1
|
Liu Y, Wu Y, Wang C, Hu W, Zou S, Ren H, Zuo Y, Qu L. MiR-127-3p enhances macrophagic proliferation via disturbing fatty acid profiles and oxidative phosphorylation in atherosclerosis. J Mol Cell Cardiol 2024; 193:36-52. [PMID: 38795767 DOI: 10.1016/j.yjmcc.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/05/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Atherosclerosis is a chronic pathology, leading to acute coronary heart disease or stroke. MiR-127 has been found significantly upregulated in advanced atherosclerosis. But its function in atherosclerosis remains unexplored. We explored the role of miR-127-3p in regulating atherosclerosis development and its downstream mechanisms. METHODS The expression profile of miR-127 in carotid atherosclerotic plaques of 23 patients with severe carotid stenosis was detected by RT-qPCR and in situ hybridization. Primary bone marrow-derived macrophages (BMDM) stimulated with oxidized low-density lipoprotein were used as an in vitro model. CCK-8, EdU, RT-qPCR, and flow cytometry were used to detect the proliferative capacity and polarization of BMDM, which were infected by lentivirus-carrying plasmid to upregulate or downregulate miR-127-3p expression, respectively. RNA sequencing combined with bioinformatic analysis and targeted fatty acid metabolomics approach were used to detect the transcriptome and lipid metabolites. The association between miR-127-3p and its target was verified by dual-luciferase activity reporting and Western blotting. Oxygen consumption rate of BMDM were detected using seahorse analysis. High-cholesterol-diet-fed low density lipoprotein deficient (LDLR-/-) mice, with-or-without carotid tandem-stenosis surgery, were treated with miR-127-3p agomir or antagomir to examine its effect on plaque development and stability. RESULTS miR-127-3p, not -5p, is elevated in human advanced carotid atheroma and its expression is positively associated with macrophage accummulation in plaques. In vitro, miR-127-3p-overexpressed macrophage exhibites increased proliferation capacity and facilitates M1 polariztion whereas the contrary trend is present in miR-127-3p-inhibited macrophage. Stearoyl-CoA desaturase-1 (SCD1) is one potential target of miR-127-3p. miR-127-3p mimics decreases the activity of 3' untranslated regions of SCD-1. Furthermore, miR-127-3p downregulates SCD1 expression, and reversing the expression of SCD1 attenuates the increased proliferation induced by miR-127-3p overexpression in macrophage. miR-127-3p overexpression could also lead to decreased content of unsaturated fatty acids (UFAs), increased content of acetyl CoA and increased level of oxidative phosphorylation. In vivo, miR-127-3p agomir significantly increases atherosclerosis progression, macrophage proliferation and decreases SCD1 expression and the content of UFAs in aortic plaques of LDLR-/- mice. Conversely, miR-127-3p antagomir attenuated atherosclerosis, macrophage proliferation in LDLR-/- mice, and enhanced carotid plaque stability in mice with vulnerable plaque induced. CONCLUSION MiR-127-3p enhances proliferation in macrophages through downregulating SCD-1 expression and decreasing the content of unsaturated fatty acid, thereby promoting atherosclerosis development and decreasing plaque stability. miR-127-3p/SCD1/UFAs might provide potential therapeutic target for anti-inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Yandong Liu
- Department of Geriatrics, 905th Hospital of PLA NAVY, Shanghai, China; Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Yicheng Wu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Chao Wang
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Weilin Hu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Sili Zou
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Huiqiong Ren
- Department of Geriatrics, 905th Hospital of PLA NAVY, Shanghai, China.
| | - Yong Zuo
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lefeng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
2
|
Chen W, Zhong Y, Yuan Y, Zhu M, Hu W, Liu N, Xing D. New insights into the suppression of inflammation and lipid accumulation by JAZF1. Genes Dis 2023; 10:2457-2469. [PMID: 37554201 PMCID: PMC10404878 DOI: 10.1016/j.gendis.2022.10.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/27/2022] [Accepted: 10/25/2022] [Indexed: 12/03/2022] Open
Abstract
Atherosclerosis is one of the leading causes of disease and death worldwide. The identification of new therapeutic targets and agents is critical. JAZF1 is expressed in many tissues and is found at particularly high levels in adipose tissue (AT). JAZF1 suppresses inflammation (including IL-1β, IL-4, IL-6, IL-8, IL-10, TNFα, IFN-γ, IAR-20, COL3A1, laminin, and MCP-1) by reducing NF-κB pathway activation and AT immune cell infiltration. JAZF1 reduces lipid accumulation by regulating the liver X receptor response element (LXRE) of the SREBP-1c promoter, the cAMP-response element (CRE) of HMGCR, and the TR4 axis. LXRE and CRE sites are present in many cytokine and lipid metabolism gene promoters, which suggests that JAZF1 regulates these genes through these sites. NF-κB is the center of the JAZF1-mediated inhibition of the inflammatory response. JAZF1 suppresses NF-κB expression by suppressing TAK1 expression. Interestingly, TAK1 inhibition also decreases lipid accumulation. A dual-targeting strategy of NF-κB and TAK1 could inhibit both inflammation and lipid accumulation. Dual-target compounds (including prodrugs) 1-5 exhibit nanomolar inhibition by targeting NF-κB and TAK1, EGFR, or COX-2. However, the NF-κB suppressing activity of these compounds is relatively low (IC50 > 300 nM). Compounds 6-14 suppress NF-κB expression with IC50 values ranging from 1.8 nM to 38.6 nM. HS-276 is a highly selective, orally bioavailable TAK1 inhibitor. Combined structural modifications of compounds using a prodrug strategy may enhance NF-κB inhibition. This review focused on the role and mechanism of JAZF1 in inflammation and lipid accumulation for the identification of new anti-atherosclerotic targets.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Yingjie Zhong
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Yang Yuan
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Meng Zhu
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Wenchao Hu
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
- Department of Endocrinology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong 266035, China
| | - Ning Liu
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Dongming Xing
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
3
|
Ye Y, Kawaguchi Y, Takeuchi A, Zhang N, Mori R, Mijiti M, Banno A, Okada T, Hiramatsu N, Nagaoka S. Rose polyphenols exert antiobesity effect in high-fat-induced obese mice by regulating lipogenic gene expression. Nutr Res 2023; 119:76-89. [PMID: 37757642 DOI: 10.1016/j.nutres.2023.09.002] [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: 06/19/2023] [Revised: 08/27/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
Obesity presents a major risk factor in the development of cardiovascular diseases. Recent reports indicate that many kinds of polyphenols have the potential to prevent metabolic diseases. We hypothesized that rose polyphenols (ROSE) have the effect of improvement in lipid metabolism. In this study, we investigated whether rose polyphenols affected lipid metabolism and exerted antiobesity. To clarify the mechanism, C57BL/6J mice were fed a high-fat diet containing 0.25% ROSE for 35 days. Compared with the control group, body weight gain and adipose tissue weight in the 0.25% ROSE group were significantly decreased. Serum cholesterol and hepatic triglyceride concentrations significantly decreased, whereas fecal triglyceride was significantly increased in the 0.25% ROSE group. Liver stearoyl-CoA desaturase 1 (Scd1), 3-hydroxy-3-methylglutaryl-CoA reductase (Hmgcr), and acyl-CoA:cholesterol acyltransferase 1 (Acat1) mRNA as well as protein stearoyl-CoA desaturase 1 concentrations were significantly lower in the 0.25% ROSE group than that in the control group. The mRNA and the protein concentrations of adipose triglyceride lipase, hormone-sensitive lipase, and peroxisomal acylcoenzyme A oxidase 1 in white adipose tissue were significantly higher in the 0.25% ROSE group than that in the control group. The components in rose polyphenols were quantified by liquid chromatography-tandem mass spectrometry, and we consider that ellagic acid plays an important role in an antiobesity effect because the ellagic acid content is the highest among polyphenols in rose polyphenols. In summary, rose polyphenols exhibit antiobesity effects by influencing lipid metabolism-related genes and proteins to promote lipolysis and suppress lipid synthesis.
Collapse
Affiliation(s)
- Yuyang Ye
- Faculty of Applied Biological Sciences, Department of Applied Life Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yuya Kawaguchi
- Faculty of Applied Biological Sciences, Department of Applied Life Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Asahi Takeuchi
- Faculty of Applied Biological Sciences, Department of Applied Life Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Ni Zhang
- Faculty of Applied Biological Sciences, Department of Applied Life Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Ryosuke Mori
- Faculty of Applied Biological Sciences, Department of Applied Life Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Maihemuti Mijiti
- Faculty of Applied Biological Sciences, Department of Applied Life Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Arata Banno
- Faculty of Applied Biological Sciences, Department of Applied Life Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | | | | | - Satoshi Nagaoka
- Faculty of Applied Biological Sciences, Department of Applied Life Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
| |
Collapse
|
4
|
Wang J, Jing J, Gong Z, Tang J, Wang L, Jia G, Liu G, Chen X, Tian G, Cai J, Kang B, Che L, Zhao H. Different Dietary Sources of Selenium Alleviate Hepatic Lipid Metabolism Disorder of Heat-Stressed Broilers by Relieving Endoplasmic Reticulum Stress. Int J Mol Sci 2023; 24:15443. [PMID: 37895123 PMCID: PMC10607182 DOI: 10.3390/ijms242015443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
As global warming continues, the phenomenon of heat stress (HS) in broilers occurs frequently. The alleviating effect of different selenium (Se) sources on HS-induced hepatic lipid metabolism disorders in broilers remains unclear. This study compared the protective effects of four Se sources (sodium selenite; selenium yeast; selenomethionine; nano-Se) on HS-induced hepatic lipid metabolism disorder and the corresponding response of selenotranscriptome in the liver of broilers. The results showed that HS-induced liver injury and hepatic lipid metabolism disorder, which were reflected in the increased activity of serum alanine aminotransferase (ALT), the increased concentration of triacylglycerol (TG) and total cholesterol (TC), the increased activity of acetyl-CoA carboxylase (ACC), diacylglycerol O-acyltransferase (DGAT) and fatty acid synthase (FAS), and the decreased activity of hepatic lipase (HL) in the liver. The hepatic lipid metabolism disorder was accompanied by the increased mRNA expression of lipid synthesis related-genes, the decreased expression of lipidolysis-related genes, and the increased expression of endoplasmic reticulum (ER) stress biomarkers (PERK, IRE1, ATF6, GRP78). The dietary supplementation of four Se sources exhibited similar protective effects. Four Se sources increased liver Se concentration and promoted the expression of selenotranscriptome and several key selenoproteins, enhanced liver antioxidant capacity and alleviated HS-induced ER stress, and thus resisted the hepatic lipid metabolism disorders of broilers exposed to HS. In conclusion, dietary supplementation of four Se sources (0.3 mg/kg) exhibited similar protective effects on HS-induced hepatic lipid metabolism disorders of broilers, and the protective effect is connected to the relieving of ER stress.
Collapse
Affiliation(s)
- Jiayi Wang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| | - Jinzhong Jing
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| | - Zhengyi Gong
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| | - Jiayong Tang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| | - Longqiong Wang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| | - Gang Jia
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| | - Guangmang Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| | - Gang Tian
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| | - Jingyi Cai
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| | - Bo Kang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
| | - Lianqiang Che
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| | - Hua Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.W.); (J.J.); (Z.G.); (J.T.); (L.W.); (G.J.); (G.L.); (X.C.); (G.T.); (J.C.); (L.C.)
| |
Collapse
|
5
|
Šarac I, Debeljak-Martačić J, Takić M, Stevanović V, Milešević J, Zeković M, Popović T, Jovanović J, Vidović NK. Associations of fatty acids composition and estimated desaturase activities in erythrocyte phospholipids with biochemical and clinical indicators of cardiometabolic risk in non-diabetic Serbian women: the role of level of adiposity. Front Nutr 2023; 10:1065578. [PMID: 37545582 PMCID: PMC10397414 DOI: 10.3389/fnut.2023.1065578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction Fatty acids (FAs) composition and desaturase activities can be altered in different metabolic conditions, but the adiposity-independent associations with clinical and biochemical indicators of cardiometabolic risk are still unclear. This study aimed to analyze the associations of FAs composition and estimated desaturase activities with anthropometric, clinical, and biochemical cardiometabolic risk indicators in non-diabetic Serbian women, and to investigate if these associations were independent of the level of adiposity and other confounders. Methods In 76 non-diabetic, otherwise healthy Serbian women, aged 24-68 years, with or without metabolic syndrome or obesity (BMI=23.6±5.6 kg/m2), FA composition in erythrocyte phospholipids was measured by gas-liquid chromatography. Desaturase activities were estimated from product/precursor FAs ratios (D9D:16:1n-7/16:0; D6D:20:3n-6/18:2n-6; D5D:20:4n-6/20:3n-6). Correlations were made with anthropometric, biochemical (serum glucose, triacylglycerols, LDL-C, HDL-C, ALT, AST, and their ratios) and clinical (blood pressure) indicators of cardiometabolic risk. Linear regression models were performed to test the independence of these associations. Results Estimated desaturase activities and certain FAs were associated with anthropometric, clinical and biochemical indicators of cardiometabolic risk: D9D, D6D, 16:1n-7 and 20:3n-6 were directly associated, while D5D and 18:0 were inversely associated. However, the associations with clinical and biochemical indicators were not independent of the associations with the level of adiposity, since they were lost after controlling for anthropometric indices. After controlling for multiple confounders (age, postmenopausal status, education, smoking, physical activity, dietary macronutrient intakes, use of supplements, alcohol consumption), the level of adiposity was the most significant predictor of desaturase activities and aforementioned FAs levels, and mediated their association with biochemical/clinical indicators. Vice versa, desaturase activities predicted the level of adiposity, but not other components of cardiometabolic risk (if the level of adiposity was accounted). While the associations of anthropometric indices with 16:1n-7, 20:3n-6, 18:0 and D9D and D6D activities were linear, the associations with D5D activity were the inverse U-shaped. The only adiposity-independent association of FAs profiles with the indicators of cardiometabolic risk was a positive association of 20:5n-3 with ALT/AST ratio, which requires further exploration. Discussion Additional studies are needed to explore the mechanisms of the observed associations.
