1
|
Guo YT, Huang JB, Xue JC, Qin SJ, Cai D, Wu QZ, Chu C, Tang CL, Xie YQ, Lin LZ, Dong GH, Zeng XW. Intrauterine exposure to long-chain perfluorocarboxylic acids (PFCAs) were associated with reduced primary bile acids in three-year-old children: Findings from a prospective birth cohort study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 360:124583. [PMID: 39038776 DOI: 10.1016/j.envpol.2024.124583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Bile acids (BAs) play a crucial role in lipid metabolism of children. However, the association between per- and polyfluoroalkyl substance (PFAS) exposure and BAs in children is scarce. To address this need, we selected 252 children from the Maoming Birth Cohort and measured 32 PFAS, encompassing short- and long-chain perfluorocarboxylic acids (PFCAs) and perfluorosulfonic acids (PFSAs) in the cord blood. Additionally, we analyzed nine primary and eight secondary BAs in the serum of three-year-old children. Generalized linear models with FDR-adjusted and Bayesian kernel machine regression (BKMR) were used to explore the associations of individual and mixture effects of PFAS and BAs. We found negative associations between cord blood long-chain PFCAs exposure and serum primary BAs in three-year-old children. For example, one ln-unit (ng/mL) increase of perfluoro-n-tridecanoic acid (PFTrDA), perfluoro-n-undecanoic acid (PFUnDA) and perfluoro-n-decanoic acid (PFDA) were associated with decreased taurochenodeoxycholic acid, with estimated percentage change of -24.28% [95% confidence interval (CI): -36.75%, -9.35%], -25.84% (95% CI: -39.67%, -8.83%), and -22.97% (95% CI: -34.45%, -9.47%) respectively. Notably, the observed associations were more pronounced in children with lower vegetable intake. Additionally, the BKMR model also demonstrated a monotonical decline in primary BAs as the PFAS mixture increased. We provided the first evidence of the association between intrauterine exposure to PFAS and its mixture with BAs in children. Further large-sample-size studies are needed to verify this finding.
Collapse
Affiliation(s)
- Yu-Ting Guo
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jin-Bo Huang
- Maoming Maternal and Child Health Hospital, Maoming, 525000, China
| | - Jing-Chuan Xue
- Guangdong Basic Research Center of Excellence for Ecological Security and Green Development, Key Laboratory for City Cluster Environmental Safety and Green Development of the Ministry of Education, School of Ecology, Environment and Resources, Guangdong University of Technology, Guangzhou, 510006, China
| | - Shuang-Jian Qin
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Dan Cai
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Environmental Protection, Guangzhou, 510655, China
| | - Qi-Zhen Wu
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chu Chu
- Guangdong Cardiovascular Institute, Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Cui-Lan Tang
- Maoming Maternal and Child Health Hospital, Maoming, 525000, China
| | - Yan-Qi Xie
- Maoming Maternal and Child Health Hospital, Maoming, 525000, China
| | - Li-Zi Lin
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Guang-Hui Dong
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiao-Wen Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
2
|
Khan M, Zhao X, Ni X, Ali S, Danzeng B, Yang H, Mushtaq M, Liang J, Xue B, Quan G. Impact of Varying Dietary Calcium Contents on the Gut Metabolomics of Yunnan Semi-Fine Wool Sheep ( Ovis aries). Metabolites 2024; 14:381. [PMID: 39057704 PMCID: PMC11278647 DOI: 10.3390/metabo14070381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Yunnan semi-fine wool (YSFW) is a recently developed dual-purpose (meat and wool) sheep breed mainly found in Yunnan Province, China. Moreover, dietary calcium is essential for animal health and productivity. The current experiment aimed to investigate the impact of dietary calcium on sheep gut metabolite profile. For this, thirty YSFW rams (male, age = 10 months, and body weight = 40.37 ± 0.49 kg) were randomized into three groups (n = 10 rams/group), followed by a completely randomized design, and the groups were allotted to one of three dietary calcium levels (Q_1 = 0.50%, Q_3 = 0.73%, and Q_5 = 0.98% on a dry basis). The rams were fed ad libitum by feeding twice a day (at 08:00 and 17:00 h/day) throughout the experimental period (44 day). On the 21st day of the experiment, fecal samples were collected from 27 rams (9/group) and untargeted metabolite profiling was performed by using ultra-performance liquid chromatography. The PCA plot showed that the Q_5 group metabolites were clustered more tightly than for Q_1 and Q_3, respectively. The tightly clustering molecules were mainly alkaloids and their derivatives, benzenoids, lignans and related compounds, lipids, nucleotides, organic acids, and nitrogenous-based derivatives. According to the Kyoto Encyclopedia of Genes and Genomes pathway analysis, these molecules potentially contribute to metabolic pathways, biosynthesis of secondary metabolites, proteinaceous compounds, and the metabolism of the protein derivatives, particularly amino acids. The PLS-DA plots revealed a significant difference between the Q_1, Q_3, and Q_5 groups, suggesting that Q_5 had a clear separation across the groups. Based on the metabolomic analysis, feeding different levels of dietary calcium significantly changed the metabolomic profile of YSFW rams, which primarily entails metabolic pathways such as energy, protein, and lipid metabolism.
