatocellular carcinoma in patients at high risk: A nationwide nested case-control study
    Gyuri Kim, Suk-Yong Jang, Chung Mo Nam, Eun Seok Kang
    Journal of Hepatology.2018; 68(3): 476.     CrossRef
  • Membrane Remodeling as a Key Player of the Hepatotoxicity Induced by Co-Exposure to Benzo[a]pyrene and Ethanol of Obese Zebrafish Larvae
    Muhammad Imran, Odile Sergent, Arnaud Tête, Isabelle Gallais, Martine Chevanne, Dominique Lagadic-Gossmann, Normand Podechard
    Biomolecules.2018; 8(2): 26.     CrossRef
  • Corticosterone-Induced Lipogenesis Activation and Lipophagy Inhibition in Chicken Liver Are Alleviated by Maternal Betaine Supplementation
    Yun Hu, Qinwei Sun, Yan Hu, Zhen Hou, Yibo Zong, Nagmeldin A Omer, Halima Abobaker, Ruqian Zhao
    The Journal of Nutrition.2018; 148(3): 316.     CrossRef
  • Simvastatin protects against acetaminophen-induced liver injury in mice
    Huan Liang, Yang Feng, Ruixia Cui, Minglong Qiu, Jingyao Zhang, Chang Liu
    Biomedicine & Pharmacotherapy.2018; 98: 916.     CrossRef
  • Effects of Pitavastatin on Insulin Sensitivity and Liver Fat: A Randomized Clinical Trial
    Laurie R Braun, Meghan N Feldpausch, Natalia Czerwonka, Julian Weiss, Karen Branch, Hang Lee, Edgar L Martinez-Salazar, Martin Torriani, Craig A Sponseller, Steven K Grinspoon, Takara L Stanley
    The Journal of Clinical Endocrinology & Metabolism.2018; 103(11): 4176.     CrossRef
  • Inactivation of SREBP-1a Phosphorylation Prevents Fatty Liver Disease in Mice: Identification of Related Signaling Pathways by Gene Expression Profiles in Liver and Proteomes of Peroxisomes
    Birgit Knebel, Sonja Hartwig, Sylvia Jacob, Ulrike Kettel, Martina Schiller, Waltraud Passlack, Cornelia Koellmer, Stefan Lehr, Dirk Müller-Wieland, Jorg Kotzka
    International Journal of Molecular Sciences.2018; 19(4): 980.     CrossRef
  • MicroRNA-124 Regulates Fatty Acid and Triglyceride Homeostasis
    Tyler A. Shaw, Ragunath Singaravelu, Megan H. Powdrill, Jordan Nhan, Nadine Ahmed, Dennis Özcelik, John Paul Pezacki
    iScience.2018; 10: 149.     CrossRef
  • Liver Disease in Singapore
    Mark Muthiah, Chern H Chong, Seng G Lim
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 66.     CrossRef
  • Accelerating Treatment of Skeletal Class II Malocclusion using Fixed Twin Block Appliances
    Snigdha Pattanaik, Navya Puvvula, Noorjahan Mohammad
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 146.     CrossRef
  • Effect of a Papain-based Chemomechanical Agent on Structure of Dentin and Bond Strength: Anin vitroStudy
    Veena S Pai, Yashwanth Gowda, Sruthi Nair
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 161.     CrossRef
  • Perception of Indian Dental Surgeons regarding Molar Incisor Hypomineralization
    Sumita Upadhyay, Jatinder K Dhillon
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 116.     CrossRef
  • Association between Obesity and Oral Health Status in Schoolchildren: A Survey in Five Districts of West Bengal, India
    Rahul Kaul, Paras Angrish, Subrata Saha, Sonali Halder, Bhaswar Bhattacharya, Malay Mitra
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 233.     CrossRef
  • Burden of Alcoholic Liver Disease: Bhutan Scenario
    Pelden Wangchuk
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 81.     CrossRef
  • Comparative Evaluation of Effects ofTriphala, Garlic Extracts, and Chlorhexidine Mouthwashes on SalivaryStreptococcus mutansCounts and Oral Hygiene Status
    Shweta Gupta, Narendra Padiyar, Bharathi Padiyar
    International Journal of Clinical Pediatric Dentistry.2018; 11(4): 299.     CrossRef
  • Evaluation of Dentin–Pulp Complex Response after Conservative Clinical Procedures in Primary Teeth
    Thais Marchini Oliveira, Bianca Mello, Tassia C Stafuzza, Luciana Vitor, Daniela Rios, Thiago Silva, Maria Machado
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 188.     CrossRef
  • Nonalcoholic Fatty Liver Disease: Time to Take the Bull by the Horns
    Preetam Nath, Shivaram P Singh
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 47.     CrossRef
  • F-18 Fluorodeoxyglucose Positron Emission Tomography/ Computed Tomography Findings of Isolated Gastric Tuberculosis mimicking Gastric Cancer and Lymphoma
    Remzi A Akdogan, Halil Rakici, Serkan Güngör, Recep Bedir, Elif Akdogan
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 93.     CrossRef
  • Clinical, Radiological, and Histological Assessment of Magnetic Nanoparticles as Pulpotomy Medicament in Primary Molars
    Manoj K Mallela, Harivinder R Konyala, Ajay R Mareddy, N Venugopal Reddy, Keerthi P Susheela
    International Journal of Clinical Pediatric Dentistry.2018; 11(4): 283.     CrossRef
  • Determination of ABO Blood Groups and Rh Typing from Dry Salivary Samples
    Laxmi Lakade, Priyam R Velani
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 100.     CrossRef
  • Orofacial Manifestations of Leukemic Children on Treatment: A Descriptive Study
    Keerthilatha M Pai, Aparna Aggarwal
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 193.     CrossRef
  • Mandibular Regional Odontodysplasia in an 8-year-old Boy showing Teeth Disorders, Gubernaculum Tracts, and Altered Bone Fractal Pattern
    Davi de Sá Cavalcante, Cristiane SR Fonteles, Thyciana R Ribeiro, Lúcio M Kurita, Alynne Vde M Pimenta, Francisco SR Carvalho, Fábio WG Costa
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 128.     CrossRef
  • Evaluation of Chemokines in the Gingival Crevicular Fluid of Children with Down Syndrome
    Harshini Togaru, Veerakishore Kumar Reddy, Naveen K Kommineni, Prathyusha Padakandla, John P Indupalli, Swapna P Nanga
    International Journal of Clinical Pediatric Dentistry.2018; 11(4): 288.     CrossRef