Collapse
Affiliation(s)
- Ivana Šarac
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jasmina Debeljak-Martačić
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marija Takić
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Vuk Stevanović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena Milešević
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Zeković
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Popović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jovica Jovanović
- Department of Occupational Health, Faculty of Medicine, University of Niš, Niš, Serbia
| | - Nevena Kardum Vidović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
6
|
Sen U, Coleman C, Sen T. Stearoyl coenzyme A desaturase-1: multitasker in cancer, metabolism, and ferroptosis. Trends Cancer 2023; 9:480-489. [PMID: 37029018 DOI: 10.1016/j.trecan.2023.03.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 04/09/2023]
Abstract
Cancer progression is a highly balanced process and is maintained by a sequence of finely tuned metabolic pathways. Stearoyl coenzyme A desaturase-1 (SCD1), the fatty enzyme that converts saturated fatty acids into monounsaturated fatty acids, is a critical modulator of the fatty acid metabolic pathway. SCD1 expression is associated with poor prognosis in several cancer types. SCD1 triggers an iron-dependent cell death called ferroptosis and elevated levels of SCD1 protect cancer cells against ferroptosis. Pharmacological inhibition of SCD1 as monotherapy and in combination with chemotherapeutic agents shows promising antitumor potential in preclinical models. In this review, we summarize the role of SCD in cancer cell progression, survival, and ferroptosis and discuss potential strategies to exploit SCD1 inhibition in future clinical trials.
Collapse
Affiliation(s)
- Utsav Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Charles Coleman
- The Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA
| | - Triparna Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
7
|
Al-Emarah MK, Kazerani HR, Taghizad F, Dehghani H, Elahi M. Anti-obesity effect of the bacterial product nisin in an NIH Swiss mouse model. Lipids Health Dis 2023; 22:23. [PMID: 36765351 PMCID: PMC9912503 DOI: 10.1186/s12944-023-01788-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
Obesity is a life-threatening metabolic disorder that predisposes individuals to other diseases. In this study, the effect of nisin, a bacteriocin produced by some bacteria, on an animal model of obesity based on selected parameters was investigated. Forty Swiss NIH mice were randomly divided into four groups and received either a placebo (saline) or nisin (25, 50, or 100 μg/kg, ip) daily for 8 weeks. The mice in all groups were fed a high-sugar diet throughout the experiment. Bodyweight and food intake were measured weekly, and at the end of the experiment, the levels of FBS, serum triglyceride, cholesterol, high-density lipoprotein, low-density lipoprotein, and hepatic enzymes were tested, and red and white blood cell counts, hemoglobin concentration, mean corpuscular volume, mean corpuscular hemoglobin, and mean corpuscular hemoglobin concentration were determined. Finally, the expression levels of some obesity-related genes, including stearoyl-CoA desaturase-1 (SCD-1), glucose transporter-4 (GLUT4), zinc finger protein 423 (zfp423), 422 (ap2), and tumor necrosis factor-alpha (TNF-α), were assessed using reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR). After the experiment, the body weights, abdominal fat, and body mass index were significantly lower in the nisin-treated groups than in the control group. The highest effect was observed with 50 μg/kg nisin. The expression of SCD-1, GLUT4, 422(ap2), and TNF-α decreased significantly following treatment with nisin. No significant differences were observed in the other studied parameters, and no toxic effects were observed for nisin under these experimental conditions. The results suggested that nisin could have antiobesity effects.
Collapse
Affiliation(s)
- M. K. Al-Emarah
- grid.411301.60000 0001 0666 1211Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran ,Faculty of Agriculture and Marshlands, University of Thi-qar, Thi-qar, Iraq
| | - H. R. Kazerani
- grid.411301.60000 0001 0666 1211Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - F. Taghizad
- grid.411301.60000 0001 0666 1211Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - H. Dehghani
- grid.411301.60000 0001 0666 1211Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran ,grid.411301.60000 0001 0666 1211Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - M. Elahi
- grid.411301.60000 0001 0666 1211Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
8
|
Kim JE, Bennett DC, Wright K, Cheng KM. Seasonal and sexual variation in mRNA expression of selected adipokine genes affecting fat deposition and metabolism of the emu (Dromaius novaehollandiae). Sci Rep 2022; 12:6325. [PMID: 35428830 PMCID: PMC9012844 DOI: 10.1038/s41598-022-10232-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
Emus are farmed for fat production. Oil rendered from their back and abdominal fat pads has good anti-oxidant and anti-inflammatory properties and has ingredients that promote cell growth. Our objective is to examine the mRNA expression of 7 emu adipokine genes (eFABP4, eSCD1, eAdipoQ, eAdipoR1, eAdipoR2, eLEP and eLepR) to identify gene markers that may help improve emu fat production. Back and abdominal fat tissues from 11 adult emus were biopsied at four time points (April, June, August and November). Total RNA was isolated and cDNA was synthesized. Gene specific primers were designed for partial cloning fragments to amplify the open reading frame of the 7 genes. eLEP was not expressed in emu fat tissue. Nucleotides and amino acids sequences of the 6 expressed gene were compared with homologs from other species and phylogenetic relationships established. Seasonal mRNA expression of each gene was assessed by quantitative RT-PCR and differential expression analysed by the 2-ΔΔCT method. The 6 expressed genes showed seasonal variation in expression and showed association of expression level with back fat adiposity. More whole-genome scanning studies are needed to develop novel molecular markers that can be applied to improve fat production in emus.
Collapse
Affiliation(s)
- Ji Eun Kim
- Faculty of Land and Food Systems, Avian Research Centre, University of British Columbia, 2357 Main Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Darin C Bennett
- Faculty of Land and Food Systems, Avian Research Centre, University of British Columbia, 2357 Main Mall, Vancouver, BC, V6T 1Z4, Canada
- Animal Science Department, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Kristina Wright
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, 570 West 7th Avenue, Vancouver, BC, V5Z 4S6, Canada
| | - Kimberly M Cheng
- Faculty of Land and Food Systems, Avian Research Centre, University of British Columbia, 2357 Main Mall, Vancouver, BC, V6T 1Z4, Canada.
| |
Collapse
|
9
|
Jing J, Yin S, Liu Y, Liu Y, Wang L, Tang J, Jia G, Liu G, Tian G, Chen X, Cai J, Kang B, Zhao H. Hydroxy Selenomethionine Alleviates Hepatic Lipid Metabolism Disorder of Pigs Induced by Dietary Oxidative Stress via Relieving the Endoplasmic Reticulum Stress. Antioxidants (Basel) 2022; 11:552. [PMID: 35326202 PMCID: PMC8945048 DOI: 10.3390/antiox11030552] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 11/18/2022] Open
Abstract
This study used 40 castrated male pigs to determine the protective effects of a new selenium molecule (hydroxy selenomethionine, OH-SeMet) on dietary oxidative stress (DOS) induced hepatic lipid metabolism disorder, and corresponding response of selenotranscriptome. The pigs were randomly grouped into 5 dietary treatments and fed a basal diet formulated with either normal corn and oils or oxidized diet in which the normal corn and oils were replaced by aged corn and oxidized oils, and supplemented with OH-SeMet at 0.0, 0.3, 0.6 and 0.9 mg Se/kg for a period of 16 weeks (n = 8). The results showed that DOS induced liver damage, increased serum alanine aminotransferase (ALT) and alkaline phosphatase (ALP) levels, decreased serum triacylglycerol (TG) level, suppressed antioxidant capacity in the liver, and changed lipid metabolism enzyme activity, thus causing lipid metabolism disorder in the liver. The DOS-induced lipid metabolism disorder was accompanied with endoplasmic reticulum (ER) stress, changes in lipid metabolism-related genes and selenotranscriptome in the liver. Dietary Se supplementation partially alleviated the negative impact of DOS on the lipid metabolism. These improvements were accompanied by increases in Se concentration, liver index, anti-oxidative capacity, selenotranscriptome especially 11 selenoprotein-encoding genes, and protein abundance of GPX1, GPX4 and SelS in the liver, as well as the decrease in SelF abundance. The Se supplementation also alleviated ER stress, restored liver lipid metabolism enzyme activity, increased the mRNA expression of lipid synthesis-related genes, and decreased the mRNA levels of lipidolysis-related genes. In conclusion, the dietary Se supplementation restored antioxidant capacity and mitigated ER stress induced by DOS, thus resisting hepatic lipid metabolism disorders that are associated with regulation of selenotranscriptome.
Collapse
Affiliation(s)
- Jinzhong Jing
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Shenggang Yin
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Yan Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Yonggang Liu
- Adisseo Asia Pacific Pte. Ltd., Singapore 188778, Singapore;
| | - Longqiong Wang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Jiayong Tang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Gang Jia
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Guangmang Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Gang Tian
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Jingyi Cai
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Bo Kang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
| | - Hua Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| |
Collapse
|
10
|
Bai J, Li J, Pan R, Zhu Y, Xiao X, Li Y, Li C. Polysaccharides from Volvariella volvacea inhibit fat accumulation in C. elegans dependent on the aak-2/nhr-49-mediated pathway. J Food Biochem 2021; 45:e13912. [PMID: 34561881 DOI: 10.1111/jfbc.13912] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 12/28/2022]
Abstract
Volvariella volvacea has bioactivities in improving immunity, anti-oxidation, and alleviating obesity, which is an excellent functional food. Polysaccharide from Volvariella volvacea (VPS), one of the main bioactive components, exerts a potential fat-lowering effect, but its exact mechanism remains unclear. In this study, the effects and molecular pathways of VPS regulate the fat deposition of Caenorhabditis elegans. Results showed that VPS at low (250 μg/ml), medium (500 μg/ml) and high (750 μg/ml) concentrations all reduced the overall fat, without inhibitory effects on the growth and movement abilities of nematode. VPS at 500 μg/ml could dramatically decrease the triglyceride (TG) level of wild-type nematode, while no significant changes in TG content were observed in mutants deficient in aak-2 (energy receptor), nhr-49 (nuclear transcription factor), fat-5, and fat-7 genes. VPS declines fat storage of C. elegans, largely through the aak-2/nhr-49-mediated fatty acid synthesis pathway, and partially the acs-2-mediated fatty acid oxidation pathway. PRACTICAL APPLICATIONS: A model illustrates the mechanism of polysaccharide from Volvariella volvacea (VPS) inhibiting fat accumulation in Caenorhabditis elegans. VPS may directly or indirectly activate the energy sensor aak-2, which governs lipid metabolism. Results demonstrate that VPS regulates fat metabolism including fatty acid oxidation (FAO) and fatty acid synthesis (FAS), rather than lipolysis. In the FAO, VPS promotes FAO by up-regulating the mRNA and protein levels of acs-2. In FAS, VPS significantly down-regulated the transcriptional regulator nhr-49 and the downstream targets fat-5, fat-6, and fat-7, thereby declining the overall fat deposition. In conclusion, VPS inhibits the fat accumulation of C. elegans largely dependent on an aak-2/nhr-49-mediated FAS pathway.
