1
|
Chang K, Hong F, Liu H, Fang Y, Wang H, Song N, Ning Y, Lu Z, Jin S, Dai Y, Ding X. FTO aggravates podocyte injury and diabetic nephropathy progression via m6A-dependent stabilization of ACC1 mRNA and promoting fatty acid metabolism. Biochem Pharmacol 2025; 235:116819. [PMID: 39988261 DOI: 10.1016/j.bcp.2025.116819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/25/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Podocyte injury associated with albuminuria and diabetic nephropathy (DN) progression. N6-methyladenosine (m6A) is a common form of epigenetic modification in eukaryotic cells and is known to be associated with a variety of disease processes. Its role in podocyte injury of DN remains poorly studied. We observed a higher expression of fat mass and obesity-associated protein (FTO) both in diabetic mice and human kidneys and DN podocytes in vitro, and the level of FTO was correlated with lipid accumulation. Furthermore, we confirmed that two selective FTO demethylation inhibitors meclofenamic acid (MA) and diacerein (DIA) administration effectively ameliorated lipotoxicity-induced podocyte injury, evidenced by restored autophagy, inhibition of apoptosis and inflammation, as well as mitigating endoplasmic reticulum stress (ERS) and mitochondrial damage both in vitro and vivo model of DN. Mechanistically, FTO demethylation inhibitors downregulated Acetyl-CoA-carboxylase 1 (ACC1) levels in db/db mice and advanced glycation end product (AGE)-treated podocytes, subsequently decreased podocyte fatty acid accumulation. ACC1 was identified as a direct FTO target in which FTO stabilizes ACC1 mRNA with the mediation of YTH domain-containing family protein 2 (YTHDF2) in an m6A-dependent manner using m6A RNA immunoprecipitation-quantitative real-time PCR (MeRIP-qPCR) and dual-luciferase reporter gene assays. Collectively, our findings demonstrate an important role of FTO mediated-m6A modification of ACC1 contributed to s lipotoxicity-mediated injury of DN podocytes, which provide fresh insights into the therapeutic strategies for DN.
Collapse
Affiliation(s)
- Kaili Chang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Kidney and Dialysis Institute of Shanghai, Kidney and Blood Purification Laboratory of Shanghai, Shanghai 200032, China
| | - Fuyuan Hong
- Division of Nephrology, Fujian Provincial Clinical College, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350122, China
| | - Hong Liu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Kidney and Dialysis Institute of Shanghai, Kidney and Blood Purification Laboratory of Shanghai, Shanghai 200032, China
| | - Yi Fang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Kidney and Dialysis Institute of Shanghai, Kidney and Blood Purification Laboratory of Shanghai, Shanghai 200032, China
| | - Hongyu Wang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Kidney and Dialysis Institute of Shanghai, Kidney and Blood Purification Laboratory of Shanghai, Shanghai 200032, China
| | - Nana Song
- Division of Nephrology, Zhongshan Hospital, Fudan University, Kidney and Dialysis Institute of Shanghai, Kidney and Blood Purification Laboratory of Shanghai, Shanghai 200032, China
| | - Yichun Ning
- Division of Nephrology, Zhongshan Hospital, Fudan University, Kidney and Dialysis Institute of Shanghai, Kidney and Blood Purification Laboratory of Shanghai, Shanghai 200032, China
| | - Zhihui Lu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Kidney and Dialysis Institute of Shanghai, Kidney and Blood Purification Laboratory of Shanghai, Shanghai 200032, China
| | - Shi Jin
- Division of Nephrology, Zhongshan Hospital, Fudan University, Kidney and Dialysis Institute of Shanghai, Kidney and Blood Purification Laboratory of Shanghai, Shanghai 200032, China
| | - Yan Dai
- Division of Nephrology, Zhongshan Hospital, Fudan University, Kidney and Dialysis Institute of Shanghai, Kidney and Blood Purification Laboratory of Shanghai, Shanghai 200032, China.
| | - Xiaoqiang Ding
- Division of Nephrology, Zhongshan Hospital, Fudan University, Kidney and Dialysis Institute of Shanghai, Kidney and Blood Purification Laboratory of Shanghai, Shanghai 200032, China.
| |
Collapse
|
2
|
Prabhakar PK, Batiha GES. Potential Therapeutic Targets for the Management of Diabetes Mellitus Type 2. Curr Med Chem 2024; 31:3167-3181. [PMID: 37125833 DOI: 10.2174/0929867330666230501172557] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/14/2023] [Accepted: 03/26/2023] [Indexed: 05/02/2023]
Abstract
Diabetes is one of the lifelong chronic metabolic diseases which is prevalent globally. There is a continuous rise in the number of people suffering from this disease with time. It is characterized by hyperglycemia, which leads to severe damage to the body's system, such as blood vessels and nerves. Diabetes occurs due to the dysfunction of pancreatic β -cell which leads to the reduction in the production of insulin or body cells unable to use insulin produce efficiently. As per the data shared International diabetes federation (IDF), there are around 415 million affected by this disease worldwide. There are a number of hit targets available that can be focused on treating diabetes. There are many drugs available and still under development for the treatment of type 2 diabetes. Inhibition of gluconeogenesis, lipolysis, fatty acid oxidation, and glucokinase activator is emerging targets for type 2 diabetes treatment. Diabetes management can be supplemented with drug intervention for obesity. The antidiabetic drug sale is the second-largest in the world, trailing only that of cancer. The future of managing diabetes will be guided by research on various novel targets and the development of various therapeutic leads, such as GLP-1 agonists, DPP-IV inhibitors, and SGLT2 inhibitors that have recently completed their different phases of clinical trials. Among these therapeutic targets associated with type 2 diabetes, this review focused on some common therapeutic targets for developing novel drug candidates of the newer generation with better safety and efficacy.
Collapse
Affiliation(s)
- Pranav Kumar Prabhakar
- Division of Research and Development, Lovely Professional University, Phagwara (Punjab) 144411, India
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| |
Collapse
|
3
|
Ali I, Khan A, Fa Z, Khan T, Wei DQ, Zheng J. Crystal structure of Acetyl-CoA carboxylase (AccB) from Streptomyces antibioticus and insights into the substrate-binding through in silico mutagenesis and biophysical investigations. Comput Biol Med 2022; 145:105439. [PMID: 35344865 DOI: 10.1016/j.compbiomed.2022.105439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 03/14/2022] [Accepted: 03/20/2022] [Indexed: 11/18/2022]
Abstract
Acetyl-CoA carboxylase (ACC) is crucial for polyketides biosynthesis and acts as an essential metabolic checkpoint. It is also an attractive drug target against obesity, cancer, microbial infections, and diabetes. However, the lack of knowledge, particularly sequence-structure function relationship to narrate ligand-enzyme binding, has hindered the progress of ACC-specific therapeutics and unnatural "natural" polyketides. Structural characterization of such enzymes will boost the opportunity to understand the substrate binding, designing new inhibitors and information regarding the molecular rules which control the substrate specificity of ACCs. To understand the substrate specificity, we determined the crystal structure of AccB (Carboxyl-transferase, CT) from Streptomyces antibioticus with a resolution of 2.3 Å and molecular modeling approaches were employed to unveil the molecular mechanism of acetyl-CoA recognition and processing. The CT domain of S. antibioticus shares a similar structural organization with the previous structures and the two steps reaction was confirmed by enzymatic assay. Furthermore, to reveal the key hotspots required for the substrate recognition and processing, in silico mutagenesis validated only three key residues (V223, Q346, and Q514) that help in the fixation of the substrate. Moreover, we also presented atomic level knowledge on the mechanism of the substrate binding, which unveiled the terminal loop (500-514) function as an opening and closing switch and pushes the substrate inside the cavity for stable binding. A significant decline in the hydrogen bonding half-life was observed upon the alanine substitution. Consequently, the presented structural data highlighted the potential key interacting residues for substrate recognition and will also help to re-design ACCs active site for proficient substrate specificity to produce diverse polyketides.
Collapse
Affiliation(s)
- Imtiaz Ali
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Abbas Khan
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Zhang Fa
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Taimoor Khan
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Dong-Qing Wei
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China; State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200030, PR China; Peng Cheng Laboratory, Vanke Cloud City Phase I Building 8, Xili Street, Nashan District, Shenzhen, Guangdong, 518055, PR China
| | - Jianting Zheng
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, PR China.
| |
Collapse
|
4
|
Wang Y, Yu W, Li S, Guo D, He J, Wang Y. Acetyl-CoA Carboxylases and Diseases. Front Oncol 2022; 12:836058. [PMID: 35359351 PMCID: PMC8963101 DOI: 10.3389/fonc.2022.836058] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/10/2022] [Indexed: 12/28/2022] Open
Abstract
Acetyl-CoA carboxylases (ACCs) are enzymes that catalyze the carboxylation of acetyl-CoA to produce malonyl-CoA. In mammals, ACC1 and ACC2 are two members of ACCs. ACC1 localizes in the cytosol and acts as the first and rate-limiting enzyme in the de novo fatty acid synthesis pathway. ACC2 localizes on the outer membrane of mitochondria and produces malonyl-CoA to regulate the activity of carnitine palmitoyltransferase 1 (CPT1) that involves in the β-oxidation of fatty acid. Fatty acid synthesis is central in a myriad of physiological and pathological conditions. ACC1 is the major member of ACCs in mammalian, mountains of documents record the roles of ACC1 in various diseases, such as cancer, diabetes, obesity. Besides, acetyl-CoA and malonyl-CoA are cofactors in protein acetylation and malonylation, respectively, so that the manipulation of acetyl-CoA and malonyl-CoA by ACC1 can also markedly influence the profile of protein post-translational modifications, resulting in alternated biological processes in mammalian cells. In the review, we summarize our understandings of ACCs, including their structural features, regulatory mechanisms, and roles in diseases. ACC1 has emerged as a promising target for diseases treatment, so that the specific inhibitors of ACC1 for diseases treatment are also discussed.
Collapse
Affiliation(s)
- Yu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science of Technology, Wuhan, China
| | - Weixing Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science of Technology, Wuhan, China
| | - Sha Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science of Technology, Wuhan, China
| | - Dingyuan Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science of Technology, Wuhan, China
| | - Jie He
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science of Technology, Wuhan, China
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yugang Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science of Technology, Wuhan, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Yugang Wang,
| |
Collapse
|
5
|
Cai J, Zang X, Wu Z, Liu J, Wang D. Altered protein S-glutathionylation depicts redox imbalance triggered by transition metal oxide nanoparticles in a breastfeeding system. NANOIMPACT 2021; 22:100305. [PMID: 35559962 DOI: 10.1016/j.impact.2021.100305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/25/2021] [Accepted: 02/19/2021] [Indexed: 06/15/2023]
Abstract
Nanosafety has become a public concern following nanotechnology development. By now, attention has seldom been paid to breastfeeding system, which is constructed by mammary physiological structure and derived substances (endogenous or exogenous), cells, tissues, organs, and individuals (mother and child), connecting environment and organism, and spans across mother-child dyad. Thus, breastfeeding system is a center of nutrients transport and a unique window of toxic susceptibility in the mother-child dyad. We applied metabolomics combined with redox proteomics to depict how nanoparticles cause metabolic burden via their spontaneous redox cycling in lactating mammary glands. Two widely used nanoparticles [titanium dioxide (nTiO2) and zinc oxide (nZnO)] were exposed to lactating mice via intranasal administration. Biodistribution and biopersistence of nTiO2 and nZnO in mammary glands destroyed its structure, reflective of significantly reduced claudin-3 protein level by 32.1% (P < 0.01) and 47.8% (P < 0.01), and significantly increased apoptosis index by 85.7 (P < 0.01) and 100.3 (P < 0.01) fold change, respectively. Airway exposure of nTiO2 trended to reduced milk production by 22.7% (P = 0.06), while nZnO significantly reduced milk production by 33.0% (P < 0.01). Metabolomics analysis revealed a metabolic shift by nTiO2 or nZnO, such as increased glycolysis (nTiO2: fold enrichment = 3.31, P < 0.05; nZnO: fold enrichment = 3.68, P < 0.05), glutathione metabolism (nTiO2: fold enrichment = 5.57, P < 0.01; nZnO: fold enrichment = 4.43, P < 0.05), and fatty acid biosynthesis (nTiO2: fold enrichment = 3.52, P < 0.05; nZnO: fold enrichment = 3.51, P < 0.05) for tissue repair at expense of lower milk fat synthesis (35.7% reduction by nTiO2; 51.8% reduction by nZnO), and finally led to oxidative stress of mammary glands. The increased GSSG/GSH ratio (57.5% increase by nTiO2; 105% increase by nZnO) with nanoparticle exposure confirmed an alteration in the redox state and a metabolic shift in mammary glands. Redox proteomics showed that nanoparticles induced S-glutathionylation (SSG) modification at Cys sites of proteins in a nanoparticle type-dependent manner. The nTiO2 induced more protein SSG modification sites (nTiO2: 21; nZnO:16), whereas nZnO induced fewer protein SSG modification sites but at deeper SSG levels (26.6% higher in average of nZnO than that of nTiO2). In detail, SSG modification by nTiO2 was characterized by Ltf at Cys423 (25.3% increase), and Trf at Cys386;395;583 (42.3%, 42.3%, 22.8% increase) compared with control group. While, SSG modification by nZnO was characterized by Trfc at Cys365 (71.3% increase) and Fasn at Cys1010 (41.0% increase). The discovery of SSG-modified proteins under airway nanoparticle exposure further supplemented the oxidative stress index and mammary injury index, and deciphered precise mechanisms of nanotoxicity into a molecular level. The unique quantitative site-specific redox proteomics and metabolomics can serve as a new technique to identify nanotoxicity and provide deep insights into nanoparticle-triggered oxidative stress, contributing to a healthy breastfeeding environment.