  • Hepatitis B Virus Infection among Health Care Workers in Indonesia
    David H Muljono, Teguh Wijayadi, Rizalinda Sjahril
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 88.     CrossRef
  • Comparison of the Effectiveness of Probiotic, Chlorhexidine-based Mouthwashes, and Oil Pulling Therapy on Plaque Accumulation and Gingival Inflammation in 10- to 12-year-old Schoolchildren: A Randomized Controlled Trial
    Saravana K Kandaswamy, Asokan Sharath, PR Geetha Priya
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 66.     CrossRef
  • Evaluation of Changes in Salivary pH after Intake of Different Eatables and Beverages in Children at Different Time Intervals
    Ankit Pachori, Haalaswamy Kambalimath, Garima Bhambhani, Garima Malhotra
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 177.     CrossRef
  • Customized Hybrid Bluegrass Appliance: An Innovative Technique
    Ziauddin Mohammad, Apeksha Bagalkotkar, Ashank Mishra, Gopi Veerala
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 141.     CrossRef
  • Postobstructive Cyst Formation in Pancreatic Duct affecting Surgical Approach
    Alper Parlakgumus, Ali Ezer
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 99.     CrossRef
  • Validating the Usage of Cariogram in 5- and 12-year-old School-going Children in Paonta Sahib, Himachal Pradesh, India: A 12-month Prospective Study
    Manish Madan, Pallav Singhal, Anu Garg, Akash Dupper
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 110.     CrossRef
  • Epidemiology of Chronic Hepatitis B in Turkey
    Hasan Ozkan
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 73.     CrossRef
  • A Comparative Evaluation of Time-dependent Changes on the Surface Hardness of Bulk Cure Composites: Anin vitroStudy
    Anindita Sarma
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 183.     CrossRef
  • Management of Hepatocellular Carcinoma: Bangladesh Perspective
    Mohammad Noor-E-Alam
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 52.     CrossRef
  • Trichobezoar: Ravenous for Hair
    Aman Kamra
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 97.     CrossRef
  • Comparative Evaluation of the Fracture Resistance of Two Different Fiber-reinforced Composite Restorations with Particulate Filler Composite Restorations
    Vanga V Narasimha Rao, Srinivas K Chandrabhatla, Vabbala R Rajasekhar
    International Journal of Clinical Pediatric Dentistry.2018; 11(4): 277.     CrossRef
  • A Comparative Evaluation of Efficacy ofStreptococcus mutansCounts in Saliva: Anin vivoStudy
    Inder K Pandit
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 94.     CrossRef
  • Liver Cancer in Nepal
    Ananta Shrestha
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 63.     CrossRef
  • Pre-eruptive Intracoronal Radiolucency in First Permanent Molar
    Mariana C Ilha, Paulo F Kramer, Simone H Ferreira, Henrique C Ruschel
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 151.     CrossRef
  • Comparative Evaluation of Various Temperature Changes on Stress Distribution in Class II Mesial-occlusal-distal Preparation restored with Different Restorative Materials: A Finite Element Analysis
    Binita Srivastava, Neorem N Devi
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 167.     CrossRef
  • A Survey on the Use of Antibiotics among the Dentists of Kolkata, West Bengal, India
    Rahul Kaul, Paras Angrish, Subrata Saha, Ashok V Sengupta, Shantanu Mukherjee
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 122.     CrossRef
  • Changing Etiology in Liver Cirrhosis in Sapporo, Japan
    Jong-Hon Kang, Takeshi Matsui
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 77.     CrossRef
  • Epidemiology, Genotype Distribution, Prognosis, Control, and Management of Viral Hepatitis B, C, D, and Hepatocellular Carcinoma in Mongolia
    Oidov Baatarkhuu, Tsagaantsooj Gerelchimeg, Dashchirev Munkh-Orshikh, Badamnachin Batsukh, Ganbold Sarangua, Jazag Amarsanaa
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 57.     CrossRef
  • Impact of Diabetes Mellitus Type 1 on Lebanese Families’ Quality of Life
    Nahla Nassif, Balsam Noueiri
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 61.     CrossRef
  • Pediatric Dental Appointments No-show: Rates and Reasons
    Anup Panda, Rupinder Bhatia, Esha C Vora
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 171.     CrossRef
  • A Case of Painless Excision
    Rupinder Bhatia, Ipshita A Suyash
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 135.     CrossRef
  • Manuka Honey: A Potent Cariostatic Agent–Anin vitroStudy
    Sapna Konde, Javaregowda P Beena, Punyatoya Sahoo, N Sunil Raj, Narayana C Kumar
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 105.     CrossRef
  • Reviewing of Research Finding of Hepatitis B Virus Infection in Lao People's Democratic Republic
    Angkham Ounavong
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 75.     CrossRef
  • Nonalcoholic Fatty Liver Disease: Identifying the Disease Burden in Sri Lanka
    Anuradha S Dassanayake
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 69.     CrossRef
  • Comparative Evaluation of Microhardness by Common Drinks on Esthetic Restorative Materials and Enamel: Anin vitroStudy
    Manish Madan, Akash Dupper, Roli Gupta, Trilok Kainthla
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 155.     CrossRef
  • Hepatocellular Carcinoma Surveillance: Benefit of Serum Alfa-fetoprotein in Real-world Practice
    Patharapan Lersritwimanmaen, Supot Nimanong
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 83.     CrossRef
  • Body Mass Index and Dental Caries: A Systematic Review