Collapse
Affiliation(s)
- Juan Bai
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun, China.,School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China.,Jiangsu Jiangnan Biotechnology Co., Ltd., Zhenjiang, China
| | - Jie Li
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Ruirong Pan
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ying Zhu
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Xiang Xiao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Yu Li
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Changtian Li
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
11
|
Taghizad F, Kazerani HR, Dehghani H, Asoodeh A, Yaghubi D. A novel approach towards obesity: The use of a bacterial product, gassericin A, in 3T3-L1 cells. Obes Res Clin Pract 2021; 15:499-505. [PMID: 34474970 DOI: 10.1016/j.orcp.2021.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND OBJECTIVE The problem of obesity and its related complications are adversely affecting human society. We studied the effects of gassericin A, a bacteriocin produced by the intestinal bacteria, on adipocyte differentiation and development. DESIGN Gassericin A was purified from Lactobacillus gasseri LA39 and was added to the culture medium of 3T3-L1 cells in two phases: Phase 1, 3T3-L1 cells were incubated with gassericin A while being induced to adipocytes (days 1-7); phase 2, the cells were incubated with the bacteriocin after being induced to adipocytes (days 8-12). The resultant changes in the pattern of expression of some of the important genes involved in adipogenesis were evaluated by RT-qPCR. The viability of cells and their numbers were also studied. RESULTS In phase 1 of the study, the levels of transcripts for stearoyl CoA desaturase (SCD-1), zinc finger protein 423 (zfp-423), and glucose transporter 4 (GLUT4) genes were significantly reduced, while that of 422ap2 gene showed a significant increment (p < 0.05). In phase 2, the zfp-423 gene showed a reduction of expression and the 422ap2 gene showed an increase in expression (p < 0.05). The other genes including UCP-1 and TNF-α did not show any significant changes in neither of the groups. Gassericin A did not affect the morphology or viability of the cells, however, the numbers of cells had nearly doubled in the treatment groups. CONCLUSION It seems that gassericin A could significantly alter the properties of adipocytes while they are in the process of development and after they have developed.
Collapse
Affiliation(s)
- Fereidoun Taghizad
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hamid Reza Kazerani
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Hesam Dehghani
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran; Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran; Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Asoodeh
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Danial Yaghubi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
12
|
Hammoud R, Pannia E, Kubant R, Metherel A, Simonian R, Pausova Z, Anderson GH. High Choline Intake during Pregnancy Reduces Characteristics of the Metabolic Syndrome in Male Wistar Rat Offspring Fed a High Fat But Not a Normal Fat Post-Weaning Diet. Nutrients 2021; 13:nu13051438. [PMID: 33923230 PMCID: PMC8145686 DOI: 10.3390/nu13051438] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/13/2021] [Accepted: 04/21/2021] [Indexed: 12/19/2022] Open
Abstract
Maternal choline intakes are below recommendations, potentially impairing the child’s later-life metabolic health. This study aims to elucidate the interaction between the choline content of the gestational diet (GD) and fat content of the post-weaning diet (PWD) on metabolic phenotype of male Wistar rats. Pregnant Wistar rats were fed a standard rodent diet (AIN-93G) with either recommended choline (RC, 1 g/kg diet choline bitartrate) or high choline (HC, 2.5-fold). Male pups were weaned to either a normal (16%) fat (NF) or a high (45%) fat (HF) diet for 17 weeks. Body weight, visceral adiposity, food intake, energy expenditure, plasma hormones, triglycerides, and hepatic fatty acids were measured. HC-HF offspring had 7% lower body weight but not food intake, and lower adiposity, plasma triglycerides, and insulin resistance compared to RC-HF. They also had increased hepatic n-3 fatty acids and a reduced n-6/n-3 and C 18:1 n-9/C18:0 ratios. In contrast, HC-NF offspring had 6–8% higher cumulative food intake and body weight, as well as increased leptin and elevated hepatic C16:1 n-7/C16:0 ratio compared to RC-NF. Therefore, gestational choline supplementation associated with improved long-term regulation of several biomarkers of the metabolic syndrome in male Wistar rat offspring fed a HF, but not a NF, PWD.
Collapse
Affiliation(s)
- Rola Hammoud
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (R.H.); (E.P.); (R.K.); (A.M.); (R.S.); (Z.P.)
| | - Emanuela Pannia
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (R.H.); (E.P.); (R.K.); (A.M.); (R.S.); (Z.P.)
| | - Ruslan Kubant
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (R.H.); (E.P.); (R.K.); (A.M.); (R.S.); (Z.P.)
| | - Adam Metherel
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (R.H.); (E.P.); (R.K.); (A.M.); (R.S.); (Z.P.)
| | - Rebecca Simonian
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (R.H.); (E.P.); (R.K.); (A.M.); (R.S.); (Z.P.)
| | - Zdenka Pausova
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (R.H.); (E.P.); (R.K.); (A.M.); (R.S.); (Z.P.)
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - G. Harvey Anderson
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (R.H.); (E.P.); (R.K.); (A.M.); (R.S.); (Z.P.)
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence: ; Tel.: +416-978-1832
| |
Collapse
|
13
|
Xiao X, Zhang X, Bai J, Li J, Zhang C, Zhao Y, Zhu Y, Zhang J, Zhou X. Bisphenol S increases the obesogenic effects of a high-glucose diet through regulating lipid metabolism in Caenorhabditis elegans. Food Chem 2020; 339:127813. [PMID: 32916401 DOI: 10.1016/j.foodchem.2020.127813] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/28/2022]
Abstract
Bisphenol S (BPS), a structural analog of Bisphenol A (BPA), has been widely used as a substitute for epoxy resin, food packaging materials, and other products due to the limited application of BPA. Studies in vivo and in vitro have indicated that BPA could induce fat accumulation like an obesogen. The main goal of this study was to investigate the role and mechanism of BPS in lipid metabolism using Caenorhabditis elegans (C. elegans) as a model. Results showed that both the overall fat deposition and the triglyceride level were significantly increased in a non-monotonically increasing trend, and the low dose of BPS (0.01 μM) exhibited a stronger influence. Additionally, BPS enhanced fat synthesis depending on daf-16, fat-5, fat-6 and fat-7, and inhibited fatty acid oxidation via nhr-49 and acs-2. This study further indicate that fat accumulation induced by BPS requires nhr-49, which also mediated the nuclear hormone signaling pathway.
Collapse
Affiliation(s)
- Xiang Xiao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Xiaowei Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Juan Bai
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Jie Li
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Caiqin Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Yansheng Zhao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Ying Zhu
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Jiayan Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Xinghua Zhou
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China.
| |
Collapse
|
14
|
El-Zeftawy M, Ali SAEM, Salah S, Hafez HS. The functional nutritional and regulatory activities of calcium supplementation from eggshell for obesity disorders management. J Food Biochem 2020; 44:e13313. [PMID: 32497284 DOI: 10.1111/jfbc.13313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022]
Abstract
The present study was to investigate the effective role of renewable sources of Ca+2 from eggshell (ES) with different doses to restrict obesity disorders. Rats were classified as follows, G1 : normal diet for 26 weeks; G2 : high-fat diet (HFD) for 26 weeks; G3 , G4 , and G5 were supplemented with HFD for 16 weeks and treated with 7.2 g Ca+2 ES/Kg rat chow, 18 g Ca+2 ES/Kg rat chow, and 2% diet containing fat (DCF), respectively, for the remaining 10 weeks. Results revealed a significant effect of the low dose of Ca+2 supplement in form of ES than high dose and 2% DCF; on basis of anthropometric parameters, lipid, leptin, adiponectin, thyroid hormones, Ca+2 , 25-hydroxyl vitamin-D, and oxidative and inflammatory parameters were regulated. Results were confirmed with the histopathological study. Therefore, it was concluded that Ca+2 supplementation can be used as a beneficial source for obesity management with anticholesterol actions. PRACTICAL APPLICATIONS: Obesity represented public health hazards. The eggshell is one of the waste products that contain a high percentage of Ca+2 . The current data exposed using a low dose of ES as a new source of Ca+2 supplement for treatment of HFD rats leads to significant enhancement of lipid profiles, liver enzymes, kidney functions, leptin, adiponectin, Ca+2 , 25(OH)-D, TSH, fT4, and PTH levels. Also, there was a reduction in weight gain, Bwt, BMI, BG, insulin, and HOMA-IR. Moreover, the oxidant-pro-oxidant system was improved in both hepatic and adipose tissues where NO and TBARS concentrations were diminished, and SOD specific activity was elevated. Additionally, TNF-α and ADAM17 expression were downregulated. Hence, it was concluded that there was good evidence that diets supplemented with ES were associated with the reduction of obesity complications especially regulating fat processing and storage in the body.
Collapse
Affiliation(s)
- Marwa El-Zeftawy
- Biochemistry Department, Faculty of Veterinary Medicine, New Valley University, New Valley, Egypt.,Biological Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Samar Abd-El Mohsen Ali
- Nutrition Department, High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Sally Salah
- Nutrition Department, Allied Medical Sciences, Pharos University, Alexandria, Egypt
| | - Hani S Hafez
- Faculty of Science, Zoology Department, Suez University, Suez, Egypt
| |
Collapse
|
15
|
Serum level of orexin A and its correlation with metabolic risk factors in type 2 diabetes mellitus patients. Int J Diabetes Dev Ctries 2019. [DOI: 10.1007/s13410-019-00728-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
16
|
Aranaz P, Navarro-Herrera D, Zabala M, Miguéliz I, Romo-Hualde A, López-Yoldi M, Martínez JA, Vizmanos JL, Milagro FI, González-Navarro CJ. Phenolic Compounds Inhibit 3T3-L1 Adipogenesis Depending on the Stage of Differentiation and Their Binding Affinity to PPARγ. Molecules 2019; 24:molecules24061045. [PMID: 30884812 PMCID: PMC6470710 DOI: 10.3390/molecules24061045] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/12/2022] Open
Abstract
Phenolic compounds might modulate adiposity. Here, we report our observation that polyphenols and phenolic acids inhibit adipogenesis in 3T3-L1 with different intensity depending on the family and the stage of differentiation. While quercetin and resveratrol inhibited lipid accumulation along the whole process of differentiation, apigenin and myricetin were active during the early and latest stages, but not intermediate, contrary to hesperidin. The activity of phenolic acids was limited to the early stages of the differentiation process, except p-coumaric and ellagic acids. This anti-adipogenic effect was accompanied by down-regulation of Scd1 and Lpl. Molecular docking analysis revealed that the inhibitory activity of these phenolic compounds over the early stages of adipogenesis exhibits a significant correlation (r = 0.7034; p = 0.005) with their binding affinity to the ligand-binding domain of PPARγ. Results show that polyphenols and phenolic acids would interact with specific residues of the receptor, which could determine their potential anti-adipogenic activity during the early stages of the differentiation. Residues Phe264, His266, Ile281, Cys285 and Met348 are the most frequently involved in these interactions, which might suggest a crucial role for these amino acids modulating the activity of the receptor. These data contribute to elucidate the possible mechanisms of phenolic compounds in the control of adipogenesis.
Collapse
Affiliation(s)
- Paula Aranaz
- Centre for Nutrition Research, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
| | - David Navarro-Herrera
- Centre for Nutrition Research, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
- Department of Biochemistry and Genetics, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
| | - María Zabala
- Centre for Nutrition Research, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
| | - Itziar Miguéliz
- Centre for Nutrition Research, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
| | - Ana Romo-Hualde
- Centre for Nutrition Research, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
| | - Miguel López-Yoldi
- Centre for Nutrition Research, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
| | - J Alfredo Martínez
- Centre for Nutrition Research, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
- Department of Nutrition, Food Science and Physiology, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
- Navarra Institute of Health Research (IdiSNA), 31008 Pamplona, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERObn); Instituto de Salud Carlos III, Monforte de Lemos 3-5, 28029 Madrid, Spain.
| | - José Luis Vizmanos
- Department of Biochemistry and Genetics, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
| | - Fermín I Milagro
- Centre for Nutrition Research, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain.