Collapse
Affiliation(s)
- Jie Cai
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| | - Xinwei Zang
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| | - Zezhong Wu
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China
| | - Jianxin Liu
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| | - Diming Wang
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| |
Collapse
|
6
|
Wada M, Yukawa K, Ogasawara H, Suzawa K, Maekawa T, Yamamoto Y, Ohta T, Lee E, Miki T. GPR52 accelerates fatty acid biosynthesis in a ligand-dependent manner in hepatocytes and in response to excessive fat intake in mice. iScience 2021; 24:102260. [PMID: 33796846 PMCID: PMC7995607 DOI: 10.1016/j.isci.2021.102260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/06/2021] [Accepted: 02/26/2021] [Indexed: 12/17/2022] Open
Abstract
Gpr52 is an orphan G-protein-coupled receptor of unknown physiological function. We found that Gpr52-deficient (Gpr52−/−) mice exhibit leanness associated with reduced liver weight, decreased hepatic de novo lipogenesis, and enhanced insulin sensitivity. Treatment of the hepatoma cell line HepG2 cells with c11, the synthetic GPR52 agonist, increased fatty acid biosynthesis, and GPR52 knockdown (KD) abolished the lipogenic action of c11. In addition, c11 induced the expressions of lipogenic enzymes (SCD1 and ELOVL6), whereas these inductions were attenuated by GPR52-KD. In contrast, cholesterol biosynthesis was not increased by c11, but its basal level was significantly suppressed by GPR52-KD. High-fat diet (HFD)-induced increase in hepatic expression of Pparg2 and its targets (Scd1 and Elovl6) was absent in Gpr52−/− mice with alleviated hepatosteatosis. Our present study showed that hepatic GPR52 promotes the biosynthesis of fatty acid and cholesterol in a ligand-dependent and a constitutive manner, respectively, and Gpr52 participates in HFD-induced fatty acid synthesis in liver. Hepatosteatosis is inherently an adaptive response to overnutrition to store energy On the other hand, it can be a pathological condition causing insulin resistance High-fat diet increases PPARγ2 expression and lipogenesis in liver via GPR52 Gpr52 ablation protects mice from developing hepatosteatosis and insulin resistance
Collapse
Affiliation(s)
- Mitsuo Wada
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan.,Pharmaceutical Frontier Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Yokohama 236-0004, Japan
| | - Kayo Yukawa
- Pharmaceutical Frontier Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Yokohama 236-0004, Japan
| | - Hiroyuki Ogasawara
- Pharmaceutical Frontier Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Yokohama 236-0004, Japan
| | - Koichi Suzawa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki 569-1125, Japan
| | - Tatsuya Maekawa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki 569-1125, Japan
| | - Yoshihisa Yamamoto
- Pharmaceutical Frontier Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Yokohama 236-0004, Japan
| | - Takeshi Ohta
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Eunyoung Lee
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan
| | - Takashi Miki
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670, Japan
| |
Collapse
|
7
|
Demir E, Aslan A. Protective effect of pristine C60 fullerene nanoparticle in combination with curcumin against hyperglycemia-induced kidney damage in diabetes caused by streptozotocin. J Food Biochem 2020; 44:e13470. [PMID: 32914898 DOI: 10.1111/jfbc.13470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022]
Abstract
The present study aims to examine the protective effects of C60 fullerene (C60), Curcumin (CUR; Curcuma longa), C60 + CUR combination against oxidative stress, apoptosis, and changes in cellular level in kidney tissue of diabetic rats. Treatment practices were administered separately to groups for 8 weeks following the approval of diabetes induction. It was observed that the treatment groups had increased antioxidant potential, decreased oxidative stress levels, decreased cholesterol, alpha tocopherol, retinol levels along with improved important changes in fatty acid metabolism compared with the diabetic group. C60, CUR, and C60 + CUR were also determined to act in the direction of reducing kidney damage by activating apoptotic pathways. It can be concluded based on these findings that C60, CUR, and especially C60 + CUR combination has beneficial properties in maintaining kidney tissue and function by effectively preventing oxidative stress, apoptotic changes, and changes at the cellular level in kidney tissue under hyperglycemia conditions. PRACTICAL APPLICATIONS: C60 and CUR have various biological activities which can be indicated as antioxidant, anti-inflammatory, anticancer, neuroprotective, and hepatoprotective. It has been reported that C60 and CUR protect the cells against oxidative injury brought about by reactive oxygen species (ROS). Data acquired from the present study puts forth that C60 and C60 + CUR may be promising agents to prevent damage induced by hyperglycemic conditions in kidney tissue.
Collapse
Affiliation(s)
- Ersin Demir
- Department of Agricultural Biotechnology, Faculty of Agriculture and Natural Sciences, Duzce University, Duzce, Turkey
| | - Abdullah Aslan
- Department of Biology-Molecular Biology and Genetics Program, Faculty of Science, Firat University, Elazig, Turkey
| |
Collapse
|
8
|
Saran AR, Dave S, Zarrinpar A. Circadian Rhythms in the Pathogenesis and Treatment of Fatty Liver Disease. Gastroenterology 2020; 158:1948-1966.e1. [PMID: 32061597 PMCID: PMC7279714 DOI: 10.1053/j.gastro.2020.01.050] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/13/2022]
Abstract
Circadian clock proteins are endogenous timing mechanisms that control the transcription of hundreds of genes. Their integral role in coordinating metabolism has led to their scrutiny in a number of diseases, including nonalcoholic fatty liver disease (NAFLD). Discoordination between central and peripheral circadian rhythms is a core feature of nearly every genetic, dietary, or environmental model of metabolic syndrome and NAFLD. Restricting feeding to a defined daily interval (time-restricted feeding) can synchronize the central and peripheral circadian rhythms, which in turn can prevent or even treat the metabolic syndrome and hepatic steatosis. Importantly, a number of proteins currently under study as drug targets in NAFLD (sterol regulatory element-binding protein [SREBP], acetyl-CoA carboxylase [ACC], peroxisome proliferator-activator receptors [PPARs], and incretins) are modulated by circadian proteins. Thus, the clock can be used to maximize the benefits and minimize the adverse effects of pharmaceutical agents for NAFLD. The circadian clock itself has the potential for use as a target for the treatment of NAFLD.
Collapse
Affiliation(s)
- Anand R. Saran
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA
| | - Shravan Dave
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, La Jolla, California; Veterans Affairs Health Sciences San Diego, La Jolla, California; Institute of Diabetes and Metabolic Health, University of California, San Diego, La Jolla, California; Center for Microbiome Innovation, University of California, San Diego, La Jolla, California.
| |
Collapse
|
9
|
Ali A, Zhang Y, Fu M, Pei Y, Wu L, Wang R, Yang G. Cystathionine gamma-lyase/H 2S system suppresses hepatic acetyl-CoA accumulation and nonalcoholic fatty liver disease in mice. Life Sci 2020; 252:117661. [PMID: 32305523 DOI: 10.1016/j.lfs.2020.117661] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/05/2020] [Accepted: 04/08/2020] [Indexed: 02/07/2023]
Abstract
AIMS Hydrogen sulfide (H2S) as a novel gasotransmitter can be endogenously produced in liver by cystathionine gamma-lyase (CSE). The dysfunctions of CSE/H2S system have been linked to various liver diseases. Acetyl-CoA is the key intermediate from the metabolism of lipid. This study examined the roles of H2S in hepatic acetyl-CoA and lipid metabolism. MATERIALS AND METHODS Both in vitro cell model and in vivo animal model of lipid accumulation were used in this study. Western blotting and real-time PCR were used for analysis of protein and mRNA expression. Acetyl-CoA was analyzed by a coupled enzyme assay, and lipid accumulation was observed with Oil Red O staining. KEY FINDINGS Incubation of human liver carcinoma (HepG2) cells with a mixture of free fatty acids (FFAs) or high glucose reduced CSE expression and H2S production, promoted intracellular accumulation of acetyl-CoA and lipid. Supply of exogenous NaHS or cysteine reduced acetyl-CoA contents and lipid accumulation, while blockage of CSE activity promoted intracellular lipid accumulation. Furthermore, H2S blocked FFAs-induced transcriptions of de novo lipogenesis, inflammation, and fibrosis-related genes. In vivo, knockout of CSE gene stimulated more hepatic acetyl-CoA and lipid accumulation in mice induced by high-fat choline-deficient diet. The expressions of lipogenesis, inflammation, and fibrosis-related genes were significantly higher in liver tissues from CSE knockout mice when compared with wild-type mice. SIGNIFICANCE CSE/H2S system is indispensable for maintaining the homeostasis of acetyl-CoA and lipid accumulation and protecting from the development of inflammation and fibrosis in liver under excessive caloric ingestion.
Collapse
Affiliation(s)
- Amr Ali
- Department of Chemistry and Biochemistry, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Yanjie Zhang
- Department of Chemistry and Biochemistry, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada; School of Life Science, Shanxi University, Taiyuan, China
| | - Ming Fu
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada; School of Human Kinetics, Laurentian University, Sudbury, Canada; Health Science North Research Institute, Sudbury, Canada
| | - Yanxi Pei
- School of Life Science, Shanxi University, Taiyuan, China
| | - Lingyun Wu
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada; School of Human Kinetics, Laurentian University, Sudbury, Canada; Health Science North Research Institute, Sudbury, Canada
| | - Rui Wang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Guangdong Yang
- Department of Chemistry and Biochemistry, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada.
| |
Collapse
|
10
|
Wang J, He W, Tsai PJ, Chen PH, Ye M, Guo J, Su Z. Mutual interaction between endoplasmic reticulum and mitochondria in nonalcoholic fatty liver disease. Lipids Health Dis 2020; 19:72. [PMID: 32284046 PMCID: PMC7155254 DOI: 10.1186/s12944-020-01210-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common metabolic syndrome. Imbalances between liver lipid output and input are the direct causes of NAFLD, and hepatic steatosis is the pathological premise and basis for NAFLD progression. Mutual interaction between endoplasmic reticulum stress (ERS) and oxidative stress play important roles in NAFLD pathogenesis. Notably, mitochondria-associated membranes (MAMs) act as a structural bridges for functional clustering of molecules, particularly for Ca2+, lipids, and reactive oxygen species (ROS) exchange. Previous studies have examined the crucial roles of ERS and ROS in NAFLD and have shown that MAM structural and functional integrity determines normal ER- mitochondria communication. Upon disruption of MAM integrity, miscommunication directly or indirectly causes imbalances in Ca2+ homeostasis and increases ERS and oxidative stress. Here, we emphasize the involvement of MAMs in glucose and lipid metabolism, chronic inflammation and insulin resistance in NAFLD and summarize MAM-targeting drugs and compounds, most of which achieve their therapeutic or ameliorative effects on NAFLD by improving MAM integrity. Therefore, targeting MAMs may be a viable strategy for NAFLD treatment. This review provides new ideas and key points for basic NAFLD research and drug development centred on mitochondria and the endoplasmic reticulum.
Collapse
Affiliation(s)
- Jin Wang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wanping He
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ping-Ju Tsai
- King-Prebiotics Biotechnology (TW) Co., LTD, 2F.-1, No. 250, Zhongshan Rd., Linkou Dist, New Taipei City, 24446, Taiwan
| | - Pei-Hsuan Chen
- King-Prebiotics Biotechnology (TW) Co., LTD, 2F.-1, No. 250, Zhongshan Rd., Linkou Dist, New Taipei City, 24446, Taiwan
| | - Manxiang Ye
- New Francisco (Yunfu City) Biotechnology Co, Ltd Swan-kan-chiau Ind. Dist., Kaofong Village, Yunfu City, Guangdong, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
11
|
Huang CH, Lin WK, Chang SH, Tsai GJ. Ganoderma lucidum culture supplement ameliorates dyslipidemia and reduces visceral fat accumulation in type 2 diabetic rats. Mycology 2020; 12:94-104. [PMID: 34026301 PMCID: PMC8128174 DOI: 10.1080/21501203.2020.1740409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Diabetic rats were daily fed with a high-cholesterol diet containing 1% or 3% freeze-dried whole submerged G. lucidum culture or its mycelia for 5 weeks. Body weight, adipose tissue weight and plasma triglyceride levels were reduced, while high-density lipoprotein-cholesterol levels were elevated in rats fed with G. lucidum powder supplement diets. Notably, G. lucidum supplements downregulated the activities of hepatic acetyl-CoA carboxylase, fatty acid synthase and lipoprotein lipase, but upregulated the activity of hormone-sensitive lipase in the perirenal adipose tissues. Moreover, G. lucidum supplements increased the faecal triglyceride excretion. Therefore, daily supplementation of submerged G. lucidum culture, especially mycelia, can ameliorate dyslipidemia and reduce visceral fat accumulation in diabetic rats fed with a high-fat diet, which is closely related to the modulation of lipid synthesis, metabolism, and excretion.