    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 228.     CrossRef
  • Evaluation of Surgical Options for Supernumerary Teeth in the Anterior Maxilla
    Gianluca Porcaro, Luca Mirabelli, Ernesto Amosso
    International Journal of Clinical Pediatric Dentistry.2018; 11(4): 294.     CrossRef
  • A Comparative Study to evaluate Parent's Ability to assess Dental Fear in their 6- to 10-year-old Children using Children's Fear Survey Schedule—Dental Subscale
    Ritika Malhotra, Anchal Sahni
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 205.     CrossRef
  • Estimation of Salivary Glucose, Calcium, Phosphorus, Alkaline Phosphatase, and Immunoglobulin A among Diabetic and Nondiabetic Children: A Case–Control Study
    Kalyani Uppu, Suzan Sahana, Ghanashyam P Madu, Aron AK Vasa, Sowjanya Nalluri
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 71.     CrossRef
  • Pedodontic Considerations in a Child with Attention Deficit Hyperactivity Disorder: Literature Review and a Case Report
    Siddhi Sinha, Prasanna Praveen, S Prathibha Rani, Athimuthu Anantharaj
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 254.     CrossRef
  • Salivary Cortisol and Alpha-amylase—Biomarkers of Stress in Children undergoing Extraction: Anin vivoStudy
    Neha Agarwal, Shefali Chaturvedy, Saurabh Chaturvedi, Yogita Chaturvedi
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 214.     CrossRef
  • Influence of Storage Media and Duration of Fragment in the Media on the Bond Strength of the Reattached Tooth Fragment
    Prashant Jalannavar
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 83.     CrossRef
  • Comparative Evaluation of Mucosal Vibrator with Topical Anesthetic Gel to reduce Pain during Administration of Local Anesthesia in Pediatric Patients: Anin vivoStudy
    Mahima Gandhi, Garima Kalia, Khushboo Rathore
    International Journal of Clinical Pediatric Dentistry.2018; 11(4): 261.     CrossRef
  • Management of Autistic Patients in Dental Office: A Clinical Update
    Jyothi S Bommangoudar
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 219.     CrossRef
  • Foreign Body causing Displacement of Immature Fractured Apical Root Fragment: An Unusual Case Report
    Aman Moda, Rajesh Singla, Preeti M Agrawal
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 247.     CrossRef
  • Validity and Reliability of the Hindi Version of the Modified Child Perceptions Questionnaire 11 to 14
    Mohit Sharma, Prasanna Kumar, Dempsy CM Mandanna
    International Journal of Clinical Pediatric Dentistry.2018; 11(4): 271.     CrossRef
  • Nonsyndromic Gingival Fibromatosis: A Rare Case Report
    Mahima Gandhi, Akshat Vijay
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 250.     CrossRef
  • Prevalence of Deleterious Oral Habits among 3- to 5-yearold Preschool Children in Bhubaneswar, Odisha, India
    Brahmananda Dutta, Tulika Verma
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 210.     CrossRef
  • Evaluation of Antimicrobial Activity of Two Endodontic Sealers: Zinc Oxide with Thyme Oil and Zinc Oxide Eugenol against Root Canal Microorganisms—Anin vitroStudy
    Manoj Chandak, Nilima Thosar, Silpi Basak
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 79.     CrossRef
  • Prevalence and Risk Factors for Dental Caries among Preschool Children: A Cross-sectional Study in Eastern India
    Vinay K Chugh, Kushal K Sahu, Ankita Chugh
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 238.     CrossRef
  • Squamous Papilloma on Hard Palate: Case Report and Literature Review
    Penmatsa Chaitanya, Satyam Martha, Ramachandran Punithvathy, Madhusudhan Reddy
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 244.     CrossRef
  • Comparison of Vitamin D Level of Children with Severe Early Childhood Caries and Children with No Caries
    Anchal Chhonkar, Vishal Arya
    International Journal of Clinical Pediatric Dentistry.2018; 11(3): 199.     CrossRef
  • Use of “Surface Analyzer” to evaluate the Effect of Two Polishing Systems on Surface Texture of Four Newer Composites
    Shefally Garg, Munish Goel, Shweta Verma, Nanika Mahajan, Bhawna Kaul, Vikas Garg
    International Journal of Clinical Pediatric Dentistry.2018; 11(4): 266.     CrossRef
  • Clinical Evaluation of Preventive Effect of Fissure Sealants on Initial Carious Lesion of Permanent Mandibular Molars Pretreated with and without Fluoride Varnish by Fluorescence Camera
    Madhagudanahalli S Lakshmi, Kudlapur T Srilatha, Bhojraj Nandlal, Seema Deshmukh
    International Journal of Clinical Pediatric Dentistry.2018; 11(2): 89.     CrossRef
  • Hepatocellular Carcinoma in Malaysia and Its Changing Trend
    Rosmawati Mohamed, Ruksana Raihan, Amirah Azzeri, Fatiha H Shabaruddin
    Euroasian Journal of Hepato-Gastroenterology.2018; 8(1): 54.     CrossRef
  • Advances in the Understanding and Treatment of Mitochondrial Fatty Acid Oxidation Disorders
    Eric S. Goetzman
    Current Genetic Medicine Reports.2017; 5(3): 132.     CrossRef
  • Hepatokines: linking nonalcoholic fatty liver disease and insulin resistance
    Ruth C. R. Meex, Matthew J. Watt
    Nature Reviews Endocrinology.2017; 13(9): 509.     CrossRef
  • In ovo injection of betaine alleviates corticosterone-induced fatty liver in chickens through epigenetic modifications
    Yun Hu, Qinwei Sun, Jie Liu, Yimin Jia, Demin Cai, Abdulrahman A. Idriss, Nagmeldin A. Omer, Ruqian Zhao
    Scientific Reports.2017;[Epub]     CrossRef
  • The therapeutic effect of silymarin in the treatment of nonalcoholic fatty disease
    Sheng Zhong, Yuxiang Fan, Qi Yan, Xingyu Fan, Bo Wu, Yujuan Han, Ying Zhang, Yong Chen, Huimao Zhang, Junqi Niu
    Medicine.2017; 96(49): e9061.     CrossRef
  • Atorvastatin reduces lipid accumulation in the liver by activating protein kinase A-mediated phosphorylation of perilipin 5
    Xing Gao, Yang Nan, Yuanlin Zhao, Yuan Yuan, Bincheng Ren, Chao Sun, Kaiyu Cao, Ming Yu, Xuyang Feng, Jing Ye
    Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids.2017; 1862(12): 1512.     CrossRef
  • Aerobic capacity mediates susceptibility for the transition from steatosis to steatohepatitis
    E. Matthew Morris, Colin S. McCoin, Julie A. Allen, Michelle L. Gastecki, Lauren G. Koch, Steven L. Britton, Justin A. Fletcher, Xiarong Fu, Wen‐Xing Ding, Shawn C. Burgess, R. Scott Rector, John P. Thyfault
    The Journal of Physiology.2017; 595(14): 4909.     CrossRef
  • Use of Statins in Patients with Chronic Liver Disease and Cirrhosis: Current Views and Prospects
    Jose Ignacio Vargas, Marco Arrese, Vijay H. Shah, Juan Pablo Arab
    Current Gastroenterology Reports.2017;[Epub]     CrossRef
  • Endogenous catalase delays high-fat diet-induced liver injury in mice
    Lingjuan Piao, Jiyeon Choi, Guideock Kwon, Hunjoo Ha
    The Korean Journal of Physiology & Pharmacology.2017; 21(3): 317.     CrossRef
  • Clinical implications of understanding the association between oxidative stress and pediatric NAFLD
    Jake P. Mann, Massimiliano Raponi, Valerio Nobili
    Expert Review of Gastroenterology & Hepatology.2017; 11(4): 371.     CrossRef
Brief Report
Beneficial Effects of Omega-3 Fatty Acids on Low Density Lipoprotein Particle Size in Patients with Type 2 Diabetes Already under Statin Therapy
Myung Won Lee, Jeong Kyung Park, Jae Won Hong, Kwang Joon Kim, Dong Yeob Shin, Chul Woo Ahn, Young Duk Song, Hong Keun Cho, Seok Won Park, Eun Jig Lee
Diabetes Metab J. 2013;37(3):207-211.   Published online June 14, 2013
DOI: https://doi.org/10.4093/dmj.2013.37.3.207
  • 4,681 View
  • 47 Download
  • 23 Crossref
AbstractAbstract PDFPubReader   