- Navarra Institute of Health Research (IdiSNA), 31008 Pamplona, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERObn); Instituto de Salud Carlos III, Monforte de Lemos 3-5, 28029 Madrid, Spain.
| | | |
Collapse
|
17
|
Zhu Y, Zhou P, Zhang L, Li J, Gao F, Zhou G. Effects of dietary crude protein levels and cysteamine supplementation on meat quality and related indices of finishing pigs. CANADIAN JOURNAL OF ANIMAL SCIENCE 2019. [DOI: 10.1139/cjas-2016-0095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this study was to investigate the effects of dietary crude protein levels and cysteamine (CS) supplementation on meat quality and related indices in longissimus dorsi muscle of finishing pigs. One hundred and twenty barrows were randomly allocated to a 2 × 2 factorial arrangement with five replicates of six pigs each. The primary variations were crude protein levels (14% or 10%) and CS supplemental levels (0 or 140 mg kg−1). After 41 d, 10 pigs per treatment were slaughtered. The results showed that low-protein level diets (LPDs) decreased Warner–Bratzler shear force (P < 0.01) and increased the content of intramuscular fat (P < 0.01). The mRNA expressions of lipogenic genes were up-regulated (P < 0.01), and the mRNA expressions of lipolytic genes were down-regulated (P < 0.01) in pigs fed LPD. LPDs increased the mRNA expressions of μ-calpain, and decreased the mRNA expression of calpastatin (P < 0.01). In addition, CS supplementation increased the mRNA expression of μ-calpain (P < 0.01). In conclusion, LPD improved the meat quality probably through regulating the lipogenesis, lipolysis, and the proteolysis process in muscle. The CS supplementation did not affect the meat quality of finishing pigs. Moreover, no significant interaction between dietary protein levels and CS supplementation for the meat quality of finishing pigs was observed.
Collapse
Affiliation(s)
- Y.P. Zhu
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
| | - P. Zhou
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
| | - L. Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
| | - J.L. Li
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
| | - F. Gao
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
| | - G.H. Zhou
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
| |
Collapse
|
18
|
Jones LM, Legge M. Plasma fatty acids as markers for desaturase and elongase activities in spinal cord injured males. J Spinal Cord Med 2019; 42:163-170. [PMID: 29319436 PMCID: PMC6419623 DOI: 10.1080/10790268.2017.1405154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To investigate the use of surrogate plasma fatty acid analysis to provide further insights into the underlying adiposity and the development of metabolic syndrome in men with spinal cord injury (SCI). DESIGN Case-control, cross-sectional study. SETTING Community-based individuals with spinal cord injury and healthy controls. PARTICIPANTS Twenty men with SCI age, height and weight matched with 20 able-bodied controls. OUTCOME MEASURES Lean tissue (LTM) and fat mass (FM) were determined using dual energy X-ray absorptiometry. Fasting blood samples were taken for analysis of fatty acids, adiponectin, insulin, glucose and leptin. Enzymatic indices were calculated using relevant fatty acids. RESULTS Total FM, leptin, stearoyl coenzyme A desaturase (SCD) Δ9 (SCD-16, 16:1/16:0, and SCD-18, 18:1/18:0) indices and Δ6 desaturase index were significantly higher (P < 0.05) in the SCI group than the controls. Significant differences between the groups was observed for several individual fatty acids. Correlational analysis revealed a different pattern between blood biomarkers and indices of SCDs, de novo lipogenesis and elongase. Associations between the desaturase and elongase indices and biomarkers in the controls followed those reported elsewhere for able bodied participants; the same associations were not observed in the SCI group. CONCLUSION We have identified disturbances in fatty acid biosynthesis in SCI individuals likely associated with the development of adipose tissue below the lesion and a decrease in LTM. Loss of LTM may disturb the normal skeletal muscle-fatty acid metabolic processes leading to the disruption of metabolic homeostasis, previously identified in persons with SCI.
Collapse
Affiliation(s)
- Lynnette M. Jones
- School of Sport and Exercise Sciences, University of Otago, Dunedin, New Zealand,Correspondence to: Lynnette M. Jones, School of Sport and Exercise Sciences, University of Otago, PO Box 56, Dunedin9054, New Zealand.
| | - Michael Legge
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
19
|
Wang H, Xu PF, Li JY, Liu XJ, Wu XY, Xu F, Xie BC, Huang XM, Zhou ZH, Kayoumu A, Liu G, Huang W. Adipose tissue transplantation ameliorates lipodystrophy-associated metabolic disorders in seipin-deficient mice. Am J Physiol Endocrinol Metab 2019; 316:E54-E62. [PMID: 30457912 DOI: 10.1152/ajpendo.00180.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Seipin deficiency is responsible for type 2 congenital generalized lipodystrophy with severe loss of adipose tissue and can lead to hepatic steatosis, insulin resistance (IR), and dyslipidemia in humans. Adipose tissue secretes many adipokines that are central to the regulation of metabolism. In this study, we investigated whether transplantation of normal adipose tissue could ameliorate severe hepatic steatosis, IR, and dyslipidemia in lipoatrophic seipin knockout (SKO) mice. Normal adipose tissue from wild-type mice was transplanted into 6-wk-old SKO mice. At 4 mo after adipose tissue transplantation (AT), the transplanted fat survived with detectable blood vessels, and the reduced levels of plasma leptin, a major adipokine, were dramatically increased. Severe hepatic steatosis, IR, and dyslipidemia in SKO mice were ameliorated after AT. In addition, abnormal hepatic lipogenesis and β-oxidation gene expression in SKO mice were improved after AT. Our results suggest that AT may be an effective treatment to improve lipodystrophy-associated metabolic disorders.
Collapse
Affiliation(s)
- Huan Wang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University , Beijing , China
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
| | - Peng-Fei Xu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
- Institute of Military Training Related Medical Science of PLA, 150th Central Hospital of PLA , Luoyang, Henan , China
| | - Jing-Yi Li
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
| | - Xue-Jing Liu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
| | - Xiao-Yue Wu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
| | - Fang Xu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
| | - Bei-Chen Xie
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
| | - Xiao-Min Huang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
| | - Zi-Hao Zhou
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
| | - Abudurexiti Kayoumu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
| | - George Liu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
| | - Wei Huang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center , Beijing , China
| |
Collapse
|
20
|
Hayakawa J, Wang M, Wang C, Han RH, Jiang ZY, Han X. Lipidomic analysis reveals significant lipogenesis and accumulation of lipotoxic components in ob/ob mouse organs. Prostaglandins Leukot Essent Fatty Acids 2018; 136:161-169. [PMID: 28110829 PMCID: PMC6203299 DOI: 10.1016/j.plefa.2017.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/03/2016] [Accepted: 01/03/2017] [Indexed: 12/13/2022]
Abstract
To further understand the role of lipogenesis and lipotoxicity in the development of obesity and diabetes, lipidomes of various organs from ob/ob mice and their wild type controls were analyzed by shotgun lipidomics at 10, 12, and 16 weeks of age. We observed that the amounts of fatty acyl (FA) chains corresponding to those from de novo synthesis (e.g., 16:0, 16:1, and 18:1 FA) were substantially elevated in ob/ob mice, consistent with increased expression of genes and proteins involved in biosynthesis. Polyunsaturated fatty acid species were moderately increased in the examined tissues of ob/ob mice, since they can only be absorbed from diets or elongated from the ingested n-3 or n-6 FA. Different profiles of FA chains between ob/ob mouse liver and skeletal muscle reflect diverging lipogenesis pathways in these organs. Amounts of vaccenic acids (i.e., 18:1(n-7) FA) in 12- and 16-week ob/ob mouse liver were significantly increased compared to their controls, indicating enhanced de novo synthesis of this acid through 16:1(n-7) FA in the liver starting at 12 weeks of age. Coincidentally, synthesis of triacylglycerol from monoacylglycerol in the liver was also increased in ob/ob mice starting at 12 weeks of age, as revealed by simulation of triacylglycerol synthesis. Moreover, levels of lipotoxic lipid classes were significantly higher in ob/ob mice than their age-matched controls, supporting the notion that elevated lipotoxic components are tightly associated with insulin resistance in ob/ob mice. Taken together, the current study revealed that lipogenesis and lipotoxicity in ob/ob mice likely contribute to insulin resistance and provides great insights into the underlying mechanisms of diabetes and obesity.
Collapse
Affiliation(s)
- Jun Hayakawa
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Miao Wang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Chunyan Wang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Rowland H Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Zhen Y Jiang
- Department of Pharmacology & Experimental Therapeutics, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Xianlin Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA.
| |
Collapse
|
21
|
Al-Muzafar HM, Amin KA. Thiazolidinedione induces a therapeutic effect on hepatosteatosis by regulating stearoyl-CoA desaturase-1, lipase activity, leptin and resistin. Exp Ther Med 2018; 16:2938-2948. [PMID: 30214514 PMCID: PMC6125847 DOI: 10.3892/etm.2018.6563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 04/06/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatosteatosis is a disease present worldwide, which presents a number of health problems. Recently, thiazolidinedione (TZD) has been used as a therapy for lipid disorders. The present study demonstrates the potential of TZD as a treatment for hepatosteatosis and its mechanism of action, particularly focusing on its role in lipid metabolism. A total of 60 (80-90 g) rats were divided into three groups: A normal group with a standard diet, a high-fat, high-carbohydrate diet (HFCD) group or a HFCD+TZD group (n=20/group). The HFCD induced hepatosteatosis over a period of 12 weeks and the HFCD+TZD group were administered TZD in weeks 13-16. Blood and tissue samples were collected to measure hepatic function, the lipid profile, metabolism and hormone biomarkers, including serum triglyceride (TG), lipoprotein lipase (LPL), stearoyl-CoA desaturase (SCD-1), leptin and resistin. The HFCD-fed rats exhibited a significant increase in serum TG, total cholesterol, low-density lipoproteins, alanine transaminase and bilirubin compared with the normal group as well as a significant decrease in high-density lipoprotein. In addition, serum leptin and resistin were significantly elevated in the HFCD group compared with the normal group. The administration of TZD significantly increased SCD-1 activity and significantly inhibited LPL activity. It also attenuated the changes in the lipid profiles and normalized serum leptin and resistin levels. The results of the present study indicated that HFCD induced lipid abnormalities associated with hypertriglyceridemia, hypercholesterolemia and hepatosteatosis. These changes resulted from disruption to leptin and resistin, which may be due to alterations in LPL and SCD-1 activity. TZD mitigated the effects of HFCD-induced hepatosteatosis, indicating a possible regulatory effect of TZD in the development of hepatosteatosis. The authors suggest that the manipulation of SCD-1 and lipase by TZD may be useful as a treatment for hepatosteatosis.
Collapse
Affiliation(s)
- Hessah Mohammed Al-Muzafar
- Department of Chemistry and Biochemistry, College of Science, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Kamal Adel Amin
- Department of Chemistry and Biochemistry, College of Science, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| |
Collapse
|
22
|
Liu C, Shen YJ, Tu QB, Zhao YR, Guo H, Wang J, Zhang L, Shi HW, Sun Y. Pedunculoside, a novel triterpene saponin extracted from Ilex rotunda, ameliorates high-fat diet induced hyperlipidemia in rats. Biomed Pharmacother 2018. [PMID: 29518607 DOI: 10.1016/j.biopha.2018.02.131] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pedunculoside (PE) is a novel triterpene saponin extracted from the dried barks of Ilex rotunda Thunb. The present study aims to explore lipid-lowering effects of PE on hyperlipidemia rat induced by high-fat diet. The rats were fed with the high-fat diet and subjected to intragastric administration of PE at doses of 30, 15, or 5 mg/kg daily for 7 weeks. The results demonstrated that treatment with PE for 7-week dramatically decreased serum total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) and reduced liver TC in hyperlipidemia rat induced by high-fat diet. Furthermore, the results also showed that PE modulated the expression of enzymes involved in lipid metabolism including peroxisome proliferator-activated receptor α (PPAR-α), sterol regulatory element-binding protein 1 (SREBP-1), fatty acid synthase (FAS) and stearoyl CoA desaturase-1 (SCD-1) mRNA in liver. Besides, PE-treated group decreased weights and diameters of epididymal adipose hyperlipidemia rat. Mechanism study demonstrated that PE regulated PPAR-γ, CCAAT/Enhancer-binding Protein α (C/EBPα)、and SREBP-1 expression as well as inhibited phosphorylation of AMPK in MDI (methylisobutylxanthine, dexamethasone, insulin) induced-3T3L1 cells. Molecular Docking confirmed interaction between PE with proteins involving PPAR-γ, C/EBPα and SREBP-1. In summary, these findings may support that PE is a novel lipid-lowering drug candidate.