Collapse
Affiliation(s)
- Chung-Hsiung Huang
- Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan
| | - Wei-Kang Lin
- Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan
| | - Shun-Hsien Chang
- Institute of Food Safety and Risk Management, National Taiwan Ocean University, Keelung, Taiwan
| | - Guo-Jane Tsai
- Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan.,Center for Marine Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan
| |
Collapse
|
12
|
Takagi H, Tanimoto K, Shimazaki A, Tonomura Y, Momosaki S, Sakamoto S, Abe K, Notoya M, Yukioka H. A Novel Acetyl-CoA Carboxylase 2 Selective Inhibitor Improves Whole-Body Insulin Resistance and Hyperglycemia in Diabetic Mice through Target-Dependent Pathways. J Pharmacol Exp Ther 2020; 372:256-263. [PMID: 31900320 DOI: 10.1124/jpet.119.263590] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 12/31/2019] [Indexed: 03/08/2025] Open
Abstract
Excess intramyocellular lipid (IMCL) deposition in skeletal muscle is closely associated with insulin resistance. Pharmacological inhibition of acetyl-CoA carboxylase (ACC) 2 offers a promising approach to treat insulin resistance through stimulation of mitochondrial fatty acid oxidation (FAO) and reduction of IMCL deposition. Previously reported experimental ACC2 inhibitors exhibited plasma glucose-lowering effects in diabetic rodents. However, their antidiabetic action may be potentially biased by off-target effects on triglyceride metabolism or by neurologic side effects. In this study, we investigated a safety profile, target dependency of its action, and antidiabetic efficacy of compound 2e, a novel olefin derivative potent ACC2 selective inhibitor. Four-day administration of suprapharmacological dose of compound 2e did not exhibit any obvious side effects in Sprague-Dawley rats. In db/db mice, single administration of compound 2e led to significantly elevated FAO and reduced IMCL deposition in skeletal muscle. In ACC2 knockout mice, treatment with pharmacological doses of compound 2e did not reduce plasma triglyceride levels, whereas A-908292, a previously reported ACC2 inhibitor, caused a significant triglyceride reduction, showing that compound 2e was devoid of off-target triglyceride-lowering activity. Chronic treatment of db/db mice with compound 2e improved hyperglycemia but did not decrease plasma triglyceride levels. Additionally, compound 2e showed significant improvements of whole-body insulin resistance in the clamp study and insulin tolerance test. Collectively, compound 2e demonstrated a good safety profile and significant antidiabetic effects through inhibition of ACC2-dependent pathways. These findings provide further evidence that selective inhibition of ACC2 is an attractive strategy against insulin resistance and type 2 diabetes. SIGNIFICANCE STATEMENT: This study shows that pharmacological inhibition of acetyl-CoA carboxylase (ACC) 2 leads to significant improvements in whole-body glucose homeostasis, independently of off-target metabolic pathways and toxicity, which were observed in previously reported ACC2 inhibitors. These findings support the concept that ACC2-selective inhibitors will be a novel remedy for treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Hiroyuki Takagi
- Shionogi & Co., Ltd., Osaka, Japan (H.T., K.T., A.S., Y.T., S.M., K.A., M.N., H.Y.) and Shionogi Techno Advance Research Co., Ltd., Osaka, Japan (S.S.)
| | - Keiichi Tanimoto
- Shionogi & Co., Ltd., Osaka, Japan (H.T., K.T., A.S., Y.T., S.M., K.A., M.N., H.Y.) and Shionogi Techno Advance Research Co., Ltd., Osaka, Japan (S.S.)
| | - Atsuyuki Shimazaki
- Shionogi & Co., Ltd., Osaka, Japan (H.T., K.T., A.S., Y.T., S.M., K.A., M.N., H.Y.) and Shionogi Techno Advance Research Co., Ltd., Osaka, Japan (S.S.)
| | - Yutaka Tonomura
- Shionogi & Co., Ltd., Osaka, Japan (H.T., K.T., A.S., Y.T., S.M., K.A., M.N., H.Y.) and Shionogi Techno Advance Research Co., Ltd., Osaka, Japan (S.S.)
| | - Sotaro Momosaki
- Shionogi & Co., Ltd., Osaka, Japan (H.T., K.T., A.S., Y.T., S.M., K.A., M.N., H.Y.) and Shionogi Techno Advance Research Co., Ltd., Osaka, Japan (S.S.)
| | - Shingo Sakamoto
- Shionogi & Co., Ltd., Osaka, Japan (H.T., K.T., A.S., Y.T., S.M., K.A., M.N., H.Y.) and Shionogi Techno Advance Research Co., Ltd., Osaka, Japan (S.S.)
| | - Kohji Abe
- Shionogi & Co., Ltd., Osaka, Japan (H.T., K.T., A.S., Y.T., S.M., K.A., M.N., H.Y.) and Shionogi Techno Advance Research Co., Ltd., Osaka, Japan (S.S.)
| | - Mitsuru Notoya
- Shionogi & Co., Ltd., Osaka, Japan (H.T., K.T., A.S., Y.T., S.M., K.A., M.N., H.Y.) and Shionogi Techno Advance Research Co., Ltd., Osaka, Japan (S.S.)
| | - Hideo Yukioka
- Shionogi & Co., Ltd., Osaka, Japan (H.T., K.T., A.S., Y.T., S.M., K.A., M.N., H.Y.) and Shionogi Techno Advance Research Co., Ltd., Osaka, Japan (S.S.)
| |
Collapse
|
13
|
Bai X, Pei R, Lei W, Zhao M, Zhang J, Tian L, Shang J. Antidiabetic Effect of Artemether in Db/Db Mice Involves Regulation of AMPK and PI3K/Akt Pathways. Front Endocrinol (Lausanne) 2020; 11:568864. [PMID: 33101203 PMCID: PMC7545317 DOI: 10.3389/fendo.2020.568864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
The traditional Chinese medicine has long been used in the treatment of diabetes, one major disease threatening the public health. It has been reported that artemether exerts antidiabetic effects on type 2 diabetes in db/db mice, however the underlying mechanisms remain unknown. In the present study, we show that artemether regulates expression of related enzymes participating in the glucose and lipid metabolism in the liver of db/db mice, which could at least partly explain the improved glucose and lipid metabolism in artemether-treated mice. Additionally, artemether also regulates expression of glycogen synthesis related enzymes in the skeletal muscle of db/db mice, supporting its promotive role in glycogen synthesis. Mechanistically, artemether activates AMPK pathway as well as PI3K/Akt pathway in the liver and skeletal muscle of db/db mice, suggesting that these two signaling pathways are both involved in the antidiabetic effects of artemether on type 2 diabetes in db/db mice. In conclusion, our study connects the antidiabetic effects of artemether to the regulation of metabolic enzymes and signaling pathways, and also provides molecular basis for the potential application of artemether in treating type 2 diabetes.
Collapse
|
14
|
Li T, Yan H, Geng Y, Shi H, Li H, Wang S, Wang Y, Xu J, Zhao G, Lu X. Target genes associated with lipid and glucose metabolism in non-alcoholic fatty liver disease. Lipids Health Dis 2019; 18:211. [PMID: 31805951 PMCID: PMC6894500 DOI: 10.1186/s12944-019-1154-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 11/26/2019] [Indexed: 12/21/2022] Open
Abstract
Background Insulin resistance (IR) and lipid peroxidation are accepted as ‘two-hit’ hypothesis of Non-alcoholic fatty liver disease (NAFLD). However, there are few published research on identifying genes which connect lipid and glucose metabolism by gene microarray. Objective To identify target genes related to lipid and glucose metabolism that might be responsible for the pathogenesis of NAFLD. Methods A rat model of NAFLD was established by feeding male rats with high-fat diet and gene expression profiles of liver tissues were determined using Agilent DNA microarray. We then investigated differentially expressed genes (DEGs) and intersection of them by using Gene Ontology (GO) and Pathway Analyses. Target genes were verified by Real-time polymerase chain reaction (RT-PCR). Results Compared with control, 932 genes, including 783 up-regulated and 149 down-regulated, exhibited differences in expression. The up-regulated genes were involved in biosynthesis, cell development, cell differentiation and down-regulated genes contributed to biological metabolic process, adipokine metabolic pathway and insulin signaling pathway. We identified genes involved in insulin signaling pathway, Notch signaling pathway and lipid synthetic process to be closely related to liver fat accumulation and insulin resistance. Among them, IGFBP7, Notch1 and HMGCR were up-regulated (2.85-fold, 3.22-fold, and 2.06-fold, respectively, all P < 0.05) and ACACB was down-regulated (2.08-fold, P < 0.01). These four genes supposed to connect lipid and glucose metabolism after GO and Pathway analyses. Conclusions These findings provide innovative information on the whole genome expression profile due to high-fat diet feeding, and bring new insight into the regulating effects of genes on the lipid and glucose metabolism of NAFLD.
Collapse
Affiliation(s)
- Ting Li
- Health Science Center, Xi'an Jiaotong University, NO.76 Yanta West Road, Xi'an, 710061, China
| | - Hua Yan
- Department of Geratology, Shaanxi Provincal People's Hospital, Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710068, China
| | - Yan Geng
- Department of Paediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Haitao Shi
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, NO.157 West 5th Road, Xi'an, 710004, China
| | - Hong Li
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, NO.157 West 5th Road, Xi'an, 710004, China
| | - Shenhao Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, NO.157 West 5th Road, Xi'an, 710004, China
| | - Yatao Wang
- Health Science Center, Xi'an Jiaotong University, NO.76 Yanta West Road, Xi'an, 710061, China
| | - Jingyuan Xu
- Health Science Center, Xi'an Jiaotong University, NO.76 Yanta West Road, Xi'an, 710061, China
| | - Gang Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, NO.157 West 5th Road, Xi'an, 710004, China
| | - Xiaolan Lu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, NO.157 West 5th Road, Xi'an, 710004, China. .,Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China.
| |
Collapse
|
15
|
Alkhouri N, Lawitz E, Noureddin M, DeFronzo R, Shulman GI. GS-0976 (Firsocostat): an investigational liver-directed acetyl-CoA carboxylase (ACC) inhibitor for the treatment of non-alcoholic steatohepatitis (NASH). Expert Opin Investig Drugs 2019; 29:135-141. [PMID: 31519114 DOI: 10.1080/13543784.2020.1668374] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: De novo lipogenesis (DNL) plays a major role in fatty acid metabolism and contributes significantly to triglyceride accumulation within the hepatocytes in patients with nonalcoholic steatohepatitis (NASH). Acetyl-CoA carboxylase (ACC) converts acetyl-CoA to malonyl CoA and is a rate-controlling step in DNL. Furthermore, malonyl-CoA is an important regulator of hepatic mitochondrial fat oxidation through its ability to inhibit carnitine palmitoyltransferase I. Therefore, inhibiting ACC pharmacologically represents an attractive approach to treating NASH.Areas covered: This article summarizes preclinical and clinical data on the efficacy and safety of the liver-targeted ACC inhibitor GS-0976 (Firsocostat) for the treatment of NASH. In a phase 2 trial that included 126 patients with NASH and fibrosis, GS-0976 20 mg daily for 12 weeks showed significant relative reduction in liver fat by 29%; however, treatment was associated with an increase in plasma triglycerides with 16 patients having levels >500 mg/dL.Expert opinion: Preclinical and preliminary clinical data support the development of GS-0976 as treatment for NASH. ACC-induced hypertriglyceridemia can be mitigated by fish oil or fibrates, but the long-term cardiovascular effects require further investigations.
Collapse
Affiliation(s)
- Naim Alkhouri
- University of Texas (UT) Health San Antonio, San Antonio, TX, USA.,Texas Liver Institute, San Antonio, TX, USA
| | - Eric Lawitz
- University of Texas (UT) Health San Antonio, San Antonio, TX, USA.,Texas Liver Institute, San Antonio, TX, USA
| | | | - Ralph DeFronzo
- University of Texas (UT) Health San Antonio, San Antonio, TX, USA.,Texas Diabetes Institute (TDI), San Antonio, TX, USA
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
16
|
Pereira RM, Rodrigues KCDC, Anaruma CP, Sant'Ana MR, de Campos TDP, Gaspar RS, Canciglieri RDS, de Melo DG, Mekary RA, da Silva ASR, Cintra DE, Ropelle ER, Pauli JR, de Moura LP. Short-term strength training reduces gluconeogenesis and NAFLD in obese mice. J Endocrinol 2019; 241:59-70. [PMID: 30878016 DOI: 10.1530/joe-18-0567] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/15/2019] [Indexed: 12/20/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has a positive correlation with obesity, insulin resistance and type 2 diabetes mellitus (T2D). The aerobic training is an important tool in combating NAFLD. However, no studies have demonstrated the molecular effects of short-term strength training on the accumulation of hepatic fat in obese mice. This study aimed to investigate the effects of short-term strength training on the mechanisms of oxidation and lipid synthesis in the liver of obese mice. The short duration protocol was used to avoid changing the amount of adipose tissue. Swiss mice were separated into three groups: lean control (CTL), sedentary obese (OB) and strength training obese (STO). The obese groups were fed a high-fat diet (HFD) and the STO group performed the strength training protocol 1 session/day for 15 days. The short-term strength training reduced hepatic fat accumulation, increasing hepatic insulin sensitivity and controlling hepatic glucose production. The obese animals increased the mRNA of lipogenic genes Fasn and Scd1 and reduced the oxidative genes Cpt1a and Ppara. On the other hand, the STO group presented the opposite results. Finally, the obese animals presented higher levels of lipogenic proteins (ACC and FAS) and proinflammatory cytokines (TNF-α and IL-1β), but the short-term strength training was efficient in reducing this condition, regardless of body weight loss. In conclusion, there was a reduction of obesity-related hepatic lipogenesis and inflammation after short-term strength training, independent of weight loss, leading to improvements in hepatic insulin sensitivity and glycemic homeostasis in obese mice. Key points: (1) Short-term strength training (STST) reduced fat accumulation and inflammation in the liver; (2) Hepatic insulin sensitivity and HPG control were increased with STST; (3) The content and activity of ACC and content of FAS were reduced with STST; (4) STST improved hepatic fat accumulation and glycemic homeostasis; (5) STST effects were observed independently of body weight change.