Beyond statin therapy for reducing low density lipoprotein cholesterol (LDL-C), additional therapeutic strategies are required to achieve more optimal reduction in cardiovascular risk among diabetic patients with dyslipidemia. To evaluate the effects and the safety of combined treatment with omega-3 fatty acids and statin in dyslipidemic patients with type 2 diabetes, we conducted a randomized, open-label study in Korea. Patients with persistent hypertriglyceridemia (≥200 mg/dL) while taking statin for at least 6 weeks were eligible. Fifty-one patients were randomized to receive either omega-3 fatty acid 4, 2 g, or no drug for 8 weeks while continuing statin therapy. After 8 weeks of treatment, the mean percentage change of low density lipoprotein (LDL) particle size and triglyceride (TG) level was greater in patients who were prescribed 4 g of omega-3 fatty acid with statin than in patients receiving statin monotherapy (2.8%±3.1% vs. 2.3%±3.6%, P=0.024; -41.0%±24.1% vs. -24.2%±31.9%, P=0.049). Coadministration of omega-3 fatty acids with statin increased LDL particle size and decreased TG level in dyslipidemic patients with type 2 diabetes. The therapy was well tolerated without significant adverse effects.

Citations

Citations to this article as recorded by  
  • Diabetic cardiac autonomic neuropathy: insulin resistance, lipid profile, and omega-3 polyunsaturated fatty acids
    Martin-Yurii Markevich, Volodymyr Segin, Victoria Serhiyenko, Alexandr Serhiyenko
    InterConf.2023; (35(163)): 213.     CrossRef
  • Atherogenic features of the fatty acid profile of erythrocyte membranes of patients with fatty liver disease of mixed genesis
    M. V. Kruchinina, A. V. Belkovets, M. V. Parulikova, A. A. Gromov
    Ateroscleroz.2023; 19(4): 350.     CrossRef
  • Omega-3 supplementation in the treatment of polycystic ovary syndrome (PCOS) – a review of clinical trials and cohort
    Vitoria Melo, Thomas Silva, Thaissa Silva, Juliana Freitas, Joselita Sacramento, Mirian Vazquez, Edilene Araujo
    Endocrine Regulations.2022; 56(1): 66.     CrossRef
  • Nutrigenetics, omega-3 and plasma lipids/lipoproteins/apolipoproteins with evidence evaluation using the GRADE approach: a systematic review
    Justine Keathley, Véronique Garneau, Valérie Marcil, David M Mutch, Julie Robitaille, Iwona Rudkowska, Gabriela Magdalena Sofian, Sophie Desroches, Marie-Claude Vohl
    BMJ Open.2022; 12(2): e054417.     CrossRef
  • N-3 fatty acid supplementation mediates lipid profile, including small dense LDL, when combined with statins: a randomized double blind placebo controlled trial
    Gediz Dogay Us, Sohail Mushtaq
    Lipids in Health and Disease.2022;[Epub]     CrossRef
  • The effect of omega-3 fatty acids and its combination with statins on lipid profile in patients with hypertriglyceridemia: A systematic review and meta-analysis of randomized controlled trials
    Yunjiao Yang, Wen Deng, Yanmei Wang, Tongyi Li, Yiding Chen, Cong Long, Qing Wen, Yue Wu, Qiu Chen
    Frontiers in Nutrition.2022;[Epub]     CrossRef
  • Study of the Healthy Effects of Different Fat Ratios Mixtures of Omega-3 to Omega-6 in Male Mice with Alloxan-Induced Diabetes
    Ali. M. Atallah, Faryal. F. Hussein
    Tikrit Journal for Agricultural Sciences.2021; 21(4): 129.     CrossRef
  • Omega-3 Fatty Acids as Druggable Therapeutics for Neurodegenerative Disorders
    Neha M. Chitre, Nader H. Moniri, Kevin S. Murnane
    CNS & Neurological Disorders - Drug Targets.2020; 18(10): 735.     CrossRef
  • Efficacy and Safety of Omega-3 Fatty Acids in Patients Treated with Statins for Residual Hypertriglyceridemia: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial
    Ji Eun Jun, In-Kyung Jeong, Jae Myung Yu, Sung Rae Kim, In Kye Lee, Kyung-Ah Han, Sung Hee Choi, Soo-Kyung Kim, Hyeong Kyu Park, Ji-Oh Mok, Yong-ho Lee, Hyuk-Sang Kwon, So Hun Kim, Ho-Cheol Kang, Sang Ah Lee, Chang Beom Lee, Kyung Mook Choi, Sung-Ho Her,
    Diabetes & Metabolism Journal.2020; 44(1): 78.     CrossRef
  • The combination of canagliflozin and omega-3 fatty acid ameliorates insulin resistance and cardiac biomarkersviamodulation of inflammatory cytokines in type 2 diabetic rats
    Mohammed Mohsen Safhi, Tarique Anwer, Gyas Khan, Rahimullah Siddiqui, Sivagurunathan Moni Sivakumar, Mohammad Firoz Alam
    The Korean Journal of Physiology & Pharmacology.2018; 22(5): 493.     CrossRef
  • Effect of diets rich in either saturated fat or n-6 polyunsaturated fatty acids and supplemented with long-chain n-3 polyunsaturated fatty acids on plasma lipoprotein profiles
    C B Dias, N Amigo, L G Wood, X Correig, M L Garg
    European Journal of Clinical Nutrition.2017; 71(11): 1297.     CrossRef
  • Effects of 12-week supplementation of marine Omega-3 PUFA-based formulation Omega3Q10 in older adults with prehypertension and/or elevated blood cholesterol
    Tian Shen, Guoqiang Xing, Jingfen Zhu, Shuxian Zhang, Yong Cai, Donghua Li, Gang Xu, Evan Xing, Jianyu Rao, Rong Shi
    Lipids in Health and Disease.2017;[Epub]     CrossRef
  • Effects of dietary saturated and n-6 polyunsaturated fatty acids on the incorporation of long-chain n-3 polyunsaturated fatty acids into blood lipids
    C B Dias, L G Wood, M L Garg
    European Journal of Clinical Nutrition.2016; 70(7): 812.     CrossRef
  • Comparative analysis of the efficacy of omega-3 fatty acids for hypertriglyceridaemia management in Korea
    H.-S. Kim, H. Kim, Y. J. Jeong, S. J. Yang, S. J. Baik, H. Lee, S.-H. Lee, J. H. Cho, I.-Y. Choi, H. W. Yim, K.-H. Yoon
    Journal of Clinical Pharmacy and Therapeutics.2016; 41(5): 508.     CrossRef
  • Effects of Omega-3 Fatty Acid Supplementation on Diabetic Nephropathy Progression in Patients with Diabetes and Hypertriglyceridemia
    Eugene Han, Yujung Yun, Gyuri Kim, Yong-ho Lee, Hye Jin Wang, Byung-Wan Lee, Bong Soo Cha, Beom Seok Kim, Eun Seok Kang, Wolf-Hagen Schunck
    PLOS ONE.2016; 11(5): e0154683.     CrossRef
  • The clinical relevance of omega-3 fatty acids in the management of hypertriglyceridemia
    James Backes, Deborah Anzalone, Daniel Hilleman, Julia Catini
    Lipids in Health and Disease.2016;[Epub]     CrossRef
  • Supercritical fluid extraction of grape seeds: extract chemical composition, antioxidant activity and inhibition of nitrite production in LPS-stimulated Raw 264.7 cells
    Concepción Pérez, María Luisa Ruiz del Castillo, Carmen Gil, Gracia Patricia Blanch, Gema Flores
    Food & Function.2015; 6(8): 2607.     CrossRef
  • Omega-3 Polyunsaturated Fatty Acids May Attenuate Streptozotocin-Induced Pancreatic β-Cell Death via Autophagy Activation in Fat1 Transgenic Mice
    Won-Min Hwang, Dong-Ho Bak, Dong Ho Kim, Ju Young Hong, Seung-Yun Han, Keun-Young Park, Kyu Lim, Dong-Mee Lim, Jae Gu Kang
    Endocrinology and Metabolism.2015; 30(4): 569.     CrossRef
  • Long-chain omega-3 fatty acids, fibrates and niacin as therapeutic options in the treatment of hypertriglyceridemia: A review of the literature
    Matthew K. Ito
    Atherosclerosis.2015; 242(2): 647.     CrossRef
  • Nutraceuticals and dyslipidaemia: Beyond the common therapeutics
    Pietro Scicchitano, Matteo Cameli, Maria Maiello, Pietro Amedeo Modesti, Maria Lorenza Muiesan, Salvatore Novo, Pasquale Palmiero, Pier Sergio Saba, Roberto Pedrinelli, Marco Matteo Ciccone
    Journal of Functional Foods.2014; 6: 11.     CrossRef
  • The effect of dietary omega-3 polyunsaturated fatty acids on plasma lipids and lipoproteins of C57BL/6 mice is age and sex specific
    K.A. Balogun, R.S. Randunu, S.K. Cheema
    Prostaglandins, Leukotrienes and Essential Fatty Acids.2014; 91(1-2): 39.     CrossRef
  • Gene-diet interactions with polymorphisms of the MGLL gene on plasma low-density lipoprotein cholesterol and size following an omega-3 polyunsaturated fatty acid supplementation: a clinical trial
    Catherine Ouellette, Iwona Rudkowska, Simone Lemieux, Benoit Lamarche, Patrick Couture, Marie-Claude Vohl
    Lipids in Health and Disease.2014;[Epub]     CrossRef
  • Saturated fat consumption may not be the main cause of increased blood lipid levels
    C.B. Dias, R. Garg, L.G. Wood, M.L. Garg
    Medical Hypotheses.2014; 82(2): 187.     CrossRef
Review
GLP-1 Receptor Agonist and Non-Alcoholic Fatty Liver Disease
Jinmi Lee, Seok-Woo Hong, Eun-Jung Rhee, Won-Young Lee
Diabetes Metab J. 2012;36(4):262-267.   Published online August 20, 2012
DOI: https://doi.org/10.4093/dmj.2012.36.4.262
  • 6,533 View
  • 86 Download
  • 64 Crossref
AbstractAbstract PDFPubReader   

Non-alcoholic fatty liver disease (NAFLD), one of the most common liver diseases, is caused by the disruption of hepatic lipid homeostasis. It is associated with insulin resistance as seen in type 2 diabetes mellitus. Glucagon-like peptide-1 (GLP-1) is an incretin that increases insulin sensitivity and aids glucose metabolism. In recent in vivo and in vitro studies, GLP-1 presents a novel therapeutic approach against NAFLD by increasing fatty acid oxidation, decreasing lipogenesis, and improving hepatic glucose metabolism. In this report, we provide an overview of the role and mechanism of GLP-1 in relieving NAFLD.