Collapse
Affiliation(s)
- Chang Liu
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China; School of Pharmacy, University of Rhode Island, RI, 02881, United States
| | - Yan-Jun Shen
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China
| | - Qing-Bo Tu
- College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China
| | - Yan-Ran Zhao
- College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China
| | - Hao Guo
- School of Pharmacy, University of Rhode Island, RI, 02881, United States; Department of Dermatology, No. 1 Hospital of China Medical University, 155N. Nanjing Street, Shenyang 110001, PR China
| | - Juan Wang
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China; College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China
| | - Li Zhang
- School of Pharmacy, University of Missouri-Kansas City, MO, 64108, United States
| | - Hua-Wei Shi
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China
| | - Yun Sun
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China; College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China.
| |
Collapse
|
23
|
Sihag J, Jones PJH. Oleoylethanolamide: The role of a bioactive lipid amide in modulating eating behaviour. Obes Rev 2018; 19:178-197. [PMID: 29124885 DOI: 10.1111/obr.12630] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 12/13/2022]
Abstract
Fatty acid ethanolamides are lipid mediators that regulate a plethora of physiological functions. One such bioactive lipid mediator, oleoylethanolamide (OEA), is a potent agonist of the peroxisome proliferator-activated receptor-alpha (PPAR-α), which modulates increased expression of the fatty acid translocase CD36 that enables the regulation of feeding behaviour. Consumption of dietary fat rich in oleic acid activates taste receptors in the gut activating specific enzymes that lead to the formation of OEA. OEA further combines with PPAR-α to enable fat oxidation in the liver, resulting in enhanced energy production. Evidence suggests that sustained ingestion of a high-fat diet abolishes the anorexic signal of OEA. Additionally, malfunction of the enterocyte that transforms oleic acid produced during fat digestion into OEA might be responsible for reduced satiety and hyperphagia, resulting in overweight and obesity. Thus, OEA anorectic signalling may be an essential element of the physiology and metabolic system regulating dietary fat intake and obesity. The evidence reviewed in this article indicates that intake of oleic acid, and thereby the resulting OEA imparting anorexic properties, is dependent on CD36, PPAR-α, enterocyte fat sensory receptors, histamine, oxytocin and dopamine; leading to increased fat oxidation and enhanced energy expenditure to induce satiety and increase feeding latency; and that a disruption in any of these systems will cease/curb fat-induced satiety.
Collapse
Affiliation(s)
- J Sihag
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, Winnipeg, Manitoba, Canada
| | - P J H Jones
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
24
|
Chen X, Ding Y, Song J, Kan J. Hypolipidaemic effect and mechanism of paprika seed oil on Sprague-Dawley rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2017; 97:4242-4249. [PMID: 28251657 DOI: 10.1002/jsfa.8300] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 06/06/2023]
Abstract
BACKGROUND Details regarding the functional properties of paprika seed oil are relatively scarce. In this study the hypolipidaemic effects and mechanisms of paprika seed oil on Sprague-Dawley rats are explored, which may improve the usage of paprika seed source and provide a theoretical basis of paprika seed oil for the alleviation of hyperlipidaemia. RESULTS In capsaicin and paprika seed oil (PSO) groups, total cholesterol (TC) and total triglyceride (TG) in serum and liver lipids of rats were significantly decreased (P < 0.05). The contents of serum HDL cholesterol were increased and the contents of serum LDL cholesterol were decreased (P < 0.05). Real-time PCR analyses revealed that the hepatic mRNA expression of fatty acid synthetase (FAS) is decreased and the expression levels of HSL is increased (P < 0.05). The mRNA expression of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) is decreased and the expression levels of low-density lipoprotein receptor (LDLR) is significantly improved (P < 0.05). The cholesterol 7-hydroxylase (CYP7A1) expression is regulated to control the cholesterol-to-bile acid transformation and cholesterol excretion is promoted. Capsaicin and unsaturated fatty acid PSO can activate and improve the mRNA expression of transient receptor potential vanilloid type-1 (TRPV1) and peroxisome proliferators-activated receptors (PPARα). CONCLUSION The hypolipidaemic effects of paprika seed oil (PSO) may be attributed to the inhibition of lipid synthesis via suppressing the expression of HMG-CoAR, CYP7A1 and FAS, meanwhile, promoting the metabolism and excretion of lipids via up-regulating the expression of LDLR, HSL, TRPV1 and PPARα. © 2017 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xuhui Chen
- College of Food Science, Southwest University, Chongqing, China
- Laboratory of Quality 1 Safety Risk Assessment for Agro-products on Storage and Preservation, Chongqing, China
| | - Yongbo Ding
- College of Food Science, Southwest University, Chongqing, China
- Laboratory of Quality 1 Safety Risk Assessment for Agro-products on Storage and Preservation, Chongqing, China
| | - Jiaxin Song
- College of Food Science, Southwest University, Chongqing, China
- Laboratory of Quality 1 Safety Risk Assessment for Agro-products on Storage and Preservation, Chongqing, China
| | - Jianquan Kan
- College of Food Science, Southwest University, Chongqing, China
- Laboratory of Quality 1 Safety Risk Assessment for Agro-products on Storage and Preservation, Chongqing, China
| |
Collapse
|
25
|
Poloni S, Spritzer PM, Mendes RH, D'Almeida V, Castro K, Sperb-Ludwig F, Kugele J, Tucci S, Blom HJ, Schwartz IVD. Leptin concentrations and SCD-1 indices in classical homocystinuria: Evidence for the role of sulfur amino acids in the regulation of lipid metabolism. Clin Chim Acta 2017; 473:82-88. [PMID: 28801090 DOI: 10.1016/j.cca.2017.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 08/05/2017] [Accepted: 08/07/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND We describe body composition, lipid metabolism and Stearoyl-CoA desaturase-1 (SCD-1) indices in patients with classical homocystinuria (HCU). METHODS Eleven treated HCU patients and 16 healthy controls were included. Body composition and bone mineral density were assessed by dual X-ray absorptiometry. Sulfur amino acids (SAA) and their derivatives (total homocysteine, cysteine, methionine, S-adenosylmethionine, S-adenosylhomocysteine, and glutathione), lipids (free fatty acids, acylcarnitines, triglycerides and lipoproteins), glucose, insulin, leptin, adiponectin, and isoprostanes were measured in plasma. Insulin resistance was evaluated by HOMA-IR. To estimate liver SCD-1 activity, SCD-16 [16:1(n-7)/16:0] and SCD-18 [18:1(n-9)/18:0] desaturation indices were determined. RESULTS In HCU patients, SCD-16 index was significantly reduced (p=0.03). A trend of an association of SCD-16 index with cysteine was observed (r=0.624, p=0.054). HCU patients displayed lower lean mass (p<0.05), with no differences in fat mass percentage. Leptin and low-density lipoprotein concentrations were lower in HCU patients (p<0.05). Femur bone mineral density Z-scores were correlated with plasma cysteine (r=0.829; p=0.04) and total homocysteine (r=-0.829; p=0.04) in HCU patients. CONCLUSIONS We report alterations in leptin and SCD-1 in HCU patients. These results agree with previous findings from epidemiologic and animal studies, and support a role for SAA on lipid homeostasis.
Collapse
Affiliation(s)
- Soraia Poloni
- Post-Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences) - Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Poli Mara Spritzer
- Gynecological Endocrinology Unit, Division of Endocrinology, Hospital de Clinicas de Porto Alegre, Department of Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Roberta H Mendes
- BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences) - Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Vânia D'Almeida
- Laboratory of Inborn Errors of Metabolism, Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Kamila Castro
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Fernanda Sperb-Ludwig
- BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences) - Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Johanna Kugele
- Laboratory for Clinical Biochemistry and Metabolism, University Medical Center, Freiburg, Germany
| | - Sara Tucci
- Laboratory for Clinical Biochemistry and Metabolism, University Medical Center, Freiburg, Germany
| | - Henk J Blom
- Laboratory for Clinical Biochemistry and Metabolism, University Medical Center, Freiburg, Germany
| | - Ida V D Schwartz
- Post-Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences) - Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
26
|
Hehar H, Ma I, Mychasiuk R. Intergenerational Transmission of Paternal Epigenetic Marks: Mechanisms Influencing Susceptibility to Post-Concussion Symptomology in a Rodent Model. Sci Rep 2017; 7:7171. [PMID: 28769086 PMCID: PMC5541091 DOI: 10.1038/s41598-017-07784-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 06/30/2017] [Indexed: 01/29/2023] Open
Abstract
Epigenetic transmission of phenotypic variance has been linked to paternal experiences prior to conception and during perinatal development. Previous reports indicate that paternal experiences increase phenotypic heterogeneity and may contribute to offspring susceptibility to post-concussive symptomology. This study sought to determine if epigenetic tags, specifically DNA methylation of promoter regions, are transmitted from rodent fathers to their sons. Using MethyLight, promoter methylation of specific genes involved in recovery from concussion and brain plasticity were analyzed in sperm and brain tissue. Promoter methylation in sperm differed based on paternal experience. Differences in methylation were often identified in both the sperm and brain tissue obtained from their sons, demonstrating transmission of epigenetic tags. For certain genes, methylation in the sperm was altered following a concussion suggesting that a history of brain injury may influence paternal transmission of traits. As telomere length is paternally inherited and linked to neurological health, this study examined paternally derived differences in telomere length, in both sperm and brain. Telomere length was consistent between fathers and their sons, and between brain and sperm, with the exception of the older fathers. Older fathers exhibited increased sperm telomere length, which was not evident in sperm or brain of their sons.
Collapse
Affiliation(s)
- Harleen Hehar
- Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Department of Psychology, Calgary, Alberta, Canada
| | - Irene Ma
- Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Department of Psychology, Calgary, Alberta, Canada
| | - Richelle Mychasiuk
- Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Department of Psychology, Calgary, Alberta, Canada.
| |
Collapse
|
27
|
Horska K, Ruda-Kucerova J, Karpisek M, Suchy P, Opatrilova R, Kotolova H. Depot risperidone-induced adverse metabolic alterations in female rats. J Psychopharmacol 2017; 31:487-499. [PMID: 28347258 DOI: 10.1177/0269881117691466] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Atypical antipsychotics are associated with adverse metabolic effects including weight gain, increased adiposity, dyslipidaemia, alterations in glucose metabolism and insulin resistance. Increasing evidence suggests that metabolic dysregulation precedes weight gain development. The aim of this study was to evaluate alterations in adipokines, hormones and basic serum biochemical parameters induced by chronic treatment with depot risperidone at two doses (20 and 40 mg/kg) in female Sprague-Dawley rats. Dose-dependent metabolic alterations induced by risperidone after 6 weeks of treatment were revealed. Concomitant to weight gain and increased liver weight, an adverse lipid profile with an elevated triglyceride level was observed in the high exposure group, administered a 40 mg/kg dose repeatedly, while the low dose exposure group, administered a 20 mg/kg dose, developed weight gain without alterations in the lipid profile and adipokine levels. An initial peak in leptin serum level after the higher dose was observed in the absence of weight gain. This finding may indicate that the metabolic alterations observed in this study are not consequent to body weight gain. Taken together, these data may support the primary effects of atypical antipsychotics on peripheral tissues.
Collapse
Affiliation(s)
- Katerina Horska
- 1 Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - Jana Ruda-Kucerova
- 2 Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Michal Karpisek
- 1 Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic.,3 R&D Department, Biovendor - Laboratorni Medicina, Brno, Czech Republic
| | - Pavel Suchy
- 1 Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - Radka Opatrilova
- 4 Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - Hana Kotolova
- 1 Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| |
Collapse
|
28
|
Nakamura K, Fukunishi S, Yokohama K, Ohama H, Tsuchimoto Y, Asai A, Tsuda Y, Higuchi K. A long-lasting dipeptidyl peptidase-4 inhibitor, teneligliptin, as a preventive drug for the development of hepatic steatosis in high-fructose diet-fed ob/ob mice. Int J Mol Med 2017; 39:969-983. [DOI: 10.3892/ijmm.2017.2899] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 02/01/2017] [Indexed: 11/06/2022] Open
|
29
|
Yokohama K, Fukunishi S, Ii M, Nakamura K, Ohama H, Tsuchimoto Y, Asai A, Tsuda Y, Higuchi K. Rosuvastatin as a potential preventive drug for the development of hepatocellular carcinoma associated with non-alcoholic fatty liver disease in mice. Int J Mol Med 2016; 38:1499-1506. [PMID: 28025996 DOI: 10.3892/ijmm.2016.2766] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) represents approximately 85% of all primary liver cancer cases. Non-alcoholic fatty liver disease (NAFLD) is one of the risk factors for HCC. NAFLD occurs in patients with components of metabolic syndrome, such as type 2 diabetes mellitus, obesity, hypertension and hyperlipidemia. Therefore, hyperlipidemia also represents a patient population at risk for HCC that can readily be identified. Rosuvastatin, a 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitor, has exhibited a more potent affinity for the active site of HMG-CoA reductase than other statins. In addition, the hepatic uptake of rosuvastatin in rats has been found to be more selective and efficient than that with other drugs. Furthermore, the cytoprotective effects of rosuvastatin against ischemic injury have been clearly reported. Thus, in this study, we aimed to determine the role of rosuvastatin as a preventive drug in HCC associated with NAFLD. STAM mice, which developed HCC from NAFLD by being fed a high-fat diet (HFD), were divided into a group in which a HFD was given to the mice for 15 weeks (n=8) and another in which a HFD supplemented with 0.00125% rosuvastatin was given to the mice for 15 weeks (n=8). Rosuvastatin inhibited the development of hepatic tumors in the mice with NAFLD induced by a specific diet both macroscopically and histologically. Rosuvastatin significantly decreased the expression levels of pro-inflammatry cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-6 and transforming growth factor (TGF)-β1. Tumor aggressiveness is mediated by angiogenic factors. Therefore, we examined the hepatic mRNA expression of vascular endothelial growth factor receptor (VEGFR), epidermal growth factor receptor (EGFR) and platelet-derived growth factor (PDGF). The hepatic expression of these factors significantly decreased in the rousvastin-fed mice. Our results thus suggest rosuvastatin that prevents carcinogenesis and improves the hepatic background. Our data suggest that rosuvastatin has potential for use as a preventive drug for the development of HCC associated with NAFLD in mice.