Collapse
Affiliation(s)
- Rodrigo Martins Pereira
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | | | - Chadi Pellegrini Anaruma
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Marcella Ramos Sant'Ana
- Laboratory of Nutritional Genomics, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | | | - Rodrigo Stellzer Gaspar
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | | | - Diego Gomes de Melo
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Rania A Mekary
- Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Social and Administrative Sciences, School of Pharmacy, MCPHS University, Boston, Massachusetts, USA
| | - Adelino Sanchez Ramos da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, USP, Ribeirão Preto, São Paulo, Brazil
| | - Dennys Esper Cintra
- Laboratory of Nutritional Genomics, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| |
Collapse
|
17
|
Li Y, Yu S, Chen L, Hu X, Zheng J, Deng X. Involvement of PPARγ/FSP27 in the pathogenic mechanism underlying insulin resistance: tipping the balance between lipogenesis and fat storage in adult catch-up growth rats. Nutr Metab (Lond) 2019; 16:11. [PMID: 30792748 PMCID: PMC6371468 DOI: 10.1186/s12986-019-0336-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 01/24/2019] [Indexed: 12/22/2022] Open
Abstract
Background Catch-up growth in adult (CUGA) is characterized by visceral fat accumulation, ectopic lipid deposition and insulin resistance (IR). Here, we investigated the determinants of these pathophysiological consequences of CUGA. Methods Rats were divided into different groups: control rats were offered normal chow ad libitum (AL), while experimental rats were put on 4-week caloric restriction (CR) initially, followed by regaining weight-matched normal chow (RN) in the RN group. General characteristics of lipid metabolism, expression level of genes in visceral adipose tissue (VAT), and glucose infusion rate (GIR60–120) by the hyperinsulinemic-euglycemic clamp were examined. Results After CR, percentage of abdominal fat mass (AFM%) was lower in the RN group than in the AL group but no difference was observed in serum non-esterified fatty acid (NEFA). Expression of fat-specific protein 27 (FSP27) was decreased in the RN group, while the expression of peroxisome proliferator-activated receptors γ (PPARγ), the key lipogenic gene, was increased. After refeeding, AFM% increased over time and serum NEFA persistently elevated in the RN group. Ectopic triglyceride contents were increased whereas insulin sensitivity was impaired. The expression of FSP27 did not follow the increase in the expression of PPARγ. Additionally, we observed a sustained increase in the expression of ATGL and CGI-58 in VAT in the RN group compared with the AL group after CR and refeeding, and a persistent shift-to-the-left of adipocyte size distribution accompanied by enhanced lipogenesis during CUGA. Conclusion The persistent CR-induced imbalance of lipogenesis/fat storage capacity might be responsible for the CUGA-associated metabolic disorders.
Collapse
Affiliation(s)
- Yan Li
- 1Department of Endocrinology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000 People's Republic of China
| | - Shan Yu
- 2Department of anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 People's Republic of China
| | - Lulu Chen
- 3Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 People's Republic of China
| | - Xiang Hu
- 3Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 People's Republic of China
| | - Juan Zheng
- 3Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 People's Republic of China
| | - Xiuling Deng
- 3Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 People's Republic of China
| |
Collapse
|
18
|
Lawitz EJ, Coste A, Poordad F, Alkhouri N, Loo N, McColgan BJ, Tarrant JM, Nguyen T, Han L, Chung C, Ray AS, McHutchison JG, Subramanian GM, Myers RP, Middleton MS, Sirlin C, Loomba R, Nyangau E, Fitch M, Li K, Hellerstein M. Acetyl-CoA Carboxylase Inhibitor GS-0976 for 12 Weeks Reduces Hepatic De Novo Lipogenesis and Steatosis in Patients With Nonalcoholic Steatohepatitis. Clin Gastroenterol Hepatol 2018; 16:1983-1991.e3. [PMID: 29705265 DOI: 10.1016/j.cgh.2018.04.042] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/15/2018] [Accepted: 04/17/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Increased de novo lipogenesis (DNL) contributes to the pathogenesis of nonalcoholic steatohepatitis (NASH). Acetyl-CoA carboxylase catalyzes the rate-limiting step in DNL. We evaluated the safety and efficacy of GS-0976, a small molecule inhibitor of acetyl-CoA carboxylase, in patients with NASH. METHODS In an open-label prospective study, patients with NASH (n = 10) received GS-0976 20 mg orally once daily for 12 weeks. NASH was diagnosed based on a proton density fat fraction estimated by magnetic resonance imaging (MRI-PDFF) ≥10% and liver stiffness by magnetic resonance elastography (MRE) ≥2.88 kPa. The contribution from hepatic DNL to plasma palmitate was measured by 14 days of heavy water labeling before and at the end of treatment. We performed the same labelling protocol in an analysis of healthy volunteers who were not given DNL (controls, n = 10). MRI-PDFF and MRE at baseline, and at weeks 4 and 12 of GS-0976 administration, were measured. We analyzed markers of liver injury and serum markers of fibrosis. RESULTS The contribution of hepatic DNL to plasma palmitate was significantly greater in patients with NASH compared with controls (43% vs 18%) (P = .003). After 12 weeks administration of GS-0976, the median hepatic DNL was reduced 22% from baseline in patients with NASH (P = .004). Compared with baseline, reductions in MRI-PDFF at week 12 (15.7% vs 9.1% at baseline; P = .006), liver stiffness by MRE (3.4 kPa vs 3.1 kPa at baseline; P = .049), TIMP metallopeptidase inhibitor 1 (275 ng/mL vs 244 ng/mL at baseline; P = .049), and serum level of alanine aminotransferase (101 U/L vs 57 U/L at baseline; P = .23) were consistent with decreased hepatic lipid content and liver injury. At week 12, 7 patients (70%) had a ≥30% decrease in MRI-PDFF. CONCLUSION In an open-label study, patients with NASH given GS-0976 for 12 weeks had reduced hepatic DNL, steatosis, and markers of liver injury. ClinicalTrials.gov no: NCT02856555.
Collapse
Affiliation(s)
- Eric J Lawitz
- Texas Liver Institute and University of Texas Health San Antonio, San Antonio, Texas.
| | - Angie Coste
- Texas Liver Institute and University of Texas Health San Antonio, San Antonio, Texas
| | - Fred Poordad
- Texas Liver Institute and University of Texas Health San Antonio, San Antonio, Texas
| | - Naim Alkhouri
- Texas Liver Institute and University of Texas Health San Antonio, San Antonio, Texas
| | - Nicole Loo
- Texas Liver Institute and University of Texas Health San Antonio, San Antonio, Texas
| | | | | | - Tuan Nguyen
- Gilead Sciences, Inc, Foster City, California
| | - Ling Han
- Gilead Sciences, Inc, Foster City, California
| | | | | | | | | | | | | | - Claude Sirlin
- University of California at San Diego, San Diego, California
| | - Rohit Loomba
- University of California at San Diego, San Diego, California
| | - Edna Nyangau
- University of California Berkeley, Berkeley, California
| | - Mark Fitch
- University of California Berkeley, Berkeley, California
| | - Kelvin Li
- University of California Berkeley, Berkeley, California
| | | |
Collapse
|
19
|
Imai N, Cohen DE. Trimming the Fat: Acetyl-CoA Carboxylase Inhibition for the Management of NAFLD. Hepatology 2018; 68:2062-2065. [PMID: 30076622 PMCID: PMC7888551 DOI: 10.1002/hep.30206] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 07/31/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Norihiro Imai
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, USA
| | - David E. Cohen
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, USA
| |
Collapse
|
20
|
Zeng N, Huang R, Li N, Jiang H, Li R, Wang F, Chen W, Xia M, Wang Q. MiR-451a attenuates free fatty acids-mediated hepatocyte steatosis by targeting the thyroid hormone responsive spot 14 gene. Mol Cell Endocrinol 2018; 474:260-271. [PMID: 29604329 DOI: 10.1016/j.mce.2018.03.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 02/06/2023]
Abstract
The thyroid hormone responsive spot 14 (THRSP) gene is a de novo lipogenesis-related gene that plays a significant role in the initiation and development of nonalcoholic fatty liver disease (NAFLD). Several previous studies had shown that endogenous and environmental factors could regulate the expression of THRSP. The role of microRNAs (miRNAs), however, in controlling THRSP expression has not been investigated. In this study, we first constructed the hepatic steatosis cell model by using a mixture of free fatty acids (FFAs; oleate/palmitate, 2:1 ratio) to treat and demonstrate the promotive role of THRSP in lipid accumulation in hepatic cells. By analyzing the photoactivatable ribonucleoside-enhanced crosslinking and immunoprecipitation (PAR-CLIP) database and performing a luciferase reporter assay, we confirmed that microRNA-451a specifically binds to mouse and human THRSP 3'UTR and inhibits its activity. Overexpression of miR-451a efficiently reduced THRSP mRNA and protein expression as well as triglyceride (TG) accumulation in cultured hepatic cells (AML12 and HepG2). Moreover, overexpression of miR-451a significantly decreases TG accumulation in the livers of mice injected with an miR-451a agomir. All these results demonstrated that miR-451a might participate in the FFA-induced hepatic steatosis by regulating the expression of the THRSP gene which represents a new potential target for NAFLD therapy.
Collapse
Affiliation(s)
- Ni Zeng
- Faculty of Preventive Medicine, A Key Laboratory of Guangzhou Environmental Pollution and Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Rong Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health and Department of Nutrition, School of Public Health, Sun Yat-sen University Guangzhou, 510080, China
| | - Nan Li
- Department of Intervention Radiology, The First Affiliated Hospital, Sun Yat-sen University Guangzhou, 510080, China
| | - Hongmei Jiang
- Faculty of Preventive Medicine, A Key Laboratory of Guangzhou Environmental Pollution and Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ruobi Li
- Faculty of Preventive Medicine, A Key Laboratory of Guangzhou Environmental Pollution and Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Fei Wang
- Faculty of Preventive Medicine, A Key Laboratory of Guangzhou Environmental Pollution and Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Wen Chen
- Faculty of Preventive Medicine, A Key Laboratory of Guangzhou Environmental Pollution and Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Min Xia
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health and Department of Nutrition, School of Public Health, Sun Yat-sen University Guangzhou, 510080, China
| | - Qing Wang
- Faculty of Preventive Medicine, A Key Laboratory of Guangzhou Environmental Pollution and Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
21
|
Tang S, Hao Y, Yuan Y, Liu R, Chen Q. Role of fibroblast growth factor receptor 4 in cancer. Cancer Sci 2018; 109:3024-3031. [PMID: 30070748 PMCID: PMC6172014 DOI: 10.1111/cas.13759] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/29/2018] [Accepted: 07/30/2018] [Indexed: 02/05/2023] Open
Abstract
Fibroblast growth factor receptors (FGFR) play a significant role in both embryonic development and in adults. Upon binding with ligands, FGFR signaling is activated and triggers various downstream signal cascades that are implicated in diverse biological processes. Aberrant regulations of FGFR signaling are detected in numerous cancers. Although FGFR4 was discovered later than other FGFR, information on the involvement of FGFR4 in cancers has significantly increased in recent years. In this review, the recent findings in FGFR4 structure, signaling transduction, physiological function, aberrant regulations, and effects in cancers as well as its potential applications as an anticancer therapeutic target are summarized.
Collapse
Affiliation(s)
- Shuya Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yilong Hao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yao Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Takagi H, Ikehara T, Kashiwagi Y, Hashimoto K, Nanchi I, Shimazaki A, Nambu H, Yukioka H. ACC2 Deletion Enhances IMCL Reduction Along With Acetyl-CoA Metabolism and Improves Insulin Sensitivity in Male Mice. Endocrinology 2018; 159:3007-3019. [PMID: 29931154 DOI: 10.1210/en.2018-00338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/15/2018] [Indexed: 12/13/2022]
Abstract
Intramyocellular lipid (IMCL) accumulation in skeletal muscle greatly contributes to lipid-induced insulin resistance. Because acetyl-coenzyme A (CoA) carboxylase (ACC) 2 negatively modulates mitochondrial fatty acid oxidation (FAO) in skeletal muscle, ACC2 inhibition is expected to reduce IMCL via elevation of FAO and to attenuate insulin resistance. However, the concept of substrate competition suggests that enhanced FAO results in reduced glucose use because of an excessive acetyl-CoA pool in mitochondria. To identify how ACC2-regulated FAO affects IMCL accumulation and glucose metabolism, we generated ACC2 knockout (ACC2-/-) mice and investigated skeletal muscle metabolites associated with fatty acid and glucose metabolism, as well as whole-body glucose metabolism. ACC2-/- mice displayed higher capacity of glucose disposal at the whole-body levels. In skeletal muscle, ACC2-/- mice exhibited enhanced acylcarnitine formation and reduced IMCL levels without alteration in glycolytic intermediate levels. Notably, these changes were accompanied by decreased acetyl-CoA content and enhanced mitochondrial pathways related to acetyl-CoA metabolism, such as the acetylcarnitine production and tricarboxylic acid cycle. Furthermore, ACC2-/- mice exhibited lower levels of IMCL and acetyl-CoA even under HFD conditions and showed protection against HFD-induced insulin resistance. Our findings suggest that ACC2 deletion leads to IMCL reduction without suppressing glucose use via an elevation in acetyl-CoA metabolism even under HFD conditions and offer new mechanistic insight into the therapeutic potential of ACC2 inhibition on insulin resistance.