Citations

Citations to this article as recorded by  
  • Hepatic function of glucagon-like peptide-1 and its based diabetes drugs
    Jia Nuo Feng, Tianru Jin
    Medical Review.2024; 4(4): 312.     CrossRef
  • Non-alcoholic fatty liver disease in type 2 diabetes: Emerging evidence of benefit of peroxisome proliferator-activated receptors agonists and incretin-based therapies
    Subhodip Pramanik, Partha Pal, Sayantan Ray
    World Journal of Methodology.2024;[Epub]     CrossRef
  • Tirzepatide against obesity and insulin-resistance: pathophysiological aspects and clinical evidence
    Salvatore Corrao, Chiara Pollicino, Dalila Maggio, Alessandra Torres, Christiano Argano
    Frontiers in Endocrinology.2024;[Epub]     CrossRef
  • Assessing the therapeutic potential of long-chain isomaltooligosaccharides in diabetic and hyperlipidemic rats
    Onrapak Reamtong, Rattiya Waeonukul, Pattaneeya Prangthip
    Diabetology & Metabolic Syndrome.2024;[Epub]     CrossRef
  • Deciphering the Gut–Liver Axis: A Comprehensive Scientific Review of Non-Alcoholic Fatty Liver Disease
    Samradhi Singh, Mona Kriti, Roberto Catanzaro, Francesco Marotta, Mustafa Malvi, Ajay Jain, Vinod Verma, Ravinder Nagpal, Rajnarayan Tiwari, Manoj Kumar
    Livers.2024; 4(3): 435.     CrossRef
  • Second Generation Anti-Obesity Medications
    Sri Nikhita Chimatapu, Steven D. Mittelman, Vibha Singhal
    Current Pediatrics Reports.2024;[Epub]     CrossRef
  • The intestine as an endocrine organ and the role of gut hormones in metabolic regulation
    Rula Bany Bakar, Frank Reimann, Fiona M. Gribble
    Nature Reviews Gastroenterology & Hepatology.2023; 20(12): 784.     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
  • From Non-Alcoholic Fatty Liver Disease to Liver Cancer: Microbiota and Inflammation as Key Players
    Avilene Rodríguez-Lara, Ascensión Rueda-Robles, María José Sáez-Lara, Julio Plaza-Diaz, Ana I. Álvarez-Mercado
    Pathogens.2023; 12(7): 940.     CrossRef
  • Investigating the Opposing Effect of Two Different Green Tea Supplements on Oxidative Stress, Mitochondrial Function and Cell Viability in HepG2 Cells
    Aparna Shil, Chris Davies, Lata Gautam, Justin Roberts, Havovi Chichger
    Journal of Dietary Supplements.2022; 19(4): 459.     CrossRef
  • Antiobesity therapeutics with complementary dual‐agonist activities at glucagon and glucagon‐like peptide 1 receptors
    Bong Gyu Park, Gyeong Min Kim, Hye‐Jin Lee, Jae Ha Ryu, Dong‐Hoon Kim, Jae‐Young Seong, Soojeong Kim, Zee‐Yong Park, Young‐Joon Kim, Jaemin Lee, Jae Il Kim
    Diabetes, Obesity and Metabolism.2022; 24(1): 50.     CrossRef
  • The anti-inflammatory feature of glucagon-like peptide-1 and its based diabetes drugs—Therapeutic potential exploration in lung injury
    Juan Pang, Jia Nuo Feng, Wenhua Ling, Tianru Jin
    Acta Pharmaceutica Sinica B.2022; 12(11): 4040.     CrossRef
  • Deficiency of peroxisomal NUDT7 stimulates de novo lipogenesis in hepatocytes
    Jinsoo Song, In-Jeoung Baek, Sujeong Park, Jinjoo Oh, Deokha Kim, Kyung Song, Mi Kyung Kim, Hye Won Lee, Byoung Kuk Jang, Eun-Jung Jin
    iScience.2022; 25(10): 105135.     CrossRef
  • Gut–Liver Axis and Non-Alcoholic Fatty Liver Disease: A Vicious Circle of Dysfunctions Orchestrated by the Gut Microbiome
    Salvatore Pezzino, Maria Sofia, Gloria Faletra, Chiara Mazzone, Giorgia Litrico, Gaetano La Greca, Saverio Latteri
    Biology.2022; 11(11): 1622.     CrossRef
  • Efficacy and safety of semaglutide for weight management: evidence from the STEP program
    Anastassia Amaro, Danny Sugimoto, Sean Wharton
    Postgraduate Medicine.2022; 134(sup1): 5.     CrossRef
  • Incretin Hormones in Obesity and Related Cardiometabolic Disorders: The Clinical Perspective
    Joanna Michałowska, Ewa Miller-Kasprzak, Paweł Bogdański
    Nutrients.2021; 13(2): 351.     CrossRef
  • Insights into the Impact of Microbiota in the Treatment of NAFLD/NASH and Its Potential as a Biomarker for Prognosis and Diagnosis
    Julio Plaza-Díaz, Patricio Solis-Urra, Jerónimo Aragón-Vela, Fernando Rodríguez-Rodríguez, Jorge Olivares-Arancibia, Ana I. Álvarez-Mercado
    Biomedicines.2021; 9(2): 145.     CrossRef
  • A comprehensive review for gut microbes: technologies, interventions, metabolites and diseases
    Changlu Qi, Ping Wang, Tongze Fu, Minke Lu, Yiting Cai, Xu Chen, Liang Cheng
    Briefings in Functional Genomics.2021; 20(1): 42.     CrossRef
  • Update on the effects of GLP-1 receptor agonists for the treatment of polycystic ovary syndrome
    Maka Siamashvili, Stephen N. Davis
    Expert Review of Clinical Pharmacology.2021; 14(9): 1081.     CrossRef
  • Mechanistic and physiological approaches of fecal microbiota transplantation in the management of NAFLD
    Manisha Gupta, Pawan Krishan, Amarjot Kaur, Sandeep Arora, Nirupma Trehanpati, Thakur Gurjeet Singh, Onkar Bedi
    Inflammation Research.2021; 70(7): 765.     CrossRef
  • Retrospective analysis (2009–2017) of factors associated with progression and regression of non-alcoholic fatty liver disease (Hepatic steatosis) in patients with type 2 diabetes seen at a tertiary diabetes centre in Southern India
    Nithyanantham Kamalraj, Madhanagopal Sathishkumar, Mani Arunvignesh, Viswanathan Baskar, Saravanan Jebarani, Anandakumar Amutha, Mohan Deepa, Coimbatore Subramanyam Shanthi Rani, Sundaramoorthy Chandru, Ranjit Unnikrishnan, Ranjit Mohan Anjana, Mardavada
    Diabetes & Metabolic Syndrome: Clinical Research & Reviews.2021; 15(5): 102261.     CrossRef
  • Gastrointestinal Hormones in Healthy Adults: Reliability of Repeated Assessments and Interrelations with Eating Habits and Physical Activity
    Silke M. Wortha, Katharina A. Wüsten, Veronica A. Witte, Nicole Bössel, Wolfram Keßler, Antje Vogelgesang, Agnes Flöel
    Nutrients.2021; 13(11): 3809.     CrossRef
  • Role of ATP-binding cassette transporter A1 in suppressing lipid accumulation by glucagon-like peptide-1 agonist in hepatocytes