Collapse
Affiliation(s)
- Muhammad Khan
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming 650224, China; (M.K.); (X.Z.); (X.N.); (B.D.); (H.Y.); (M.M.); (J.L.)
- Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Jindian, Panlong District, Kunming 650224, China
| | - Xiaoqi Zhao
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming 650224, China; (M.K.); (X.Z.); (X.N.); (B.D.); (H.Y.); (M.M.); (J.L.)
- Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Jindian, Panlong District, Kunming 650224, China
| | - Xiaojun Ni
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming 650224, China; (M.K.); (X.Z.); (X.N.); (B.D.); (H.Y.); (M.M.); (J.L.)
- Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Jindian, Panlong District, Kunming 650224, China
| | - Sikandar Ali
- Zhejiang Vegamax Biotechnology Co., Ltd., Huzhou 313300, China;
| | - Baiji Danzeng
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming 650224, China; (M.K.); (X.Z.); (X.N.); (B.D.); (H.Y.); (M.M.); (J.L.)
- Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Jindian, Panlong District, Kunming 650224, China
| | - Hongyuan Yang
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming 650224, China; (M.K.); (X.Z.); (X.N.); (B.D.); (H.Y.); (M.M.); (J.L.)
- Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Jindian, Panlong District, Kunming 650224, China
| | - Maida Mushtaq
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming 650224, China; (M.K.); (X.Z.); (X.N.); (B.D.); (H.Y.); (M.M.); (J.L.)
- Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Jindian, Panlong District, Kunming 650224, China
| | - Jiachong Liang
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming 650224, China; (M.K.); (X.Z.); (X.N.); (B.D.); (H.Y.); (M.M.); (J.L.)
- Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Jindian, Panlong District, Kunming 650224, China
| | - Bai Xue
- Animal Nutrition Institute, Sichuan Agricultural University, Wenjiang District, Chengdu 611137, China
| | - Guobo Quan
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming 650224, China; (M.K.); (X.Z.); (X.N.); (B.D.); (H.Y.); (M.M.); (J.L.)
- Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Jindian, Panlong District, Kunming 650224, China
| |
Collapse
|
3
|
Cheng Y, Liang S, Zhang S, Hui X. Thermogenic Fat as a New Obesity Management Tool: From Pharmaceutical Reagents to Cell Therapies. Biomedicines 2024; 12:1474. [PMID: 39062047 PMCID: PMC11275133 DOI: 10.3390/biomedicines12071474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is a complex medical condition caused by a positive imbalance between calorie intake and calorie consumption. Brown adipose tissue (BAT), along with the newly discovered "brown-like" adipocytes (called beige cells), functions as a promising therapeutic tool to ameliorate obesity and metabolic disorders by burning out extra nutrients in the form of heat. Many studies in animal models and humans have proved the feasibility of this concept. In this review, we aim to summarize the endeavors over the last decade to achieve a higher number/activity of these heat-generating adipocytes. In particular, pharmacological compounds, especially agonists to the β3 adrenergic receptor (β3-AR), are reviewed in terms of their feasibility and efficacy in elevating BAT function and improving metabolic parameters in human subjects. Alternatively, allograft transplantation of BAT and the transplantation of functional brown or beige adipocytes from mesenchymal stromal cells or human induced pluripotent stem cells (hiPSCs) make it possible to increase the number of these beneficial adipocytes in patients. However, practical and ethical issues still need to be considered before the therapy can eventually be applied in the clinical setting. This review provides insights and guidance on brown- and beige-cell-based strategies for the management of obesity and its associated metabolic comorbidities.