Collapse
Affiliation(s)
- Hiroyuki Takagi
- Drug Discovery and Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Tatsuya Ikehara
- Biomarker Research and Development Department, Shionogi & Co., Ltd., Osaka, Japan
| | - Yuto Kashiwagi
- Biomarker Research and Development Department, Shionogi & Co., Ltd., Osaka, Japan
| | - Kumi Hashimoto
- Drug Discovery and Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Isamu Nanchi
- Drug Discovery and Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Atsuyuki Shimazaki
- Drug Discovery and Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Hirohide Nambu
- Drug Discovery and Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Hideo Yukioka
- Drug Discovery and Disease Research Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| |
Collapse
|
23
|
Jang S, Gornicki P, Marjanovic J, Bass E, P Iurcotta T, Rodriguez P, Austin J, Haselkorn R. Activity and structure of human acetyl-CoA carboxylase targeted by a specific inhibitor. FEBS Lett 2018; 592:2048-2058. [PMID: 29772612 DOI: 10.1002/1873-3468.13097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 01/01/2023]
Abstract
We have studied a series of human acetyl-CoA carboxylase (ACC) 1 and ACC2 proteins with deletions and/or Ser to Ala substitutions of the known phosphorylation sites. In vitro dephosphorylation/phosphorylation experiments reveal a substantial level of phosphorylation of human ACCs produced in insect cells. Our results are consistent with AMPK phosphorylation of Ser29 , Ser80 , Ser1,201 , and Ser1,216 . Phosphorylation of the N-terminal regulatory domain decreases ACC1 activity, while phosphorylation of residues in the ACC central domain has no effect. Inhibition of the activity by phosphorylation is significantly more profound at citrate concentrations below 2 mm. Furthermore, deletion of the N-terminal domain facilitates structural changes induced by citrate, including conversion of ACC dimers to linear polymers. We have also identified ACC2 amino acid mutations affecting specific inhibition of the isozyme by compound CD-017-0191. They form two clusters separated by 60-90 Å: one located in the vicinity of the BC active site and the other one in the vicinity of the ACC1 phosphorylation sites in the central domain, suggesting a contribution of the interface of two ACC dimers in the polymer to the inhibitor binding site.
Collapse
Affiliation(s)
- SoRi Jang
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Piotr Gornicki
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Jasmina Marjanovic
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Ethan Bass
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Toni P Iurcotta
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Pedro Rodriguez
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Jotham Austin
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Robert Haselkorn
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
24
|
Organic cation transporter 1 (OCT1) modulates multiple cardiometabolic traits through effects on hepatic thiamine content. PLoS Biol 2018; 16:e2002907. [PMID: 29659562 PMCID: PMC5919692 DOI: 10.1371/journal.pbio.2002907] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 04/26/2018] [Accepted: 03/20/2018] [Indexed: 01/07/2023] Open
Abstract
A constellation of metabolic disorders, including obesity, dysregulated lipids, and elevations in blood glucose levels, has been associated with cardiovascular disease and diabetes. Analysis of data from recently published genome-wide association studies (GWAS) demonstrated that reduced-function polymorphisms in the organic cation transporter, OCT1 (SLC22A1), are significantly associated with higher total cholesterol, low-density lipoprotein (LDL) cholesterol, and triglyceride (TG) levels and an increased risk for type 2 diabetes mellitus, yet the mechanism linking OCT1 to these metabolic traits remains puzzling. Here, we show that OCT1, widely characterized as a drug transporter, plays a key role in modulating hepatic glucose and lipid metabolism, potentially by mediating thiamine (vitamin B1) uptake and hence its levels in the liver. Deletion of Oct1 in mice resulted in reduced activity of thiamine-dependent enzymes, including pyruvate dehydrogenase (PDH), which disrupted the hepatic glucose–fatty acid cycle and shifted the source of energy production from glucose to fatty acids, leading to a reduction in glucose utilization, increased gluconeogenesis, and altered lipid metabolism. In turn, these effects resulted in increased total body adiposity and systemic levels of glucose and lipids. Importantly, wild-type mice on thiamine deficient diets (TDs) exhibited impaired glucose metabolism that phenocopied Oct1 deficient mice. Collectively, our study reveals a critical role of hepatic thiamine deficiency through OCT1 deficiency in promoting the metabolic inflexibility that leads to the pathogenesis of cardiometabolic disease. The liver is the major organ for glucose and lipid metabolism; impairment in liver energy metabolism is often found in metabolic disorders. Traditionally, excesses in macronutrients (fat and glucose) are linked to the development of metabolic disorders. Our study provides evidence that imbalances in a micronutrient, vitamin B1 (thiamine), can serve as an etiological cause of lipid and glucose disorders and implicates the organic cation transporter, OCT1, in these disorders. OCT1 is a key determinant of thiamine levels in the liver. In humans, reduced-function polymorphisms of OCT1 significantly associate with high LDL cholesterol levels. Using Oct1 knockout mice, we show that reduced OCT1-mediated thiamine uptake in the liver leads to reduced levels of TPP—the active metabolite of thiamine—and decreased activity of key TPP-dependent enzymes. As a result, a shift from glucose to fatty acid oxidation occurs, leading to imbalances in key metabolic intermediates, alterations in metabolic flux pathways, and disruptions of various metabolic regulatory mechanisms. The extensive characterization of Oct1 knockout mice provides evidence for the molecular mechanisms responsible for various metabolic traits and indicates an important role for imbalances in micronutrients in cardiometabolic disorders.
Collapse
|
25
|
Samuel VT, Shulman GI. Nonalcoholic Fatty Liver Disease as a Nexus of Metabolic and Hepatic Diseases. Cell Metab 2018; 27:22-41. [PMID: 28867301 PMCID: PMC5762395 DOI: 10.1016/j.cmet.2017.08.002] [Citation(s) in RCA: 511] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/01/2017] [Accepted: 08/01/2017] [Indexed: 12/15/2022]
Abstract
NAFLD is closely linked with hepatic insulin resistance. Accumulation of hepatic diacylglycerol activates PKC-ε, impairing insulin receptor activation and insulin-stimulated glycogen synthesis. Peripheral insulin resistance indirectly influences hepatic glucose and lipid metabolism by increasing flux of substrates that promote lipogenesis (glucose and fatty acids) and gluconeogenesis (glycerol and fatty acid-derived acetyl-CoA, an allosteric activator of pyruvate carboxylase). Weight loss with diet or bariatric surgery effectively treats NAFLD, but drugs specifically approved for NAFLD are not available. Some new pharmacological strategies act broadly to alter energy balance or influence pathways that contribute to NAFLD (e.g., agonists for PPAR γ, PPAR α/δ, FXR and analogs for FGF-21, and GLP-1). Others specifically inhibit key enzymes involved in lipid synthesis (e.g., mitochondrial pyruvate carrier, acetyl-CoA carboxylase, stearoyl-CoA desaturase, and monoacyl- and diacyl-glycerol transferases). Finally, a novel class of liver-targeted mitochondrial uncoupling agents increases hepatocellular energy expenditure, reversing the metabolic and hepatic complications of NAFLD.
Collapse
Affiliation(s)
- Varman T Samuel
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06510, USA; Veterans Affairs Medical Center, West Haven, CT 06516, USA.
| | - Gerald I Shulman
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
26
|
Hampe L, Xu C, Harris PWR, Chen J, Liu M, Middleditch M, Radjainia M, Wang Y, Mitra AK. Synthetic peptides designed to modulate adiponectin assembly improve obesity-related metabolic disorders. Br J Pharmacol 2017; 174:4478-4492. [PMID: 28945274 DOI: 10.1111/bph.14050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/21/2017] [Accepted: 09/07/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Adiponectin, an adipokine possessing profound insulin-sensitizing and anti-inflammatory properties, is a potent biotherapeutic agent . The trimeric adiponectin subunit assembles into hexameric and functionally important higher molecular weight (HMW) forms, controlled by the endoplasmic reticulum protein 44 (ERp44). Obesity-induced ER stress decreases the HMW form in serum, contributing to the development of insulin resistance and Type 2 diabetes. In this study, a panel of synthetic peptides, designed to target ERp44-adiponectin interactions, were tested for their effects on circulating levels of HMW adiponectin. EXPERIMENTAL APPROACH Peptides derived from the ERp44 binding region of adiponectin and immunoglobulin IgM were synthesized with or without a cell-penetrating sequence. Cultures of 3T3-L1 adipocytes were incubated with the peptides for assessing the assembly and secretion of HMW adiponectin. Mice given standard chow or a high-fat diet were treated acutely or chronically, with the peptides to investigate the therapeutic effects on insulin sensitivity and energy metabolism. RESULTS The designed peptides interfered with ERp44-adiponectin interactions and modulated adiponectin assembly and release from adipocytes. In particular, IgM-derived peptides facilitated the release of endogenous adiponectin (especially the HMW form) from adipose tissue, enhanced its circulating level and the ratio of HMW-to-total-adiponectin in obese mice. Long-term treatment of mice fed with high-fat diet by IgM-derived peptides reduced the circulating lipid levels and improved insulin sensitivity. CONCLUSIONS AND IMPLICATIONS Targeting ERp44-adiponectin interactions with short peptides represents an effective strategy to treat of obesity-related metabolic disorders, such as insulin resistance and Type 2 diabetes.
Collapse
Affiliation(s)
- Lutz Hampe
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Cheng Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong
| | - Paul W R Harris
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Jie Chen
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong
| | - Ming Liu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong
| | - Martin Middleditch
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Mazdak Radjainia
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong
| | - Alok K Mitra
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
27
|
Scott F, Gonzalez Malagon SG, O'Brien BA, Fennema D, Veeravalli S, Coveney CR, Phillips IR, Shephard EA. Identification of Flavin-Containing Monooxygenase 5 (FMO5) as a Regulator of Glucose Homeostasis and a Potential Sensor of Gut Bacteria. Drug Metab Dispos 2017; 45:982-989. [PMID: 28646079 PMCID: PMC5539585 DOI: 10.1124/dmd.117.076612] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/13/2017] [Indexed: 11/24/2022] Open
Abstract
We have previously identified flavin-containing monooxygenase 5 (FMO5) as a regulator of metabolic aging. The aim of the present study was to investigate the role of FMO5 in glucose homeostasis and the impact of diet and gut flora on the phenotype of mice in which the Fmo5 gene has been disrupted (Fmo5-/- mice). In comparison with wild-type (WT) counterparts, Fmo5-/- mice are resistant to age-related changes in glucose homeostasis and maintain the higher glucose tolerance and insulin sensitivity characteristic of young animals. When fed a high-fat diet, they are protected against weight gain and reduction of insulin sensitivity. The phenotype of Fmo5-/- mice is independent of diet and the gut microbiome and is determined solely by the host genotype. Fmo5-/- mice have metabolic characteristics similar to those of germ-free mice, indicating that FMO5 plays a role in sensing or responding to gut bacteria. In WT mice, FMO5 is present in the mucosal epithelium of the gastrointestinal tract where it is induced in response to a high-fat diet. In comparison with WT mice, Fmo5-/- mice have fewer colonic goblet cells, and they differ in the production of the colonic hormone resistin-like molecule βFmo5-/- mice have lower concentrations of tumor necrosis factor α in plasma and of complement component 3 in epididymal white adipose tissue, indicative of improved inflammatory tone. Our results implicate FMO5 as a regulator of body weight and of glucose disposal and insulin sensitivity and, thus, identify FMO5 as a potential novel therapeutic target for obesity and insulin resistance.
Collapse
Affiliation(s)
- Flora Scott
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Sandra G Gonzalez Malagon
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Brett A O'Brien
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Diede Fennema
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Sunil Veeravalli
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Clarissa R Coveney
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Ian R Phillips
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Elizabeth A Shephard
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| |
Collapse
|
28
|
Genomic structural variations for cardiovascular and metabolic comorbidity. Sci Rep 2017; 7:41268. [PMID: 28120895 PMCID: PMC5264603 DOI: 10.1038/srep41268] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 12/19/2016] [Indexed: 12/19/2022] Open
Abstract
The objective of this study was to identify genes targeted by both copy number and copy-neutral changes in the right coronary arteries in the area of advanced atherosclerotic plaques and intact internal mammary arteries derived from the same individuals with comorbid coronary artery disease and metabolic syndrome. The artery samples from 10 patients were screened for genomic imbalances using array comparative genomic hybridization. Ninety high-confidence, identical copy number variations (CNVs) were detected. We also identified eight copy-neutral changes (cn-LOHs) > 1.5 Mb in paired arterial samples in 4 of 10 individuals. The frequencies of the two gains located in the 10q24.31 (ERLIN1) and 12q24.11 (UNG, ACACB) genomic regions were evaluated in 33 paired arteries and blood samples. Two patients contained the gain in 10q24.31 (ERLIN1) and one patient contained the gain in 12q24.11 (UNG, ACACB) that affected only the blood DNA. An additional two patients harboured these CNVs in both the arteries and blood. In conclusion, we discovered and confirmed a gain of the 10q24.31 (ERLIN1) and 12q24.11 (UNG, ACACB) genomic regions in patients with coronary artery disease and metabolic comorbidity. Analysis of DNA extracted from blood indicated a possible somatic origin for these CNVs.