    Jingya Lyu, Hitomi Imachi, Kensaku Fukunaga, Seisuke Sato, Toshihiro Kobayashi, Tao Dong, Takanobu Saheki, Mari Matsumoto, Hisakazu Iwama, Huanxiang Zhang, Koji Murao
    Molecular Metabolism.2020; 34: 16.     CrossRef
  • Effects of synbiotic consumption on lipid profile: a systematic review and meta-analysis of randomized controlled clinical trials
    Amir Hadi, Ehsan Ghaedi, Saman Khalesi, Makan Pourmasoumi, Arman Arab
    European Journal of Nutrition.2020; 59(7): 2857.     CrossRef
  • Glucagon‐like peptide‐1 receptor agonists (GLP‐1 RAs) for the management of nonalcoholic fatty liver disease (NAFLD): A systematic review
    Xiaodan Lv, Yongqiang Dong, Lingling Hu, Feiyu Lu, Changyu Zhou, Shaoyou Qin
    Endocrinology, Diabetes & Metabolism.2020;[Epub]     CrossRef
  • Genetic engineering of novel super long-acting Exendin-4 chimeric protein for effective treatment of metabolic and cognitive complications of obesity
    Jong Youl Lee, Taehoon Park, Eunmi Hong, Reeju Amatya, Kyung-Ah Park, Young-Hoon Park, Kyoung Ah Min, Minki Jin, Sumi Lee, Seungmi Hwang, Gu Seob Roh, Meong Cheol Shin
    Biomaterials.2020; 257: 120250.     CrossRef
  • A look to the future in non‐alcoholic fatty liver disease: Are glucagon‐like peptide‐1 analogues or sodium‐glucose co‐transporter‐2 inhibitors the answer?
    Rebecca K. Vincent, David M. Williams, Marc Evans
    Diabetes, Obesity and Metabolism.2020; 22(12): 2227.     CrossRef
  • A systematic review and meta-analysis of probiotic consumption and metabolic status of athletes
    Atefeh As’Habi, Maryam Nazari, Hossein Hajianfar, Arman Arab, Zeinab Faghfoori
    International Journal of Food Properties.2020; 23(1): 941.     CrossRef
  • The Gut Barrier, Intestinal Microbiota, and Liver Disease: Molecular Mechanisms and Strategies to Manage
    Julio Plaza-Díaz, Patricio Solís-Urra, Fernando Rodríguez-Rodríguez, Jorge Olivares-Arancibia, Miguel Navarro-Oliveros, Francisco Abadía-Molina, Ana I. Álvarez-Mercado
    International Journal of Molecular Sciences.2020; 21(21): 8351.     CrossRef
  • Pathophysiology of NAFLD and NASH in Experimental Models: The Role of Food Intake Regulating Peptides
    L. Kořínková, V. Pražienková, L. Černá, A. Karnošová, B. Železná, J. Kuneš, Lenka Maletínská
    Frontiers in Endocrinology.2020;[Epub]     CrossRef
  • Have a heart: failure to increase GLP-1 caused by heart failure increases the risk of diabetes
    Michael J. Ryan
    Clinical Science.2020; 134(23): 3119.     CrossRef
  • Unraveling the Role of Leptin in Liver Function and Its Relationship with Liver Diseases
    Maite Martínez-Uña, Yaiza López-Mancheño, Carlos Diéguez, Manuel A. Fernández-Rojo, Marta G. Novelle
    International Journal of Molecular Sciences.2020; 21(24): 9368.     CrossRef
  • The Role of GLP1 in Rat Steatotic and Non-Steatotic Liver Transplantation from Cardiocirculatory Death Donors
    Cindy G. Avalos-de León, Mónica B. Jiménez-Castro, María Eugenia Cornide-Petronio, Araní Casillas-Ramírez, Carmen Peralta
    Cells.2019; 8(12): 1599.     CrossRef
  • LRG ameliorates steatohepatitis by activating the AMPK/mTOR/SREBP1 signaling pathway in C57BL/6J mice fed a high‑fat diet
    Tao Hao, Hongying Chen, Sisi Wu, Haoming Tian
    Molecular Medicine Reports.2019;[Epub]     CrossRef
  • Liraglutide alters hepatic metabolism in high-fat fed obese mice: A bioinformatic prediction and functional analysis
    Isabelle Arruda Barbosa, Eloá Mangabeira Santos, Alanna Fernandes Paraíso, Pablo Vinicyus Ferreira Chagas, Luís Paulo Oliveira, João Marcus Oliveira Andrade, Lucyana Conceição Farias, Bruna Mara Aparecida de Carvalho, Alfredo Maurício Batista de Paula, An
    Meta Gene.2019; 20: 100553.     CrossRef
  • Dipeptidyl peptidase‐4 inhibitors and aerobic exercise synergistically protect against liver injury in ovariectomized rats
    Nagat Younan, Samah Elattar, Mira Farouk, Laila Rashed, Suzanne Estaphan
    Physiological Reports.2019;[Epub]     CrossRef
  • Microbial Metabolites Determine Host Health and the Status of Some Diseases
    Panida Sittipo, Jae-won Shim, Yun Lee
    International Journal of Molecular Sciences.2019; 20(21): 5296.     CrossRef
  • Compound K attenuates glucose intolerance and hepatic steatosis through AMPK-dependent pathways in type 2 diabetic OLETF rats
    Yoo-Cheol Hwang, Da-Hee Oh, Moon Chan Choi, Sang Yeoul Lee, Kyu-Jeong Ahn, Ho-Yeon Chung, Sung-Jig Lim, Sung Hyun Chung, In-Kyung Jeong
    The Korean Journal of Internal Medicine.2018; 33(2): 347.     CrossRef
  • Liraglutide attenuates partial warm ischemia-reperfusion injury in rat livers
    Ahmed A. Abdelsameea, Noha A.T. Abbas, Samar M. Abdel Raouf
    Naunyn-Schmiedeberg's Archives of Pharmacology.2017; 390(3): 311.     CrossRef
  • Natural alkaloid bouchardatine ameliorates metabolic disorders in high‐fat diet‐fed mice by stimulating the sirtuin 1/liver kinase B‐1/AMPK axis
    Yong Rao, Hong Yu, Lin Gao, Yu‐Ting Lu, Zhao Xu, Hong Liu, Lian‐Quan Gu, Ji‐Ming Ye, Zhi‐Shu Huang
    British Journal of Pharmacology.2017; 174(15): 2457.     CrossRef
  • Non-alcoholic fatty liver disease and dyslipidemia: An update
    Niki Katsiki, Dimitri P. Mikhailidis, Christos S. Mantzoros
    Metabolism.2016; 65(8): 1109.     CrossRef
  • Exendin-4 Inhibits Hepatic Lipogenesis by Increasing β-Catenin Signaling
    Mi Hae Seo, Jinmi Lee, Seok-Woo Hong, Eun-Jung Rhee, Se Eun Park, Cheol Young Park, Ki Won Oh, Sung Woo Park, Won-Young Lee, Catherine Mounier
    PLOS ONE.2016; 11(12): e0166913.     CrossRef
  • The Relationship between Type 2 Diabetes Mellitus and Non-Alcoholic Fatty Liver Disease Measured by Controlled Attenuation Parameter
    Young Eun Chon, Kwang Joon Kim, Kyu Sik Jung, Seung Up Kim, Jun Yong Park, Do Young Kim, Sang Hoon Ahn, Chae Yoon Chon, Jae Bock Chung, Kyeong Hye Park, Ji Cheol Bae, Kwang-Hyub Han
    Yonsei Medical Journal.2016; 57(4): 885.     CrossRef