Collapse
Affiliation(s)
- Keisuke Yokohama
- Second Department of Internal Medicine, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Shinya Fukunishi
- Second Department of Internal Medicine, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Masaaki Ii
- Department of Pharmacology, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Ken Nakamura
- Second Department of Internal Medicine, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Hideko Ohama
- Second Department of Internal Medicine, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Yusuke Tsuchimoto
- Second Department of Internal Medicine, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Akira Asai
- Second Department of Internal Medicine, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Yasuhiro Tsuda
- Second Department of Internal Medicine, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Kazuhide Higuchi
- Second Department of Internal Medicine, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| |
Collapse
|
30
|
Niewiadomski J, Zhou JQ, Roman HB, Liu X, Hirschberger LL, Locasale JW, Stipanuk MH. Effects of a block in cysteine catabolism on energy balance and fat metabolism in mice. Ann N Y Acad Sci 2016; 1363:99-115. [PMID: 26995761 DOI: 10.1111/nyas.13021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 12/08/2015] [Accepted: 01/19/2016] [Indexed: 01/14/2023]
Abstract
To gain further insights into the effects of elevated cysteine levels on energy metabolism and the possible mechanisms underlying these effects, we conducted studies in cysteine dioxygenase (Cdo1)-null mice. Cysteine dioxygenase (CDO) catalyzes the first step of the major pathway for cysteine catabolism. When CDO is absent, tissue and plasma cysteine levels are elevated, resulting in enhanced flux of cysteine through desulfhydration reactions. When Cdo1-null mice were fed a high-fat diet, they gained more weight than their wild-type controls, regardless of whether the diet was supplemented with taurine. Cdo1-null mice had markedly lower leptin levels, higher feed intakes, and markedly higher abundance of hepatic stearoyl-CoA desaturase 1 (SCD1) compared to wild-type control mice, and these differences were not affected by the fat or taurine content of the diet. Thus, reported associations of elevated cysteine levels with greater weight gain and with elevated hepatic Scd1 expression are also seen in the Cdo1-null mouse model. Hepatic accumulation of acylcarnitines suggests impaired mitochondrial β-oxidation of fatty acids in Cdo1-null mice. The strong associations of elevated cysteine levels with excess H2 S production and impairments in energy metabolism suggest that H2 S signaling could be involved.
Collapse
Affiliation(s)
| | - James Q Zhou
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Heather B Roman
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Xiaojing Liu
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | | | - Jason W Locasale
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Martha H Stipanuk
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| |
Collapse
|
31
|
Ruan J, Zhang Y, Yuan J, Xin L, Xia J, Liu N, Mu Y, Chen Y, Yang S, Li K. A long-term high-fat, high-sucrose diet in Bama minipigs promotes lipid deposition and amyotrophy by up-regulating the myostatin pathway. Mol Cell Endocrinol 2016; 425:123-32. [PMID: 26850224 DOI: 10.1016/j.mce.2016.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 01/29/2016] [Accepted: 02/01/2016] [Indexed: 01/08/2023]
Abstract
Skeletal muscle is as an important regulator of blood glucose and glycolipid metabolism and is closely related to motor ability. The underlying mechanisms by which dietary ectopic lipids in skeletal muscle prevents muscle growth remain elusive. We utilized miniature Bama swine as a model to mimic human obesity using prolonged dietary induction. After 23 months on a high-fat, high-sucrose diet, metabolic disorders were induced in the animals, which exhibited increased body weight, extensive lipid deposition in the skeletal muscle and amyotrophy. Microarray profiles demonstrated the up-regulation of genes related to fat deposition and muscle growth inhibition. We outline a clear potential pathway that in combination with increased 11β-hydroxysteroid dehydrogenase type 1, promotes expression of a major inhibitor, myostatin, by converting corticosterone to cortisol, which leads to the growth inhibition of skeletal muscle. This research provides new insights into the treatment of muscle diseases induced by obesity.
Collapse
Affiliation(s)
- Jinxue Ruan
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan West Road, Beijing, 100193, PR China; Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Science, Jilin University, Changchun, 130012, PR China
| | - Yuanyuan Zhang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, PR China
| | - Jing Yuan
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan West Road, Beijing, 100193, PR China; College of Animal Science, Yangtz University, Jinzhou, 434023, Hubei, PR China
| | - Leilei Xin
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan West Road, Beijing, 100193, PR China
| | - Jihan Xia
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan West Road, Beijing, 100193, PR China
| | - Nan Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan West Road, Beijing, 100193, PR China; Agricutural Genomes Institute at Shenzhen, CAAS, Shenzhen, 518120, PR China
| | - Yulian Mu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan West Road, Beijing, 100193, PR China
| | - Yaoxing Chen
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, PR China
| | - Shulin Yang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan West Road, Beijing, 100193, PR China.
| | - Kui Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan West Road, Beijing, 100193, PR China; Agricutural Genomes Institute at Shenzhen, CAAS, Shenzhen, 518120, PR China
| |
Collapse
|
32
|
Sujishi T, Fukunishi S, Ii M, Nakamura K, Yokohama K, Ohama H, Tsuchimoto Y, Asai A, Tsuda Y, Higuchi K. Sitagliptin can inhibit the development of hepatic steatosis in high-fructose diet-fed ob/ob mice. J Clin Biochem Nutr 2015; 57:244-53. [PMID: 26566312 PMCID: PMC4639593 DOI: 10.3164/jcbn.15-84] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 08/03/2015] [Indexed: 12/14/2022] Open
Abstract
The beneficial effect of dipeptidyl peptidase-4 inhibition on diet-induced extra-pancreatic effects, especially on liver tissue remains poorly understood. Thus, we made the experimental designs as follows; five-week-old male ob/ob mice, which develop type 2 diabetic mellitus and nonalcoholic fatty liver disease by taking a high-carbohydrate diet (HCD), were divided into a group in which a HCD was given for 8 weeks as control, and another in which a HCD added with 0.0018% sitagliptin was given for 8 weeks. Hepatic steatosis was seen in all mice, but the mean grade of steatosis in the sitagliptin-administrated mice was significantly decreased. The acetyl-CoA concentrations were lower in sitagliptin-administrated mice, although the differences were not significant. However, the malonyl-CoA concentrations were significantly lower in sitagliptin-administrated mice. The expression of acetyl-CoA carboxylase 1 was inhibited in sitagliptin-administrated mice, irrespective of expressions of carbohydrate responsive element-binding protein (ChREBP) or sterol regulatory element-binding protein (SREBP)-1c. In conclusion, sitagliptin may affect the development of nonalcoholic fatty liver disease by inhibiting the production of malonyl-CoA and thus synthesis of fatty acids in the liver.
Collapse
Affiliation(s)
- Tetsuya Sujishi
- 2nd Department of Internal Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Shinya Fukunishi
- 2nd Department of Internal Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Masaaki Ii
- Department of Pharmacology, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Ken Nakamura
- 2nd Department of Internal Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Keisuke Yokohama
- 2nd Department of Internal Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Hideko Ohama
- 2nd Department of Internal Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Yusuke Tsuchimoto
- 2nd Department of Internal Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Akira Asai
- 2nd Department of Internal Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Yasuhiro Tsuda
- 2nd Department of Internal Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Kazuhide Higuchi
- 2nd Department of Internal Medicine, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| |
Collapse
|
33
|
Stearoyl-CoA Desaturase-1: Is It the Link between Sulfur Amino Acids and Lipid Metabolism? BIOLOGY 2015; 4:383-96. [PMID: 26046927 PMCID: PMC4498306 DOI: 10.3390/biology4020383] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 05/02/2015] [Accepted: 05/14/2015] [Indexed: 12/11/2022]
Abstract
An association between sulfur amino acids (methionine, cysteine, homocysteine and taurine) and lipid metabolism has been described in several experimental and population-based studies. Changes in the metabolism of these amino acids influence serum lipoprotein concentrations, although the underlying mechanisms are still poorly understood. However, recent evidence has suggested that the enzyme stearoyl-CoA desaturase-1 (SCD-1) may be the link between these two metabolic pathways. SCD-1 is a key enzyme for the synthesis of monounsaturated fatty acids. Its main substrates C16:0 and C18:0 and products palmitoleic acid (C16:1) and oleic acid (C18:1) are the most abundant fatty acids in triglycerides, cholesterol esters and membrane phospholipids. A significant suppression of SCD-1 has been observed in several animal models with disrupted sulfur amino acid metabolism, and the activity of SCD-1 is also associated with the levels of these amino acids in humans. This enzyme also appears to be involved in the etiology of metabolic syndromes because its suppression results in decreased fat deposits (regardless of food intake), improved insulin sensitivity and higher basal energy expenditure. Interestingly, this anti-obesogenic phenotype has also been described in humans and animals with sulfur amino acid disorders, which is consistent with the hypothesis that SCD-1 activity is influenced by these amino acids, in particularly cysteine, which is a strong and independent predictor of SCD-1 activity and fat storage. In this narrative review, we discuss the evidence linking sulfur amino acids, SCD-1 and lipid metabolism.
Collapse
|
34
|
Burdeos GC, Nakagawa K, Abe T, Kimura F, Miyazawa T. Tocotrienol modulates crucial lipid metabolism-related genes in differentiated 3T3-L1 preadipocytes. Food Funct 2015; 5:2221-7. [PMID: 25055730 DOI: 10.1039/c4fo00463a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Obesity and other lipid metabolism-related diseases have become more prevalent in recent years due to drastic lifestyle changes and dietary patterns. Unsaturated vitamin E, tocotrienol (T3), represents one of the most fascinating naturally occurring compounds that has the potential to influence a broad range of mechanisms underlying abnormal lipid metabolism processes. However, its efficacy and mechanism have been uncertain due to scarcity of data concerning the effect of T3 on lipid metabolism. In this study, we report a series of fascinating experimental findings on how T3 affects lipid metabolism in differentiated 3T3-L1 preadipocytes. Treatment with T3 (25 μM), especially δ and γ isomers, inhibited the accumulation of triglyceride and lipid droplets in differentiated 3T3-L1 cells. This manifestation was supported by mRNA and protein expression of crucial lipid metabolism-related genes. The present study provides a novel set of data pertaining to the possibility of T3 as an anti-metabolic disorder agent.