Collapse
|
29
|
Alrob OA, Khatib S, Naser SA. MicroRNAs 33, 122, and 208: a potential novel targets in the treatment of obesity, diabetes, and heart-related diseases. J Physiol Biochem 2016; 73:307-314. [PMID: 27966196 DOI: 10.1007/s13105-016-0543-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/06/2016] [Indexed: 12/17/2022]
Abstract
Despite decades of research, obesity and diabetes remain major health problems in the USA and worldwide. Among the many complications associated with diabetes is an increased risk of cardiovascular diseases, including myocardial infarction and heart failure. Recently, microRNAs have emerged as important players in heart disease and energy regulation. However, little work has investigated the role of microRNAs in cardiac energy regulation. Both human and animal studies have reported a significant increase in circulating free fatty acids and triacylglycerol, increased cardiac reliance on fatty acid oxidation, and subsequent decrease in glucose oxidation which all contributes to insulin resistance and lipotoxicity seen in obesity and diabetes. Importantly, MED13 was initially identified as a negative regulator of lipid accumulation in Drosophilia. Various metabolic genes were downregulated in MED13 transgenic heart, including sterol regulatory element-binding protein. Moreover, miR-33 and miR-122 have recently revealed as key regulators of lipid metabolism. In this review, we will focus on the role of microRNAs in regulation of cardiac and total body energy metabolism. We will also discuss the pharmacological and non-pharmacological interventions that target microRNAs for the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Osama Abo Alrob
- Faculty of Pharmacy, Yarmouk University, P.O Box 566, Irbid, 21163, Jordan.
| | - Said Khatib
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Saleh A Naser
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
30
|
Cheng J, Zhang T, Ji H, Tao K, Guo J, Wei W. Functional characterization of AMP-activated protein kinase signaling in tumorigenesis. Biochim Biophys Acta Rev Cancer 2016; 1866:232-251. [PMID: 27681874 DOI: 10.1016/j.bbcan.2016.09.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022]
Abstract
AMP-activated protein kinase (AMPK) is a ubiquitously expressed metabolic sensor among various species. Specifically, cellular AMPK is phosphorylated and activated under certain stressful conditions, such as energy deprivation, in turn to activate diversified downstream substrates to modulate the adaptive changes and maintain metabolic homeostasis. Recently, emerging evidences have implicated the potential roles of AMPK signaling in tumor initiation and progression. Nevertheless, a comprehensive description on such topic is still in scarcity, especially in combination of its biochemical features with mouse modeling results to elucidate the physiological role of AMPK signaling in tumorigenesis. Hence, we performed this thorough review by summarizing the tumorigenic role of each component along the AMPK signaling, comprising of both its upstream and downstream effectors. Moreover, their functional interplay with the AMPK heterotrimer and exclusive efficacies in carcinogenesis were chiefly explained among genetically altered mice models. Importantly, the pharmaceutical investigations of AMPK relevant medications have also been highlighted. In summary, in this review, we not only elucidate the potential functions of AMPK signaling pathway in governing tumorigenesis, but also potentiate the future targeted strategy aiming for better treatment of aberrant metabolism-associated diseases, including cancer.
Collapse
Affiliation(s)
- Ji Cheng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Tao Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hongbin Ji
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai 200031, People's Republic of China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China.
| | - Jianping Guo
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
31
|
Biotin augments acetyl CoA carboxylase 2 gene expression in the hypothalamus, leading to the suppression of food intake in mice. Biochem Biophys Res Commun 2016; 476:134-9. [DOI: 10.1016/j.bbrc.2016.04.152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 04/27/2016] [Indexed: 01/19/2023]
|
32
|
Mahmood S, Birkaya B, Rideout TC, Patel MS. Lack of mitochondria-generated acetyl-CoA by pyruvate dehydrogenase complex downregulates gene expression in the hepatic de novo lipogenic pathway. Am J Physiol Endocrinol Metab 2016; 311:E117-27. [PMID: 27166281 PMCID: PMC4967143 DOI: 10.1152/ajpendo.00064.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/03/2016] [Indexed: 12/20/2022]
Abstract
During the absorptive state, the liver stores excess glucose as glycogen and synthesizes fatty acids for triglyceride synthesis for export as very low density lipoproteins. For de novo synthesis of fatty acids from glucose, the mitochondrial pyruvate dehydrogenase complex (PDC) is the gatekeeper for the generation of acetyl-CoA from glucose-derived pyruvate. Here, we tested the hypothesis that limiting the supply of PDC-generated acetyl-CoA from glucose would have an impact on expression of key genes in the lipogenic pathway. In the present study, although the postnatal growth of liver-specific PDC-deficient (L-PDCKO) male mice was largely unaltered, the mice developed hyperinsulinemia with lower blood glucose levels in the fed state. Serum and liver lipid triglyceride and cholesterol levels remained unaltered in L-PDCKO mice. Expression of several key genes (ACL, ACC1) in the lipogenic pathway and their upstream regulators (LXR, SREBP1, ChREBP) as well as several genes in glucose metabolism (Pklr, G6pd2, Pck1) and fatty acid oxidation (FAT, Cpt1a) was downregulated in livers from L-PDCKO mice. Interestingly, there was concomitant upregulation of lipogenic genes in adipose tissue from L-PDCKO mice. Although, the total hepatic acetyl-CoA content remained unaltered in L-PDCKO mice, modified acetylation profiles of proteins in the nuclear compartment suggested an important role for PDC-generated acetyl-CoA in gene expression in de novo fatty acid synthesis in the liver. This finding has important implications for the regulation of hepatic lipid synthesis in pathological states.
Collapse
Affiliation(s)
- Saleh Mahmood
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, and
| | - Barbara Birkaya
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, and
| | - Todd C Rideout
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| | - Mulchand S Patel
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, and
| |
Collapse
|
33
|
Softic S, Cohen DE, Kahn CR. Role of Dietary Fructose and Hepatic De Novo Lipogenesis in Fatty Liver Disease. Dig Dis Sci 2016; 61:1282-93. [PMID: 26856717 PMCID: PMC4838515 DOI: 10.1007/s10620-016-4054-0] [Citation(s) in RCA: 462] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 01/21/2016] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a liver manifestation of metabolic syndrome. Overconsumption of high-fat diet (HFD) and increased intake of sugar-sweetened beverages are major risk factors for development of NAFLD. Today the most commonly consumed sugar is high fructose corn syrup. Hepatic lipids may be derived from dietary intake, esterification of plasma free fatty acids (FFA) or hepatic de novo lipogenesis (DNL). A central abnormality in NAFLD is enhanced DNL. Hepatic DNL is increased in individuals with NAFLD, while the contribution of dietary fat and plasma FFA to hepatic lipids is not significantly altered. The importance of DNL in NAFLD is further established in mouse studies with knockout of genes involved in this process. Dietary fructose increases levels of enzymes involved in DNL even more strongly than HFD. Several properties of fructose metabolism make it particularly lipogenic. Fructose is absorbed via portal vein and delivered to the liver in much higher concentrations as compared to other tissues. Fructose increases protein levels of all DNL enzymes during its conversion into triglycerides. Additionally, fructose supports lipogenesis in the setting of insulin resistance as fructose does not require insulin for its metabolism, and it directly stimulates SREBP1c, a major transcriptional regulator of DNL. Fructose also leads to ATP depletion and suppression of mitochondrial fatty acid oxidation, resulting in increased production of reactive oxygen species. Furthermore, fructose promotes ER stress and uric acid formation, additional insulin independent pathways leading to DNL. In summary, fructose metabolism supports DNL more strongly than HFD and hepatic DNL is a central abnormality in NAFLD. Disrupting fructose metabolism in the liver may provide a new therapeutic option for the treatment of NAFLD.
Collapse
Affiliation(s)
- Samir Softic
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, One Joslin Place, Boston, MA, 02215, USA
- Department of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - David E Cohen
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - C Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, One Joslin Place, Boston, MA, 02215, USA.
| |
Collapse
|
34
|
Bertini V, Orsini A, Bonuccelli A, Cambi F, Del Pistoia M, Vannozzi I, Toschi B, Saggese G, Simi P, Valetto A. 17q12 microduplications: a challenge for clinicians. Am J Med Genet A 2016; 167A:674-6. [PMID: 25691423 DOI: 10.1002/ajmg.a.36905] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 11/14/2014] [Indexed: 11/10/2022]
Abstract
In the recent years, some cases of 17q12 deletions and duplications have been reported, but the clinical impact of these imbalances is still to be fully elucidated. In particular, 17q12 duplications elude syndrome classification, since they are associated with a wide phenotypic spectrum, ranging from very mild to quite severe phenotypes. Here, two unrelated patients with the same 1.2 Mb microduplication of 17q12 are reported. Comparing these patients' phenotype with those previously published, it emerges that the more patients reported, the more difficult is finding common characteristics, even in presence of exactly the same genetic anomaly. The role of the genes duplicated in this region and the impact of this chromosomal imbalance are discussed.
Collapse
Affiliation(s)
- V Bertini
- Cytogenetics and Molecular Genetics Unit, Children Department, AOUP, Pisa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Dai W, Panserat S, Kaushik S, Terrier F, Plagnes-Juan E, Seiliez I, Skiba-Cassy S. Hepatic fatty acid biosynthesis is more responsive to protein than carbohydrate in rainbow trout during acute stimulations. Am J Physiol Regul Integr Comp Physiol 2016; 310:R74-86. [DOI: 10.1152/ajpregu.00281.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 10/20/2015] [Indexed: 12/21/2022]
Abstract
The link between dietary carbohydrate/protein and de novo lipogenesis (DNL) remains debatable in carnivorous fish. We aimed to evaluate and compare the response of hepatic lipogenic gene expression to dietary carbohydrate intake/glucose and dietary protein intake/amino acids (AAs) during acute stimulations using both in vivo and in vitro approaches. For the in vivo trial, three different diets and a controlled-feeding method were employed to supply fixed amount of dietary protein or carbohydrate in a single meal; for the in vitro trial, primary hepatocytes were stimulated with a low or high level of glucose (3 mM or 20 mM) and a low or high level of AAs (one-fold or four-fold concentrated AAs). In vitro data showed that a high level of AAs upregulated the expression of enzymes involved in DNL [fatty acid synthase (FAS) and ATP citrate lyase (ACLY)], lipid bioconversion [elongation of very long chain fatty acids like-5 (Elovl5), Elovl2, Δ6 fatty acyl desaturase (D6D) and stearoyl-CoA desaturase-1 (SCD1)], NADPH production [glucose-6-phosphate dehydrogenase (G6PDH) and malic enzyme (ME)], and transcriptional factor sterol regulatory element binding protein 1-like, while a high level of glucose only elevated the expression of ME. Data in trout liver also showed that high dietary protein intake induced higher lipogenic gene expression (FAS, ACLY, and Elovl2) regardless of dietary carbohydrate intake, while high carbohydrate intake markedly suppressed the expression of acetyl-CoA carboxylase (ACC) and Elovl5. Overall, we conclude that, unlike rodents or humans, hepatic fatty acid biosynthetic gene expression in rainbow trout is more responsive to dietary protein intake/AAs than dietary carbohydrate intake/glucose during acute stimulations. This discrepancy probably represents one important physiological and metabolic difference between carnivores and omnivores.
Collapse
Affiliation(s)
- Weiwei Dai
- Institut National de la Recherche Agronomique, UR 1067 Nutrition Métabolisme, Aquaculture, Aquapôle, CD 918, Saint-Pée-sur-Nivelle, France
| | - Stéphane Panserat
- Institut National de la Recherche Agronomique, UR 1067 Nutrition Métabolisme, Aquaculture, Aquapôle, CD 918, Saint-Pée-sur-Nivelle, France
| | - Sadasivam Kaushik
- Institut National de la Recherche Agronomique, UR 1067 Nutrition Métabolisme, Aquaculture, Aquapôle, CD 918, Saint-Pée-sur-Nivelle, France
| | - Frédéric Terrier
- Institut National de la Recherche Agronomique, UR 1067 Nutrition Métabolisme, Aquaculture, Aquapôle, CD 918, Saint-Pée-sur-Nivelle, France
| | - Elisabeth Plagnes-Juan
- Institut National de la Recherche Agronomique, UR 1067 Nutrition Métabolisme, Aquaculture, Aquapôle, CD 918, Saint-Pée-sur-Nivelle, France
| | - Iban Seiliez
- Institut National de la Recherche Agronomique, UR 1067 Nutrition Métabolisme, Aquaculture, Aquapôle, CD 918, Saint-Pée-sur-Nivelle, France
| | - Sandrine Skiba-Cassy
- Institut National de la Recherche Agronomique, UR 1067 Nutrition Métabolisme, Aquaculture, Aquapôle, CD 918, Saint-Pée-sur-Nivelle, France
| |
Collapse
|
36
|
Castro C, Freitag J, Berod L, Lochner M, Sparwasser T. Microbe-associated immunomodulatory metabolites: Influence on T cell fate and function. Mol Immunol 2015; 68:575-84. [DOI: 10.1016/j.molimm.2015.07.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 06/29/2015] [Accepted: 07/21/2015] [Indexed: 01/30/2023]
|
37
|
Biotin-dependent functions in adiposity: a study of monozygotic twin pairs. Int J Obes (Lond) 2015; 40:788-95. [PMID: 26601567 DOI: 10.1038/ijo.2015.237] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/16/2015] [Accepted: 11/06/2015] [Indexed: 12/28/2022]
|
38
|
Huang T, Sun J, Wang Q, Gao J, Liu Y. Synthesis, Biological Evaluation and Molecular Docking Studies of Piperidinylpiperidines and Spirochromanones Possessing Quinoline Moieties as Acetyl-CoA Carboxylase Inhibitors. Molecules 2015; 20:16221-34. [PMID: 26370948 PMCID: PMC6332354 DOI: 10.3390/molecules200916221] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 08/30/2015] [Accepted: 09/01/2015] [Indexed: 01/22/2023] Open
Abstract
Acetyl-coenzyme A carboxylases (ACCs) play critical roles in the regulation of fatty acid metabolism and have been targeted for the development of drugs against obesity, diabetes and other metabolic diseases. Two series of compounds possessing quinoline moieties were designed, synthesized and evaluated for their potential to inhibit acetyl-CoA carboxylases. Most compounds showed moderate to good ACC inhibitory activities and compound 7a possessed the most potent biological activities against ACC1 and ACC2, with IC50 values of 189 nM and 172 nM, respectively, comparable to the positive control. Docking simulation was performed to position compound 7a into the active site of ACC to determine a probable binding model.