  • The role of the gut microbiota in NAFLD
    Christopher Leung, Leni Rivera, John B. Furness, Peter W. Angus
    Nature Reviews Gastroenterology & Hepatology.2016; 13(7): 412.     CrossRef
  • Extrapancreatic effects of incretin hormones: evidence for weight‐independent changes in morphological aspects and oxidative status in insulin‐sensitive organs of the obese nondiabetic Zucker rat (ZFR)
    Ides M. Colin, Henri Colin, Ines Dufour, Charles‐Edouard Gielen, Marie‐Christine Many, Jean Saey, Bernard Knoops, Anne‐Catherine Gérard
    Physiological Reports.2016;[Epub]     CrossRef
  • A Guide to Non-Alcoholic Fatty Liver Disease in Childhood and Adolescence
    Jonathan Temple, Paul Cordero, Jiawei Li, Vi Nguyen, Jude Oben
    International Journal of Molecular Sciences.2016; 17(6): 947.     CrossRef
  • Extrapancreatic Effect of Glucagon like Peptide-1
    In-Kyung Jeong
    The Korean Journal of Medicine.2015; 89(4): 404.     CrossRef
  • Green Tea Extract Rich in Epigallocatechin-3-Gallate Prevents Fatty Liver by AMPK Activation via LKB1 in Mice Fed a High-Fat Diet
    Aline B. Santamarina, Juliana L. Oliveira, Fernanda P. Silva, June Carnier, Laís V. Mennitti, Aline A. Santana, Gabriel H. I. de Souza, Eliane B. Ribeiro, Cláudia M. Oller do Nascimento, Fábio S. Lira, Lila M. Oyama, Patricia Aspichueta
    PLOS ONE.2015; 10(11): e0141227.     CrossRef
  • Acarbose, lente carbohydrate, and prebiotics promote metabolic health and longevity by stimulating intestinal production of GLP-1
    Mark F McCarty, James J DiNicolantonio
    Open Heart.2015; 2(1): e000205.     CrossRef
  • The Glucagon-Like Peptide-1 Analogue Liraglutide Inhibits Oxidative Stress and Inflammatory Response in the Liver of Rats with Diet-Induced Non-alcoholic Fatty Liver Disease
    Huiting Gao, Zhigang Zeng, Han Zhang, Xiaoli Zhou, Lichang Guan, Weiping Deng, Lishu Xu
    Biological & Pharmaceutical Bulletin.2015; 38(5): 694.     CrossRef
  • Glucagon-like polypeptide agonists in type 2 diabetes mellitus: efficacy and tolerability, a balance
    Sri Harsha Tella, Marc S. Rendell
    Therapeutic Advances in Endocrinology and Metabolism.2015; 6(3): 109.     CrossRef
  • Gut Microbiota: Association with NAFLD and Metabolic Disturbances
    E. Lau, D. Carvalho, P. Freitas
    BioMed Research International.2015; 2015: 1.     CrossRef
  • Acarbose: safe and effective for lowering postprandial hyperglycaemia and improving cardiovascular outcomes
    James J DiNicolantonio, Jaikrit Bhutani, James H O'Keefe
    Open Heart.2015; 2(1): e000327.     CrossRef
  • Nonalcoholic fatty liver disease and bariatric surgery in adolescents
    AiXuan Holterman, Juan Gurria, Smita Tanpure, Nerina DiSomma
    Seminars in Pediatric Surgery.2014; 23(1): 49.     CrossRef
  • Pediatric non-alcoholic fatty liver disease: New insights and future directions
    Pierluigi Marzuillo
    World Journal of Hepatology.2014; 6(4): 217.     CrossRef
  • 4Ps medicine of the fatty liver: the research model of predictive, preventive, personalized and participatory medicine—recommendations for facing obesity, fatty liver and fibrosis epidemics
    Francesca Maria Trovato, Daniela Catalano, Giuseppe Musumeci, Guglielmo M Trovato
    EPMA Journal.2014;[Epub]     CrossRef
  • The Role of Medications for the Management of Patients with NAFLD
    Natalia Mazzella, Laura M. Ricciardi, Arianna Mazzotti, Giulio Marchesini
    Clinics in Liver Disease.2014; 18(1): 73.     CrossRef
  • Randomised clinical trial: the beneficial effects of VSL#3 in obese children with non‐alcoholic steatohepatitis
    A. Alisi, G. Bedogni, G. Baviera, V. Giorgio, E. Porro, C. Paris, P. Giammaria, L. Reali, F. Anania, V. Nobili
    Alimentary Pharmacology & Therapeutics.2014; 39(11): 1276.     CrossRef
  • The cardiometabolic benefits of glycine: Is glycine an ‘antidote’ to dietary fructose?
    Mark F McCarty, James J DiNicolantonio
    Open Heart.2014; 1(1): e000103.     CrossRef
  • Pediatric non-alcoholic fatty liver disease: an increasing public health issue
    S. Berardis, E. Sokal
    European Journal of Pediatrics.2014; 173(2): 131.     CrossRef
  • Glucagon‐like peptide‐1 analogue, liraglutide, improves liver fibrosis markers in obese women with polycystic ovary syndrome and nonalcoholic fatty liver disease
    H. Kahal, G. Abouda, A. S. Rigby, A. M. Coady, E. S. Kilpatrick, S. L. Atkin
    Clinical Endocrinology.2014; 81(4): 523.     CrossRef
  • Comparative analysis of plasma metabolomics response to metabolic challenge tests in healthy subjects and influence of the FTO obesity risk allele
    Simone Wahl, Susanne Krug, Cornelia Then, Anna Kirchhofer, Gabi Kastenmüller, Tina Brand, Thomas Skurk, Melina Claussnitzer, Cornelia Huth, Margit Heier, Christa Meisinger, Annette Peters, Barbara Thorand, Christian Gieger, Cornelia Prehn, Werner Römisch-
    Metabolomics.2014; 10(3): 386.     CrossRef
  • Role of thiazolidinediones, insulin sensitizers, in non‐alcoholic fatty liver disease
    Eugene Chang, Cheol‐Young Park, Sung Woo Park
    Journal of Diabetes Investigation.2013; 4(6): 517.     CrossRef
  • Therapeutic options in pediatric non alcoholic fatty liver disease: current status and future directions
    Pietro Vajro, Selvaggia Lenta, Claudio Pignata, Mariacarolina Salerno, Roberta D’Aniello, Ida De Micco, Giulia Paolella, Giancarlo Parenti
    Italian Journal of Pediatrics.2012; 38(1): 55.     CrossRef
Original Article
Dietary Oleate Has Beneficial Effects on Every Step of Non-Alcoholic Fatty Liver Disease Progression in a Methionine- and Choline-Deficient Diet-Fed Animal Model
Ji Young Lee, Jae Hoon Moon, Jong Suk Park, Byung-Wan Lee, Eun Seok Kang, Chul Woo Ahn, Hyun Chul Lee, Bong Soo Cha
Diabetes Metab J. 2011;35(5):489-496.   Published online October 31, 2011
DOI: https://doi.org/10.4093/dmj.2011.35.5.489
  • 30,896 View
  • 45 Download
  • 17 Crossref
AbstractAbstract PDFPubReader   
Background