Collapse
Affiliation(s)
- Gregor Carpentero Burdeos
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan.
| | | | | | | | | |
Collapse
|
35
|
Yosunkaya Ş, Okur HK, Can Ü, Zamani A, Kutlu R. Impact of Continuous Positive Airway Pressure Treatment on Leptin Levels in Patients with Obstructive Sleep Apnea Syndrome. Metab Syndr Relat Disord 2015; 13:272-7. [PMID: 25867002 DOI: 10.1089/met.2014.0161] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND There is growing evidence that leptin regulation is altered in obstructive sleep apnea syndrome (OSAS). Several potential mechanisms have been purported to explain how sleep apnea may alter leptin levels. We investigated whether repeated apneas, hypoxia, or excessive daytime sleepiness influenced the levels of leptin in OSAS patients. We also evaluated whether a 3-month continuous positive airway pressure (CPAP) treatment affected leptin levels in patients. METHODS Randomly selected 31 untreated, otherwise healthy male, overweight [body mass index (BMI) >25 kg/m(2)] obstructive sleep apnea syndrome (OSAS) patients [apnea-hypopnea index (AHI) ≥15] and 25 control (AHI <5) were included in this study. To confirm the diagnosis, all subjects underwent standard polysomnography. Serum samples were taken at 07:00-08:00 a.m. after overnight fasting. The OSAS patients that had regular CPAP treatment (n=26) were re-evaulated 3 months later. RESULTS Leptin levels (50.5±17.5 grams/L in OSAS and 56.3±25.5 grams/L in controls) and lipid profiles (TC, TGs, HDL-C, and LDL-C) between patient and control groups did not differ (P>0.05). Leptin levels were not correlated with the AHI, oxygen saturation, or excessive daytime sleepiness. CPAP treatment did not significantly change the (BMI), waist and neck circumference, or leptin levels in OSAS patients. Furthermore, we found no correlation between the decrease in serum leptin levels and parameters that were improved by CPAP treatment. CONCLUSION Leptin levels and lipid profile of overweight subjects with and without OSAS were not different, and our results suggest that OSAS-related parameters and CPAP treatment do not play a significant role in the serum leptin levels.
Collapse
Affiliation(s)
- Şebnem Yosunkaya
- 1 Meram Medical Faculty, Department of Chest Disease, Necmettin Erbakan University , Konya, Turkey
| | - Hacer Kuzu Okur
- 2 Department of Chest Diseases and Thoracic Surgery, Fatih Sultan Mehmet Education and Research Hospital , Istanbul, Turkey
| | - Ümmügülsüm Can
- 3 Department of Biochemistry, Konya Education and Research Hospital , Konya, Turkey
| | - Adil Zamani
- 4 Meram Medical Faculty, Department of Chest Disease, Necmettin Erbakan University , Konya, Turkey
| | - Ruhuşen Kutlu
- 5 Meram Medical Faculty, Department of Family Physician, Necmettin Erbakan University , Konya, Turkey
| |
Collapse
|
36
|
Jia S, Hu Y, Zhang W, Zhao X, Chen Y, Sun C, Li X, Chen K. Hypoglycemic and hypolipidemic effects of neohesperidin derived from Citrus aurantium L. in diabetic KK-Aymice. Food Funct 2015; 6:878-86. [DOI: 10.1039/c4fo00993b] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The present study is to investigate the possible hypoglycemic and hypolipidemic effects of neohesperidin (NHP) derived fromCitrus aurantiumL.in vivo.
Collapse
Affiliation(s)
- Sheng Jia
- Laboratory of Fruit Quality Biology
- Zhejiang University
- Hangzhou
- China
| | - Ying Hu
- Department of Ultrasonography
- The First Affiliated Hospital
- College of Medicine
- Zhejiang University
- Hangzhou
| | - Wenna Zhang
- Laboratory of Fruit Quality Biology
- Zhejiang University
- Hangzhou
- China
| | - Xiaoyong Zhao
- Laboratory of Fruit Quality Biology
- Zhejiang University
- Hangzhou
- China
| | - Yanhong Chen
- Laboratory Animal Centre of Zhejiang University
- Hangzhou
- China
| | - Chongde Sun
- Laboratory of Fruit Quality Biology
- Zhejiang University
- Hangzhou
- China
- Agricultural Products Processing Technology Key Laboratory of Zhejiang Province
| | - Xian Li
- Laboratory of Fruit Quality Biology
- Zhejiang University
- Hangzhou
- China
- Agricultural Products Processing Technology Key Laboratory of Zhejiang Province
| | - Kunsong Chen
- Laboratory of Fruit Quality Biology
- Zhejiang University
- Hangzhou
- China
- Agricultural Products Processing Technology Key Laboratory of Zhejiang Province
| |
Collapse
|
37
|
Trombley S, Mustafa A, Schmitz M. Regulation of the seasonal leptin and leptin receptor expression profile during early sexual maturation and feed restriction in male Atlantic salmon, Salmo salar L., parr. Gen Comp Endocrinol 2014; 204:60-70. [PMID: 24818969 DOI: 10.1016/j.ygcen.2014.04.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/28/2014] [Accepted: 04/21/2014] [Indexed: 12/22/2022]
Abstract
In mammals, leptin acts as an adiposity signal and is a crucial link between nutritional status and the reproductive axis. So far the link between leptin and energy balance during sexual maturation in teleosts has been poorly investigated. In this study, seasonal gene expression changes in two leptin genes (lepa1 and lepa2) and the leptin receptor were investigated during early sexual maturation in male Atlantic salmon parr under fully fed (control) and feed restricted conditions from April through September. Both Atlantic salmon lepa1 and lepa2 in the liver and lepr in the brain were significantly down-regulated in non-maturing control males in early spring, coinciding with the start of the growth and fat accumulation. In maturing control males, hepatic leptin expression increased during mid-spermatogenesis and lepa1 and lepa2 mRNA levels were up-regulated by 7.7 and 49 times respectively during final maturation. For the first time in a fish species, a significant up-regulation of lepr expression was observed in the testis throughout mid to late spermatogenesis. Feed restriction decreased the incidence of sexual maturation by 53% and highly up-regulated both leptin genes in the liver and the leptin receptor in the pituitary. This study shows that hepatic lepa1 and lepa2 expression and lepr expression in the testis is affected by early sexual maturation in male Atlantic salmon parr. Fast growth and high fat stores are associated with low leptin levels while feed restriction has a stimulatory effect on hepatic leptin and leptin receptor gene expression in the pituitary, suggesting a role for leptin other than that as an adiposity signal.
Collapse
Affiliation(s)
- Susanne Trombley
- Department of Organismal Biology, Comparative Physiology, Evolutionary Biology Centre, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Arshi Mustafa
- Department of Organismal Biology, Comparative Physiology, Evolutionary Biology Centre, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Monika Schmitz
- Department of Organismal Biology, Comparative Physiology, Evolutionary Biology Centre, Uppsala University, SE-752 36 Uppsala, Sweden.
| |
Collapse
|
38
|
Liu S, Lin SJ, Li G, Kim E, Chen YT, Yang DR, Tan MHE, Yong EL, Chang C. Differential roles of PPARγ vs TR4 in prostate cancer and metabolic diseases. Endocr Relat Cancer 2014; 21:R279-300. [PMID: 24623743 DOI: 10.1530/erc-13-0529] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ, NR1C3) and testicular receptor 4 nuclear receptor (TR4, NR2C2) are two members of the nuclear receptor (NR) superfamily that can be activated by several similar ligands/activators including polyunsaturated fatty acid metabolites, such as 13-hydroxyoctadecadienoic acid and 15-hydroxyeicosatetraenoic acid, as well as some anti-diabetic drugs such as thiazolidinediones (TZDs). However, the consequences of the transactivation of these ligands/activators via these two NRs are different, with at least three distinct phenotypes. First, activation of PPARγ increases insulin sensitivity yet activation of TR4 decreases insulin sensitivity. Second, PPARγ attenuates atherosclerosis but TR4 might increase the risk of atherosclerosis. Third, PPARγ suppresses prostate cancer (PCa) development and TR4 suppresses prostate carcinogenesis yet promotes PCa metastasis. Importantly, the deregulation of either PPARγ or TR4 in PCa alone might then alter the other receptor's influences on PCa progression. Knocking out PPARγ altered the ability of TR4 to promote prostate carcinogenesis and knocking down TR4 also resulted in TZD treatment promoting PCa development, indicating that both PPARγ and TR4 might coordinate with each other to regulate PCa initiation, and the loss of either one of them might switch the other one from a tumor suppressor to a tumor promoter. These results indicate that further and detailed studies of both receptors at the same time in the same cells/organs may help us to better dissect their distinct physiological roles and develop better drug(s) with fewer side effects to battle PPARγ- and TR4-related diseases including tumor and cardiovascular diseases as well as metabolic disorders.
Collapse
Affiliation(s)
- Su Liu
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Shin-Jen Lin
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Gonghui Li
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Eungseok Kim
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Yei-Tsung Chen
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Dong-Rong Yang
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - M H Eileen Tan
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Eu Leong Yong
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Chawnshang Chang
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, TaiwanGeorge Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| |
Collapse
|
39
|
Saito E, Okada T, Abe Y, Odaka M, Kuromori Y, Iwata F, Hara M, Mugishima H, Kitamura Y. Relationship between estimated fatty acid desaturase activities and abdominal adiposity in Japanese children. Obes Res Clin Pract 2014; 8:e201-98. [DOI: 10.1016/j.orcp.2013.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 03/12/2013] [Accepted: 03/12/2013] [Indexed: 10/27/2022]
|
40
|
Lin SJ, Zhang Y, Liu NC, Yang DR, Li G, Chang C. Minireview: Pathophysiological roles of the TR4 nuclear receptor: lessons learned from mice lacking TR4. Mol Endocrinol 2014; 28:805-21. [PMID: 24702179 DOI: 10.1210/me.2013-1422] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Testicular nuclear receptor 4 (TR4), also known as NR2C2, belongs to the nuclear receptor superfamily and shares high homology with the testicular nuclear receptor 2. The natural ligands of TR4 remained unclear until the recent discoveries of several energy/lipid sensors including the polyunsaturated fatty acid metabolites, 13-hydroxyoctadecadienoic acid and 15-hydroxyeicosatetraenoic acid, and their synthetic ligands, thiazolidinediones, used for treatment of diabetes. TR4 is widely expressed throughout the body and particularly concentrated in the testis, prostate, cerebellum, and hippocampus. It has been shown to play important roles in cerebellar development, forebrain myelination, folliculogenesis, gluconeogenesis, lipogenesis, muscle development, bone development, and prostate cancer progression. Here we provide a comprehensive summary of TR4 signaling including its upstream ligands/activators/suppressors, transcriptional coactivators/repressors, downstream targets, and their in vivo functions with potential impacts on TR4-related diseases. Importantly, TR4 shares similar ligands/activators with another key nuclear receptor, peroxisome proliferator-activated receptor γ, which raised several interesting questions about how these 2 nuclear receptors may collaborate with or counteract each other's function in their related diseases. Clear dissection of such molecular mechanisms and their differential roles in various diseases may help researchers to design new potential drugs with better efficacy and fewer side effects to battle TR4 and peroxisome proliferator-activated receptor γ involved diseases.
Collapse
Affiliation(s)
- Shin-Jen Lin
- George Whipple Laboratory for Cancer Research (S.-J.L., Y.Z., N.-C.L., C.C.), Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14646; Department of Urology (D.-R.Y.), the Second Affiliated Hospital of Suzhou University, Suzhou, 215004 China; Chawnshang Chang Liver Cancer Center and Department of Urology (G.L.), Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016 China; and Sex Hormone Research Center (C.C.), China Medical University/Hospital, Taichung, 404 Taiwan
| | | | | | | | | | | |
Collapse
|
41
|
Suburu J, Gu Z, Chen H, Chen W, Zhang H, Chen YQ. Fatty acid metabolism: Implications for diet, genetic variation, and disease. FOOD BIOSCI 2013; 4:1-12. [PMID: 24511462 DOI: 10.1016/j.fbio.2013.07.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cultures across the globe, especially Western societies, are burdened by chronic diseases such as obesity, metabolic syndrome, cardiovascular disease, and cancer. Several factors, including diet, genetics, and sedentary lifestyle, are suspected culprits to the development and progression of these health maladies. Fatty acids are primary constituents of cellular physiology. Humans can acquire fatty acids by de novo synthesis from carbohydrate or protein sources or by dietary consumption. Importantly, regulation of their metabolism is critical to sustain balanced homeostasis, and perturbations of such can lead to the development of disease. Here, we review de novo and dietary fatty acid metabolism and highlight recent advances in our understanding of the relationship between dietary influences and genetic variation in fatty acid metabolism and their role in chronic diseases.