Collapse
Affiliation(s)
- Tonghui Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical College, Xuzhou 221004, Jiangsu, China.
| | - Jie Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical College, Xuzhou 221004, Jiangsu, China.
| | - Qianqian Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical College, Xuzhou 221004, Jiangsu, China.
| | - Jian Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical College, Xuzhou 221004, Jiangsu, China.
| | - Yi Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical College, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
39
|
Cordonier EL, Adjam R, Teixeira DC, Onur S, Zbasnik R, Read PE, Döring F, Schlegel VL, Zempleni J. Resveratrol compounds inhibit human holocarboxylase synthetase and cause a lean phenotype in Drosophila melanogaster. J Nutr Biochem 2015; 26:1379-84. [PMID: 26303405 DOI: 10.1016/j.jnutbio.2015.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/22/2015] [Accepted: 07/08/2015] [Indexed: 02/06/2023]
Abstract
Holocarboxylase synthetase (HLCS) is the sole protein-biotin ligase in the human proteome. HLCS has key regulatory functions in intermediary metabolism, including fatty acid metabolism, and in gene repression through epigenetic mechanisms. The objective of this study was to identify food-borne inhibitors of HLCS that alter HLCS-dependent pathways in metabolism and gene regulation. When libraries of extracts from natural products and chemically pure compounds were screened for HLCS inhibitor activity, resveratrol compounds in grape materials caused an HLCS inhibition of >98% in vitro. The potency of these compounds was piceatannol>resveratrol>piceid. Grape-borne compounds other than resveratrol metabolites also contributed toward HLCS inhibition, e.g., p-coumaric acid and cyanidin chloride. HLCS inhibitors had meaningful effects on body fat mass. When Drosophila melanogaster brummer mutants, which are genetically predisposed to storing excess amounts of lipids, were fed diets enriched with grape leaf extracts and piceid, body fat mass decreased by more than 30% in males and females. However, Drosophila responded to inhibitor treatment with an increase in the expression of HLCS, which elicited an increase in the abundance of biotinylated carboxylases in vivo. We conclude that mechanisms other than inhibition of HLCS cause body fat loss in flies. We propose that the primary candidate is the inhibition of the insulin receptor/Akt signaling pathway.
Collapse
Affiliation(s)
- Elizabeth L Cordonier
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, 316 Ruth Leverton Hall, Lincoln, NE 68583-0806, USA
| | - Riem Adjam
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, 316 Ruth Leverton Hall, Lincoln, NE 68583-0806, USA
| | - Daniel Camara Teixeira
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, 316 Ruth Leverton Hall, Lincoln, NE 68583-0806, USA
| | - Simone Onur
- Abteilung Molekulare Prävention, Institut für Humanernährung und Lebensmittelkunde, Universität Kiel, Heinrich-Hecht-Platz 10, 24118 Kiel, Germany
| | - Richard Zbasnik
- Department of Food Science and Technology, University of Nebraska-Lincoln, 326 Filley Hall, Lincoln, NE 68583-0806, USA
| | - Paul E Read
- Department of Agronomy, University of Nebraska-Lincoln, 377 Plant Science Hall, Lincoln, NE 68583-0724, USA
| | - Frank Döring
- Abteilung Molekulare Prävention, Institut für Humanernährung und Lebensmittelkunde, Universität Kiel, Heinrich-Hecht-Platz 10, 24118 Kiel, Germany
| | - Vicki L Schlegel
- Department of Food Science and Technology, University of Nebraska-Lincoln, 326 Filley Hall, Lincoln, NE 68583-0806, USA
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, 316 Ruth Leverton Hall, Lincoln, NE 68583-0806, USA.
| |
Collapse
|
40
|
Valsangkar DS, Downs SM. Acetyl CoA carboxylase inactivation and meiotic maturation in mouse oocytes. Mol Reprod Dev 2015; 82:679-93. [PMID: 26043180 DOI: 10.1002/mrd.22505] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 05/09/2015] [Indexed: 12/24/2022]
Abstract
In mouse oocytes, meiotic induction by pharmacological activation of PRKA (adenosine monophosphate-activated protein kinase; formerly known as AMPK) or by hormones depends on stimulation of fatty acid oxidation (FAO). PRKA stimulates FAO by phosphorylating and inactivating acetyl CoA carboxylase (ACAC; formerly ACC), leading to decreased malonyl CoA levels and augmenting fatty-acid transport into mitochondria. We investigated a role for ACAC inactivation in meiotic resumption by testing the effect of two ACAC inhibitors, CP-640186 and Soraphen A, on mouse oocytes maintained in meiotic arrest in vitro. These inhibitors significantly stimulated the resumption of meiosis in arrested cumulus cell-enclosed oocytes, denuded oocytes, and follicle-enclosed oocytes. This stimulation was accompanied by an increase in FAO. Etomoxir, a malonyl CoA analogue, prevented meiotic resumption as well as the increase in FAO induced by ACAC inhibition. Citrate, an ACAC activator, and CBM-301106, an inhibitor of malonyl CoA decarboxylase, which converts malonyl CoA to acetyl CoA, suppressed both meiotic induction and FAO induced by follicle-stimulating hormone, presumably by maintaining elevated malonyl CoA levels. Mouse oocyte-cumulus cell complexes contain both isoforms of ACAC (ACACA and ACACB); when wild-type and Acacb(-/-) oocytes characteristics were compared, we found that these single-knockout oocytes showed a significantly higher FAO level and a reduced ability to maintain meiotic arrest, resulting in higher rates of germinal vesicle breakdown. Collectively, these data support the model that ACAC inactivation contributes to the maturation-promoting activity of PRKA through stimulation of FAO.
Collapse
Affiliation(s)
- Deepa S Valsangkar
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| | - Stephen M Downs
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| |
Collapse
|
41
|
Lee YA, Wallace MC, Friedman SL. Pathobiology of liver fibrosis: a translational success story. Gut 2015; 64:830-41. [PMID: 25681399 PMCID: PMC4477794 DOI: 10.1136/gutjnl-2014-306842] [Citation(s) in RCA: 682] [Impact Index Per Article: 68.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/07/2015] [Indexed: 12/12/2022]
Abstract
Reversibility of hepatic fibrosis and cirrhosis following antiviral therapy for hepatitis B or C has advanced the prospect of developing antifibrotic therapies for patients with chronic liver diseases, especially non-alcoholic steatohepatitis. Mechanisms of fibrosis have focused on hepatic stellate cells, which become fibrogenic myofibroblasts during injury through 'activation', and are at the nexus of efforts to define novel drug targets. Recent studies have clarified pathways of stellate cell gene regulation and epigenetics, emerging pathways of fibrosis regression through the recruitment and amplification of fibrolytic macrophages, nuanced responses of discrete inflammatory cell subsets and the identification of the 'ductular reaction' as a marker of severe injury and repair. Based on our expanded knowledge of fibrosis pathogenesis, attention is now directed towards strategies for antifibrotic therapies and regulatory challenges for conducting clinical trials with these agents. New therapies are attempting to: 1) Control or cure the primary disease or reduce tissue injury; 2) Target receptor-ligand interactions and intracellular signaling; 3) Inhibit fibrogenesis; and 4) Promote resolution of fibrosis. Progress is urgently needed in validating non-invasive markers of fibrosis progression and regression that can supplant biopsy and shorten the duration of clinical trials. Both scientific and clinical challenges remain, however the past three decades of steady progress in understanding liver fibrosis have contributed to an emerging translational success story, with realistic hopes for antifibrotic therapies to treat patients with chronic liver disease in the near future.
Collapse
Affiliation(s)
- Youngmin A Lee
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael C Wallace
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
42
|
Oh GS, Lee GG, Yoon J, Oh WK, Kim SW. Selective inhibition of liver X receptor α-mediated lipogenesis in primary hepatocytes by licochalcone A. Chin Med 2015; 10:8. [PMID: 25937827 PMCID: PMC4416341 DOI: 10.1186/s13020-015-0037-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 04/13/2015] [Indexed: 11/24/2022] Open
Abstract
Background Sterol regulatory element binding protein-1c (SREBP-1c) is a regulator of the lipogenic pathway and is transcriptionally activated by liver X receptor α (LXRα). This study aims to investigate phytochemicals inhibiting the autonomous transactivity of LXRα with potentials as SREBP-1c inhibitors. Licochalcone A (LicA) is a flavonoid isolated from licorice root of Glycyrrhiza plant. Methods The effects of 238 natural chemicals on autonomous transactivity of LXRα were determined by the Gal4-TK-luciferase reporter system. The inclusion criteria for chemical selection was significant (P < 0.05) inhibition of autonomous transactivity of LXRα from three independent experiments. Transcript levels of mouse primary hepatocytes were measured by conventional or quantitative RT-PCR. Luciferase assay was used to assess synthetic or natural promoter activities of LXRα target genes. The effect of LicA on lipogenic activity was evaluated by measuring cellular triglycerides in mouse primary hepatocytes. The recruitment of RNA polymerase II to the LXR response element (LXRE) region was examined by chromatin immunoprecipitation. Results Among 238 natural compounds, LicA considerably inhibited the autonomous transactivity of LXRα and decreased the LXRα-dependent expression of SREBP-1c. LicA inhibited not only LXRα-dependent activation of the synthetic LXRE promoter but also that of the natural SREBP-1c promoter. As a consequence, LicA reduced the LXRα agonist-stimulated transcription of several lipogenic genes. Furthermore, LXRα-dependent hepatic lipid accumulation was repressed by LicA in mouse primary hepatocytes. Interestingly, the LXRα-dependent activation of ATP-binding cassette transporter A1 (ABCA1) and ATP-binding cassette transporter G1 (ABCG1), other LXR target genes involved in reverse cholesterol transport (RCT), was not inhibited by LicA. LicA hindered the recruitment of RNA polymerase II to the LXRE of the SREBP-1c gene, but not of the ABCA1 gene. Conclusions LicA is a selective inhibitor of LXRα, repressing lipogenic LXRα target genes but not RCT-related LXRα target genes.
Collapse
Affiliation(s)
- Gyun-Sik Oh
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736 Korea ; Bio-medical Institute of Technology, University of Ulsan College of Medicine, Seoul, 138-736 Korea
| | - Gang Gu Lee
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736 Korea ; Bio-medical Institute of Technology, University of Ulsan College of Medicine, Seoul, 138-736 Korea
| | - Jin Yoon
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736 Korea ; Bio-medical Institute of Technology, University of Ulsan College of Medicine, Seoul, 138-736 Korea
| | - Won Keun Oh
- Korea Bioactive Natural Material Bank, College of Pharmacy, Seoul National University, Seoul, 151-742 Korea
| | - Seung-Whan Kim
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736 Korea ; Bio-medical Institute of Technology, University of Ulsan College of Medicine, Seoul, 138-736 Korea
| |
Collapse
|
43
|
Montgomery MK, Turner N. Mitochondrial dysfunction and insulin resistance: an update. Endocr Connect 2015; 4:R1-R15. [PMID: 25385852 PMCID: PMC4261703 DOI: 10.1530/ec-14-0092] [Citation(s) in RCA: 374] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 11/10/2014] [Indexed: 12/11/2022]
Abstract
Mitochondrial dysfunction has been implicated in the development of insulin resistance (IR); however, a large variety of association and intervention studies as well as genetic manipulations in rodents have reported contrasting results. Indeed, even 39 years after the first publication describing a relationship between IR and diminished mitochondrial function, it is still unclear whether a direct relationship exists, and more importantly if changes in mitochondrial capacity are a cause or consequence of IR. This review will take a journey through the past and summarise the debate about the occurrence of mitochondrial dysfunction and its possible role in causing decreased insulin action in obesity and type 2 diabetes. Evidence is presented from studies in various human populations, as well as rodents with genetic manipulations of pathways known to affect mitochondrial function and insulin action. Finally, we have discussed whether mitochondria are a potential target for the treatment of IR.
Collapse
Affiliation(s)
- Magdalene K Montgomery
- Department of PharmacologyUNSW Medicine, School of Medical Sciences, University of New South Wales, Kensington, Sydney, New South Wales 2052, Australia
| | - Nigel Turner
- Department of PharmacologyUNSW Medicine, School of Medical Sciences, University of New South Wales, Kensington, Sydney, New South Wales 2052, Australia
| |
Collapse
|
44
|
Larsen MC, Bushkofsky JR, Gorman T, Adhami V, Mukhtar H, Wang S, Reeder SB, Sheibani N, Jefcoate CR. Cytochrome P450 1B1: An unexpected modulator of liver fatty acid homeostasis. Arch Biochem Biophys 2015; 571:21-39. [PMID: 25703193 DOI: 10.1016/j.abb.2015.02.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/23/2015] [Accepted: 02/10/2015] [Indexed: 12/12/2022]
Abstract
Cytochrome P450 1b1 (Cyp1b1) expression is absent in mouse hepatocytes, but present in liver endothelia and activated stellate cells. Increased expression during adipogenesis suggests a role of Cyp1b1 metabolism in fatty acid homeostasis. Wild-type C57BL/6j (WT) and Cyp1b1-null (Cyp1b1-ko) mice were provided low or high fat diets (LFD and HFD, respectively). Cyp1b1-deletion suppressed HFD-induced obesity, improved glucose tolerance and prevented liver steatosis. Suppression of lipid droplets in sinusoidal hepatocytes, concomitant with enhanced glycogen granules, was a consistent feature of Cyp1b1-ko mice. Cyp1b1 deletion altered the in vivo expression of 560 liver genes, including suppression of PPARγ, stearoyl CoA desaturase 1 (Scd1) and many genes stimulated by PPARα, each consistent with this switch in energy storage mechanism. Ligand activation of PPARα in Cyp1b1-ko mice by WY-14643 was, nevertheless, effective. Seventeen gene changes in Cyp1b1-ko mice correspond to mouse transgenic expression that attenuated diet-induced diabetes. The absence of Cyp1b1 in mouse hepatocytes indicates participation in energy homeostasis through extra-hepatocyte signaling. Extensive sexual dimorphism in hepatic gene expression suggests a developmental impact of estrogen metabolism by Cyp1b1. Suppression of Scd1 and increased leptin turnover support enhanced leptin participation from the hypothalamus. Cyp1b1-mediated effects on vascular cells may underlie these changes.