Non-alcoholic fatty liver disease (NAFLD) is increasingly recognized as a major cause of liver-related morbidity and mortality. The underlying mechanisms of disease progression remain poorly understood, and primary therapy of NAFLD is not yet established. We investigated the effects of dietary oleate on the development and progression of NAFLD in a methionine- and choline-deficient (MCD) diet-fed animal model.

Methods

A total of 30 C57BL/6J mice were randomly divided into three groups (n=10 in each group) and fed various experimental diets for four weeks: chow, MCD diet, or OMCD (MCD diet with oleate, 0.5 mg/g/day). Liver samples were examined for steatohepatitis and fibrosis parameters and associated genes.

Results

Additional dietary oleate dramatically reduced MCD diet-induced hepatic steatosis. Hepatic carbohydrate responsive element-binding protein was overexpressed in MCD diet-fed mice, and dietary oleate prevented this overexpression (P<0.001). Dietary oleate partially prevented MCD diet-induced serum level increases in aspartate aminotransferase and alanine aminotransferase (P<0.001, respectively). The mRNA expressions of hepatic monocyte chemoattractant protein 1, tumor necrosis factor-α and matrix metalloproteinase-9 were increased in MCD diet-fed mice, and this overexpression of inflammatory molecules was prevented by dietary oleate (P<0.001). Hepatic pericellular fibrosis was observed in MCD diet-fed mice, and dietary oleate prevented this fibrosis. Altogether, dietary oleate prevented MCD diet-induced hepatic steatosis, inflammation and fibrosis.

Conclusion

Dietary oleate has beneficial effects in every step of NAFLD development and progression and could be a nutritional option for NAFLD prevention and treatment.

Citations

Citations to this article as recorded by  
  • Bidirectional association between NAFLD and gallstone disease: a systematic review and meta-analysis of observational studies
    Shengying Gu, Shanshan Hu, Shuowen Wang, Chendong Qi, Chenyang Shi, Guorong Fan
    Expert Review of Gastroenterology & Hepatology.2023; 17(3): 283.     CrossRef
  • The Effect of Bioactive Aliment Compounds and Micronutrients on Non-Alcoholic Fatty Liver Disease
    Camelia Munteanu, Betty Schwartz
    Antioxidants.2023; 12(4): 903.     CrossRef
  • Single‐cell transcriptomics stratifies organoid models of metabolic dysfunction‐associated steatotic liver disease
    Anja Hess, Stefan D Gentile, Amel Ben Saad, Raza‐Ur Rahman, Tim Habboub, Daniel S Pratt, Alan C Mullen
    The EMBO Journal.2023;[Epub]     CrossRef
  • Histopathological Examination of the Effects of Tocilizumab and Dexamethasone on the Liver in Rats of Oleic Acid induced Acute Lung Injury
    Funda TERZİ, Hüseyin Serkan EROL
    Balıkesır Health Sciences Journal.2022;[Epub]     CrossRef
  • Identifying Lipid Metabolites Influenced by Oleic Acid Administration Using High-Performance Liquid Chromatography–Mass Spectrometry-Based Lipidomics
    Chao Xu, Dan Song, Askild L. Holck, Youyou Zhou, Rong Liu
    ACS Omega.2020; 5(20): 11314.     CrossRef
  • Causative and Sanative dynamicity of ChREBP in Hepato-Metabolic disorders
    P. Vineeth Daniel, Prosenjit Mondal
    European Journal of Cell Biology.2020; 99(8): 151128.     CrossRef
  • PPARδ attenuates hepatic steatosis through autophagy-mediated fatty acid oxidation
    Lei Tong, Long Wang, Shuangshuang Yao, Lina Jin, Jian Yang, Yifei Zhang, Guang Ning, Zhiguo Zhang
    Cell Death & Disease.2019;[Epub]     CrossRef
  • Butyrate Protects Mice Against Methionine–Choline-Deficient Diet-Induced Non-alcoholic Steatohepatitis by Improving Gut Barrier Function, Attenuating Inflammation and Reducing Endotoxin Levels
    Jianzhong Ye, Longxian Lv, Wenrui Wu, Yating Li, Ding Shi, Daiqiong Fang, Feifei Guo, Huiyong Jiang, Ren Yan, Wanchun Ye, Lanjuan Li
    Frontiers in Microbiology.2018;[Epub]     CrossRef
  • Olive oil combined with Lycium barbarum polysaccharides attenuates liver apoptosis and inflammation induced by carbon tetrachloride in rats
    Yun-Yun Chiang, Jane C.-J. Chao
    Journal of Functional Foods.2018; 48: 329.     CrossRef
  • Dietary oleic acid regulates hepatic lipogenesis through a liver X receptor-dependent signaling
    Simon Ducheix, Alexandra Montagner, Arnaud Polizzi, Frédéric Lasserre, Marion Régnier, Alice Marmugi, Fadila Benhamed, Justine Bertrand-Michel, Laila Mselli-Lakhal, Nicolas Loiseau, Pascal G. Martin, Jean-Marc Lobaccaro, Laurent Ferrier, Catherine Postic,
    PLOS ONE.2017; 12(7): e0181393.     CrossRef
  • Is hepatic lipogenesis fundamental for NAFLD/NASH? A focus on the nuclear receptor coactivator PGC-1β
    Simon Ducheix, Maria Carmela Vegliante, Gaetano Villani, Nicola Napoli, Carlo Sabbà, Antonio Moschetta
    Cellular and Molecular Life Sciences.2016; 73(20): 3809.     CrossRef
  • Metformin alleviates hepatosteatosis by restoring SIRT1-mediated autophagy induction via an AMP-activated protein kinase-independent pathway
    Young Mi Song, Yong-ho Lee, Ji-Won Kim, Dong-Sik Ham, Eun-Seok Kang, Bong Soo Cha, Hyun Chul Lee, Byung-Wan Lee
    Autophagy.2015; 11(1): 46.     CrossRef
  • Adipokines and proinflammatory cytokines, the key mediators in the pathogenesis of nonalcoholic fatty liver disease
    Sanja Stojsavljević
    World Journal of Gastroenterology.2014; 20(48): 18070.     CrossRef
  • Microglial Cell Activation Increases Saturated and Decreases Monounsaturated Fatty Acid Content, but Both Lipid Species are Proinflammatory
    Emily B. Button, Andrew S. Mitchell, Marcia M. Domingos, Jessica H.‐J. Chung, Ryan M. Bradley, Ashkan Hashemi, Phillip M. Marvyn, Ashley C. Patterson, Ken D. Stark, Joe Quadrilatero, Robin E. Duncan
    Lipids.2014; 49(4): 305.     CrossRef
  • Modeling progressive non-alcoholic fatty liver disease in the laboratory mouse
    Jesse D. Riordan, Joseph H. Nadeau
    Mammalian Genome.2014; 25(9-10): 473.     CrossRef
  • Rapid chromatographic method to decipher distinct alterations in lipid classes in NAFLD/NASH
    Stephan Laggai, Yvette Simon, Theo Ranssweiler, Alexandra K Kiemer, Sonja M Kessler
    World Journal of Hepatology.2013; 5(10): 558.     CrossRef
  • Dimethyl sulfoxide reduces hepatocellular lipid accumulation through autophagy induction
    Young Mi Song, Sun-Ok Song, Yong-Keun Jung, Eun-Seok Kang, Bong Soo Cha, Hyun Chul Lee, Byung-Wan Lee
    Autophagy.2012; 8(7): 1085.     CrossRef

Diabetes Metab J : Diabetes & Metabolism Journal
Close layer
TOP