Collapse
Affiliation(s)
- Janel Suburu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhennan Gu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P.R. China ; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Haiqin Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P.R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P.R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P.R. China
| | - Yong Q Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P.R. China ; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
42
|
Freshwater Clam Extract Ameliorates Triglyceride and Cholesterol Metabolism through the Expression of Genes Involved in Hepatic Lipogenesis and Cholesterol Degradation in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:830684. [PMID: 23476706 PMCID: PMC3583048 DOI: 10.1155/2013/830684] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 12/26/2012] [Accepted: 01/06/2013] [Indexed: 12/20/2022]
Abstract
The freshwater clam (Corbicula spp.) is a popular edible bivalve and has been used as a folk remedy for liver disease in Asia. As a Chinese traditional medicine, it is said that freshwater clam ameliorates alcoholic intoxication and cholestasis. In this study, to estimate the practical benefit of freshwater clam extract (FCE), we compared the effects of FCE and soy protein isolate (SPI) on triglyceride and cholesterol metabolism in rats. FCE and SPI lowered serum cholesterol, and FCE tended to reduce serum triglycerides. FCE enhanced fecal sterol excretion and hepatic mRNA levels of CYP7A1 and ABCG5 more substantially than SPI; however, both diets reduced hepatic cholesterol. Both of the diets similarly suppressed liver lipids improved Δ9-desaturated fatty acid profile, and FCE was associated with a reduction in FAS and SCD1 mRNA levels. Hepatic transcriptome analysis revealed that inhibition of lipogenesis-related gene expression may contribute to downregulation of hepatic triglycerides by FCE. FCE would have better potential benefits for preventing metabolic disorders, through greater improvement of metabolism of triglycerides and cholesterol, likely through a mechanism similar to SPI.
Collapse
|
43
|
Trombley S, Schmitz M. Leptin in fish: possible role in sexual maturation in male Atlantic salmon. FISH PHYSIOLOGY AND BIOCHEMISTRY 2013; 39:103-106. [PMID: 23053613 DOI: 10.1007/s10695-012-9731-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 09/27/2012] [Indexed: 06/01/2023]
Abstract
Leptin plays an important role in energy homeostasis and reproductive function in mammals, while its function in fish is still poorly understood, especially its role in reproduction. In the present study, leptin gene expression and circulating leptin plasma levels were measured during sexual maturation in male Atlantic salmon parr. In maturing male salmon, higher hepatic leptin (lepa1) gene expression levels were observed during mid-spermatogenesis compared to immature fish. An upregulation of leptin during sexual maturation has also been observed in a few other teleost species. The physiological significance of elevated leptin levels during the maturational process in teleosts remains to be explored.
Collapse
Affiliation(s)
- S Trombley
- Department of Organismal Biology, Comparative Physiology, Evolutionary Biology Center, Uppsala University, Norbyvägen 18A, 752 36, Uppsala, Sweden
| | | |
Collapse
|
44
|
Hodson L, Fielding BA. Stearoyl-CoA desaturase: rogue or innocent bystander? Prog Lipid Res 2013; 52:15-42. [DOI: 10.1016/j.plipres.2012.08.002] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 08/27/2012] [Accepted: 08/27/2012] [Indexed: 02/07/2023]
|
45
|
Wang J, Zhao S, Song X, Pan H, Li W, Zhang Y, Gao S, Chen D. Low protein diet up-regulate intramuscular lipogenic gene expression and down-regulate lipolytic gene expression in growth–finishing pigs. Livest Sci 2012. [DOI: 10.1016/j.livsci.2012.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
46
|
Zhang XH, Huang B, Choi SK, Seo JS. Anti-obesity effect of resveratrol-amplified grape skin extracts on 3T3-L1 adipocytes differentiation. Nutr Res Pract 2012; 6:286-93. [PMID: 22977681 PMCID: PMC3439571 DOI: 10.4162/nrp.2012.6.4.286] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 07/01/2012] [Accepted: 07/09/2012] [Indexed: 11/09/2022] Open
Abstract
Resveratrol (3,4,5-trihydroxy-trans-stilbene), a phytoalexin found in grape skin, grape products, and peanuts as well as red wine, has been reported to have various biological and pharmacological properties. The purpose of this study was to investigate the anti-obesity effect of resveratrol-amplified grape skin extracts on adipocytes. The anti-obesity effects of grape skin extracts were investigated by measuring proliferation and differentiation in 3T3-L1 cells. The effect of grape skin ethanol extracts on cell proliferation was detected by the MTS assay. The morphological changes and degree of adipogenesis of preadipocyte 3T3-L1 cells were measured by Oil Red-O staining assay. Treatment with extracts of resveratrol-amplified grape skin decreased lipid accumulation and glycerol-3-phosphate dehydrogenase activity without affecting 3T3-L1 cell viability. Grape skin extract treatment resulted in significantly attenuated expression of key adipogenic transcription factors, including peroxisome proliferator-activated receptor, CCAAT/enhancer-binding proteins, and their target genes (FAS, aP2, SCD-1, and LPL). These results indicate that resveratrol-amplified grape skin extracts may be useful for preventing obesity by regulating lipid metabolism.
Collapse
Affiliation(s)
- Xian-Hua Zhang
- Department of Food and Nutrition, Yeungnam University, 214-1, Dae-dong, Gyeongsan-si, Gyeongbuk 712-749, Korea
| | | | | | | |
Collapse
|
47
|
Gillies PJ, Bhatia SK, Belcher LA, Hannon DB, Thompson JT, Vanden Heuvel JP. Regulation of inflammatory and lipid metabolism genes by eicosapentaenoic acid-rich oil. J Lipid Res 2012; 53:1679-89. [PMID: 22556214 DOI: 10.1194/jlr.m022657] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Omega-3-PUFAs, eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), are associated with prevention of various aspects of metabolic syndrome. In the present studies, the effects of oil rich in EPA on gene expression and activation of nuclear receptors was examined and compared with other ω3-PUFAs. The EPA-rich oil (EO) altered the expression of FA metabolism genes in THP-1 cells, including stearoyl CoA desaturase (SCD) and FA desaturase-1 and -2 (FASDS1 and -2). Other ω3-PUFAs resulted in a similar gene expression response for a subset of genes involved in lipid metabolism and inflammation. In reporter assays, EO activated human peroxisome proliferator-activated receptor α (PPARα) and PPARβ/γ with minimal effects on PPARγ, liver X receptor, retinoid X receptor, farnesoid X receptor, and retinoid acid receptor γ (RARγ); these effects were similar to that observed for purified EPA. When serum from a 6 week clinical intervention with dietary supplements containing olive oil (control), DHA, or two levels of EPA were applied to THP-1 cells, the expression of SCD and FADS2 decreased in the cells treated with serum from the ω3-PUFA-supplemented individuals. Taken together, these studies indicate regulation of gene expression by EO that is consistent with treating aspects of dyslipidemia and inflammation.
Collapse
Affiliation(s)
- Peter J Gillies
- Central Research and Development, DuPont Experimental Station, E328/140B, Wilmington, DE 19880, USA
| | | | | | | | | | | |
Collapse
|
48
|
Duarte FO, Sene-Fiorese M, Cheik NC, Maria ASLS, de Aquino AE, Oishi JC, Rossi EA, Garcia de Oliveira Duarte AC, Dâmaso AR. Food restriction and refeeding induces changes in lipid pathways and fat deposition in the adipose and hepatic tissues in rats with diet-induced obesity. Exp Physiol 2012; 97:882-94. [PMID: 22467759 DOI: 10.1113/expphysiol.2011.064121] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The aim of this study was to determine the effects of successive cycles of a moderately restrictive diet and refeeding with a high-fat diet on the metabolism of the adipose and hepatic tissues of obese rats. Rats were assigned to the following groups: a chow diet; a high-fat diet; a moderate caloric restriction; or a moderate caloric restriction plus refeeding. Some animals in each group were given [1-(14)C]triolein intragastrically, while others received an intraperitoneal injection of 3 mCi (3)H(2)O. All animals were killed by decapitation. The retroperitoneal, visceral epididymal and omental white adipose tissues, brown adipose tissue, liver and blood were immediately removed. The lipid uptake from the diet, in vivo rate of lipogenesis, percentage of fat, lipid profile and leptin concentration were analysed. The high-fat diet promoted an increase in fatty liver (P ≤ 0.05), adiposity mass (P ≤ 0.05) and the plasma concentration of leptin (P ≤ 0.05) and a decreased lipid uptake in white adipose tissue depots (P ≤ 0.05) in relation to the chow diet. The moderate caloric restriction did not reverse the changes promoted by the high-fat diet but induced a small decrease in adiposity, which was reversed after refeeding, and the animals maintained a dyslipidaemic profile and high fat deposition in the liver. We can conclude that the high-fat diet and subsequent moderate caloric restriction plus refeeding increased the risks of developing visceral obesity, dyslipidaemia and non-alcoholic fatty liver disease, which suggests that this type of experimental protocol can be used to study mechanisms related to the metabolic syndrome.
Collapse
Affiliation(s)
- Fernanda Oliveira Duarte
- Departamento de Educação Física e Motricidade-DEFMH, Universidade Federal de São Carlos-UFSCar Rodovia Washington Luís, Km 235-SP-310, São Carlos-São Paulo-Brasil, CEP: 13565-905.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Vatier C, Gautier JF, Vigouroux C. Therapeutic use of recombinant methionyl human leptin. Biochimie 2012; 94:2116-25. [PMID: 22464954 DOI: 10.1016/j.biochi.2012.03.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 03/13/2012] [Indexed: 01/11/2023]
Abstract
Recombinant methionyl human leptin (r-metHuLeptin) was first used as a replacement therapy in patients bearing inactivating mutations in the leptin gene. In this indication, it was shown since 1999 to be very efficient in inducing a dramatic weight loss in rare children and adults with severe obesity due to the lack of leptin. These first clinical trials clearly showed that r-metHuLeptin acted centrally to reduce food intake, inducing loss of fat mass, and to correct metabolic alterations, immune and neuroendocrine defects. A few years later, r-metHuLeptin was also shown to reverse the metabolic complications associated with lipodystrophic syndromes, due to primary defects in fat storage, which induce leptin deficiency. The beneficial effects, which could be mediated by central and/or peripheral mechanisms, are thought to mainly involve the lowering effects of leptin on ectopic lipid storage, in particular in liver and muscles, reducing insulin resistance. Interestingly, r-metHuLeptin therapy also reversed the hypothalamic-pituitary-gonadal axis dysfunctions associated with hypothalamic amenorrhea. However, if r-metHuLeptin treatment has been shown to be dramatically efficient in leptin-deficient states, its very limited effect in inducing weight loss in common obese patients revealed that, in patients with adequate leptin secretion, mechanisms of leptin resistance and leptin tolerance prevent r-metHuLeptin from inducing any additional effects. This review will present the current data about the effects of r-metHuLeptin therapy in humans, and discuss the recent perspectives of this therapy in new indications.
Collapse
Affiliation(s)
- Camille Vatier
- INSERM, UMR_S938, Centre de Recherches Saint-Antoine, Paris F-75012, France.
| | | | | |
Collapse
|
50
|
Côté I, Yasari S, Pighon A, Barsalani R, Rabasa-Lhoret R, Prud'homme D, Lavoie JM. Liver fat accumulation may be dissociated from adiposity gain in ovariectomized rats. Climacteric 2012; 15:594-601. [PMID: 22268446 DOI: 10.3109/13697137.2011.637650] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The purpose of the present study was to establish a model of rats prone and resistant to intra-abdominal fat accumulation in response to ovariectomy (Ovx-P and Ovx-R) and to determine its relationship with molecular biomarkers. DESIGN Two experiments were conducted in which female rats were either sham-operated (Sham) or ovariectomized (Ovx). In the first experiment, ovariectomized rats were stratified into three tertiles based on intra-abdominal adipose tissue mass. To strengthen the Ovx-P/Ovx-R model, we conducted a second experiment in which the numbers of rats in each group were extended and in which different molecular markers were measured. At the end of a 6-8-week period, ovariectomized rats that displayed the lower abdominal fat accumulation (lower tertile) were labelled as Ovx-R and those in the upper tertile as Ovx-P. RESULTS Ovx-R rats displayed similar abdominal fat gain to Sham rats whereas Ovx-P rats depicted abdominal fat mass twice as high as that of Sham and Ovx-R rats. Despite the difference in abdominal adiposity, liver fat content was ~50% higher (p < 0.01) in both Ovx-R and Ovx-P rats compared to Sham rats. In addition, both Ovx-R and Ovx-P rats depicted higher HOMA-IR scores (p < 0.05) and lower (p < 0.01) hepatic gene expression of leptin receptor-b and -e, microsomal transfer protein (MTP), and diacylglycerol acyltransferase-2 (DGAT-2) compared to Sham rats. CONCLUSION The present findings indicate that estrogen withdrawal-induced hepatic steatosis and associated insulin resistance may be dissociated from abdominal fat accumulation and suggest that a decrease in leptin action through a down-regulation of leptin receptors and a decrease in very low density lipoprotein production through a down-regulation of MTP and DGAT-2 may be factors responsible for this observation in the absence of peripheral fat gain.
Collapse
Affiliation(s)
- I Côté
- Department of Kinesiology, Université de Montréal, Montréal, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|