Collapse
Affiliation(s)
- Michele Campaigne Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Justin R Bushkofsky
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, United States; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53706, United States
| | - Tyler Gorman
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Vaqar Adhami
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, United States
| | - Hasan Mukhtar
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, United States
| | - Suqing Wang
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Scott B Reeder
- Department of Radiology, University of Wisconsin, Madison, WI 53706, United States; Department of Medical Physics, University of Wisconsin, Madison, WI 53706, United States; Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53706, United States; Department of Medicine, University of Wisconsin, Madison, WI 53706, United States; Department of Emergency Medicine, University of Wisconsin, Madison, WI 53706, United States
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53706, United States
| | - Colin R Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States; Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, United States; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53706, United States.
| |
Collapse
|
45
|
Otero YF, Stafford JM, McGuinness OP. Pathway-selective insulin resistance and metabolic disease: the importance of nutrient flux. J Biol Chem 2015; 289:20462-9. [PMID: 24907277 DOI: 10.1074/jbc.r114.576355] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hepatic glucose and lipid metabolism are altered in metabolic disease (e.g. obesity, metabolic syndrome, and Type 2 diabetes). Insulin-dependent regulation of glucose metabolism is impaired. In contrast, lipogenesis, hypertriglyceridemia, and hepatic steatosis are increased. Because insulin promotes lipogenesis and liver fat accumulation, to explain the elevation in plasma and tissue lipids, investigators have suggested the presence of pathway-selective insulin resistance. In this model, insulin signaling to glucose metabolism is impaired, but insulin signaling to lipid metabolism is intact. We discuss the evidence for the differential regulation of hepatic lipid and glucose metabolism. We suggest that the primary phenotypic driver is altered substrate delivery to the liver, as well as the repartitioning of hepatic nutrient handling. Specific alterations in insulin signaling serve to amplify the alterations in hepatic substrate metabolism. Thus, hyperinsulinemia and its resultant increased signaling may facilitate lipogenesis, but are not the major drivers of the phenotype of pathway-selective insulin resistance.
Collapse
|
46
|
Keshk WA, Noeman SA. Impact of Chicory-Supplemented Diet on HMG-CoA Reductase, Acetyl-CoA Carboxylase, Visfatin and Anti-Oxidant Status in Triton WR-1339-Induced Hyperlipidemia. J Food Biochem 2015. [DOI: 10.1111/jfbc.12115] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Walaa A. Keshk
- Medical Biochemistry Department; Faculty of Medicine; Tanta University; El-Geish Street Tanta El-Gharbia Egypt
| | - Saad A. Noeman
- Medical Biochemistry Department; Faculty of Medicine; Tanta University; El-Geish Street Tanta El-Gharbia Egypt
| |
Collapse
|
47
|
Duarte JAG, Carvalho F, Pearson M, Horton JD, Browning JD, Jones JG, Burgess SC. A high-fat diet suppresses de novo lipogenesis and desaturation but not elongation and triglyceride synthesis in mice. J Lipid Res 2014; 55:2541-53. [PMID: 25271296 DOI: 10.1194/jlr.m052308] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Intracellular lipids and their synthesis contribute to the mechanisms and complications of obesity-associated diseases. We describe an NMR approach that provides an abbreviated lipidomic analysis with concurrent lipid biosynthetic fluxes. Following deuterated water administration, positional isotopomer analysis by deuterium NMR of specific lipid species was used to examine flux through de novo lipogenesis (DNL), FA elongation, desaturation, and TG-glycerol synthesis. The NMR method obviated certain assumptions regarding sites of enrichment and exchangeable hydrogens required by mass isotope methods. The approach was responsive to genetic and pharmacological gain or loss of function of DNL, elongation, desaturation, and glyceride synthesis. BDF1 mice consuming a high-fat diet (HFD) or matched low-fat diet for 35 weeks were examined across feeding periods to determine how flux through these pathways contributes to diet induced fatty liver and obesity. HFD mice had increased rates of FA elongation and glyceride synthesis. However DNL was markedly suppressed despite insulin resistance and obesity. We conclude that most hepatic TGs in the liver of HFD mice were formed from the reesterification of existing or ingested lipids, not DNL.
Collapse
Affiliation(s)
- Joao A G Duarte
- Advanced Imaging Research Center-Division of Metabolic Mechanisms of Disease, The University of Texas Southwestern Medical Center, Dallas, TX Center for Neurosciences and Cell Biology, Department of Zoology, University of Coimbra, Coimbra, Portugal
| | - Filipa Carvalho
- Center for Neurosciences and Cell Biology, Department of Zoology, University of Coimbra, Coimbra, Portugal
| | - Mackenzie Pearson
- Advanced Imaging Research Center-Division of Metabolic Mechanisms of Disease, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Jay D Horton
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Jeffrey D Browning
- Advanced Imaging Research Center-Division of Metabolic Mechanisms of Disease, The University of Texas Southwestern Medical Center, Dallas, TX Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX
| | - John G Jones
- Center for Neurosciences and Cell Biology, Department of Zoology, University of Coimbra, Coimbra, Portugal
| | - Shawn C Burgess
- Advanced Imaging Research Center-Division of Metabolic Mechanisms of Disease, The University of Texas Southwestern Medical Center, Dallas, TX Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
48
|
Zhang D, Tong X, Arthurs B, Guha A, Rui L, Kamath A, Inoki K, Yin L. Liver clock protein BMAL1 promotes de novo lipogenesis through insulin-mTORC2-AKT signaling. J Biol Chem 2014; 289:25925-35. [PMID: 25063808 PMCID: PMC4162191 DOI: 10.1074/jbc.m114.567628] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 07/15/2014] [Indexed: 12/15/2022] Open
Abstract
The clock protein BMAL1 (brain and muscle Arnt-like protein 1) participates in circadian regulation of lipid metabolism, but its contribution to insulin AKT-regulated hepatic lipid synthesis is unclear. Here we used both Bmal1(-/-) and acute liver-specific Bmal1-depleted mice to study the role of BMAL1 in refeeding-induced de novo lipogenesis in the liver. Both global deficiency and acute hepatic depletion of Bmal1 reduced lipogenic gene expression in the liver upon refeeding. Conversely, Bmal1 overexpression in mouse liver by adenovirus was sufficient to elevate the levels of mRNA of lipogenic enzymes. Bmal1(-/-) primary mouse hepatocytes displayed decreased levels of de novo lipogenesis and lipogenic enzymes, supporting the notion that BMAL1 regulates lipid synthesis in hepatocytes in a cell-autonomous manner. Both refed mouse liver and insulin-treated primary mouse hepatocytes showed impaired AKT activation in the case of either Bmal1 deficiency or Bmal1 depletion by adenoviral shRNA. Restoring AKT activity by a constitutively active mutant of AKT nearly normalized de novo lipogenesis in Bmal1(-/-) hepatocytes. Finally, Bmal1 deficiency or knockdown decreased the protein abundance of RICTOR, the key component of the mTORC2 complex, without affecting the gene expression of key factors of insulin signaling. Thus, our study uncovered a novel metabolic function of hepatic BMAL1 that promotes de novo lipogenesis via the insulin-mTORC2-AKT signaling during refeeding.
Collapse
Affiliation(s)
- Deqiang Zhang
- From the Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Xin Tong
- From the Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Blake Arthurs
- From the Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Anirvan Guha
- From the Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Liangyou Rui
- From the Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Avani Kamath
- From the Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Ken Inoki
- From the Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Lei Yin
- From the Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| |
Collapse
|
49
|
Park S, Hwang IW, Makishima Y, Perales-Clemente E, Kato T, Niederländer NJ, Park EY, Terzic A. Spot14/Mig12 heterocomplex sequesters polymerization and restrains catalytic function of human acetyl-CoA carboxylase 2. J Mol Recognit 2014; 26:679-88. [PMID: 24277613 PMCID: PMC4283044 DOI: 10.1002/jmr.2313] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 08/20/2013] [Accepted: 08/21/2013] [Indexed: 01/17/2023]
Abstract
Acetyl-CoA carboxylase 2 (ACC2) is an isoform of ACC functioning as a negative regulator of fatty acid β-oxidation. Spot14, a thyroid hormone responsive protein, and Mig12, a Spot14 paralog, have recently been identified as regulators of fatty acid synthesis targeting ACC1, a distinctive subtype of ACC. Here, we examined whether Spot14/Mig12 modulates ACC2. Nanoscale protein topography mapped putative protein-protein interactions between purified human Spot14/Mig12 and ACC2, validated by functional assays. Human ACC2 displayed consistent enzymatic activity, and homogeneous particle distribution was probed by atomic force microscopy. Citrate-induced polymerization and enzymatic activity of ACC2 were restrained by the addition of the recombinant Spot14/Mig12 heterocomplex but only partially by the oligo-heterocomplex, demonstrating that the heterocomplex is a designated metabolic inhibitor of human ACC2. Moreover, Spot14/Mig12 demonstrated a sequestering role preventing an initial ACC2 nucleation step during filamentous polymer formation. Thus, the Spot14/Mig12 heterocomplex controls human ACC2 polymerization and catalytic function, emerging as a previously unrecognized molecular regulator in catalytic lipid metabolism.
Collapse
Affiliation(s)
- Sungjo Park
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA; Marriott Heart Disease Research Program, Division of Cardiovascular Diseases, Departments of Medicine, Molecular Pharmacology and Experimental Therapeutics, and Medical Genetics, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Martius G, Alwahsh SM, Rave-Fränk M, Hess CF, Christiansen H, Ramadori G, Malik IA. Hepatic fat accumulation and regulation of FAT/CD36: an effect of hepatic irradiation. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:5379-5392. [PMID: 25197426 PMCID: PMC4152116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 07/16/2014] [Indexed: 06/03/2023]
Abstract
Irradiation is known to induce inflammation and affect fat metabolic pathways. The current study investigates hepatic fat accumulation and fatty acid transportation in a rat model of single dose liver irradiation (25-Gy). Rat livers were selectively irradiated in-vivo (25-Gy), sham-irradiated rats served as controls. Hepatic lipids were studied by colorimetric assays in liver and serum. Intracellular lipids, protein and mRNA were studied by Nile red staining, immunohistology, Western Blot analysis and RT-PCR in liver, respectively. Changes in FAT/CD36 expression were studied in-vitro in a human monocyte cell line U937 after irradiation in presence or absence of infliximab (IFX). Nile Red staining of liver cryosections showed a quick (12-48 h) increase in fat droplets. Accordingly, hepatic triglycerides (TG) and free fatty acids (FFA) were elevated. An early increase (3-6 h) in the serum level of HDL-C, TG and cholesterol was measured after single dose irradiation followed by a decrease thereafter. Furthermore, expression of the fat transporter protein FAT/CD36 was increased, immunohistochemistry revealed basolateral and cytoplasmic expression in hepatocytes. Moreover, apolipoprotein-B100, -C3 and enzymes (acetyl-CoA carboxylase, lipoprotein-lipase, carnitine-palmitoyltransferase, malonyl-CoA-decarboxylase) involved in fat metabolism were induced at 12-24 h. Early activation of the NFkβ pathway (IκBα) by TNF-α was seen, followed by a significant elevation of serum markers for liver damage (AST and GLDH). TNF-α blockage by anti-TNF-α in cell culture (U937) prevented the increase of FAT/CD36 caused by irradiation. Selective liver irradiation is a model for rapid induction of steatosis hepatis and fat accumulation could be triggered by irradiation-induced inflammatory mediators (e.g. TNF-α).
Collapse
Affiliation(s)
- Gesa Martius
- Department of Gastroenterology and Endocrinology, University Medical Center GoettingenD-37075 Goettingen, Germany
| | - Salamah Mohammad Alwahsh
- Department of Gastroenterology and Endocrinology, University Medical Center GoettingenD-37075 Goettingen, Germany
| | - Margret Rave-Fränk
- Department of Radiotherapy and Radiooncology, University Medical Center GoettingenD-37075 Goettingen, Germany
| | - Clemens Friedrich Hess
- Department of Radiotherapy and Radiooncology, University Medical Center GoettingenD-37075 Goettingen, Germany
| | - Hans Christiansen
- Department of Radiotherapy and Radiooncology, University Medical Center GoettingenD-37075 Goettingen, Germany
- Current address: Department of Radiation Oncology, Medizinische Hochschule HannoverCarl-Neuberg-Straße, 30625 Hannover, Germany
| | - Giuliano Ramadori
- Department of Gastroenterology and Endocrinology, University Medical Center GoettingenD-37075 Goettingen, Germany
| | - Ihtzaz Ahmed Malik
- Department of Gastroenterology and Endocrinology, University Medical Center GoettingenD-37075 Goettingen, Germany
| |
Collapse
|