Collapse
Affiliation(s)
- Ying Cheng
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China;
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (S.L.); (S.Z.)
| | - Shiqing Liang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (S.L.); (S.Z.)
| | - Shuhan Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (S.L.); (S.Z.)
| | - Xiaoyan Hui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; (S.L.); (S.Z.)
| |
Collapse
|
4
|
Xu Y, Zhou C, Zong M, Zhu J, Guo X, Sun Z. High-protein high-konjac glucomannan diets changed glucose and lipid metabolism by modulating colonic microflora and bile acid profiles in healthy mouse models. Food Funct 2024; 15:4446-4461. [PMID: 38563504 DOI: 10.1039/d4fo00159a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
High protein and fiber diets are becoming increasingly popular for weight loss; however, the benefits or risks of high protein and fiber diets with a normal calorie level for healthy individuals still need to be elucidated. In this study, we explored the role and mechanisms of long-term high protein and/or konjac glucomannan diets on the metabolic health of healthy mouse models. We found that high konjac glucomannan contents improved the glucose tolerance of mice and both high protein and high konjac glucomannan contents improved the serum lipid profile but increased the TNF-α levels. In the liver, high dietary protein contents reduced the expression of the FASN gene related to fatty acid synthesis. Interactions of dietary protein and fiber were shown in the signaling pathways related to lipid and glucose metabolism of the liver and the inflammatory status of the colon, wherein the high protein and high konjac glucomannan diet downregulated the expression of the SREBF1 and FXR genes in the liver and downregulated the expression of TNF-α genes in the colon compared to the high protein diet. High konjac glucomannan contents reduced the colonic secondary bile acid levels including DCA and LCA; this was largely associated with the changed microbiota profile and also contributed to improved lipid and glucose homeostasis. In conclusion, high protein diets improved lipid homeostasis and were not a risk to metabolic health, while high fiber diets improved glucose and lipid homeostasis by modulating colonic microbiota and bile acid profiles, and a high protein diet supplemented with konjac glucomannan might improve hepatic lipid homeostasis and colonic inflammation in healthy mouse models through long-term intervention.
Collapse
Affiliation(s)
- Yetong Xu
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Chengyu Zhou
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Minyue Zong
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Junwei Zhu
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Xutong Guo
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Zhihong Sun
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| |
Collapse
|
5
|
Abiri B, Amini S, Hejazi M, Hosseinpanah F, Zarghi A, Abbaspour F, Valizadeh M. Tea's anti-obesity properties, cardiometabolic health-promoting potentials, bioactive compounds, and adverse effects: A review focusing on white and green teas. Food Sci Nutr 2023; 11:5818-5836. [PMID: 37823174 PMCID: PMC10563719 DOI: 10.1002/fsn3.3595] [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: 03/16/2023] [Revised: 07/02/2023] [Accepted: 07/22/2023] [Indexed: 10/13/2023] Open
Abstract
Tea is one of the most commonly consumed beverages in the world. Morocco, Japan, and China have consumed green tea for centuries. White tea, which is a variety of green teas, is very popular in China and is highly revered for its taste. Presently, both teas are consumed in other countries around the world, even as functional ingredients, and novel research is constantly being conducted in these areas. We provide an update on the health benefits of white and green teas in this review, based on recent research done to present. After a general introduction, we focused on tea's anti-obesity and human health-promoting potential, adverse effects, and new approaches to tea and its bioactive compounds. It has been found that the health benefits of tea are due to its bioactive components, mainly phenolic compounds. Of these, catechins are the most abundant. This beverage (or its extracts) has potential anti-inflammatory and antioxidant properties, which could contribute to body weight control and the improvement of several chronic diseases. However, some studies have mentioned the possibility of toxic effects; therefore, reducing tea consumption is a good idea, especially during the last trimester of pregnancy. Additionally, new evidence will provide insight into the possible effects of tea on the human gut microbiota, and even on the viruses responsible for SARS-CoV-2. A beverage such as this may favor beneficial gut microbes, which may have important implications due to the influence of gut microbiota on human health.
Collapse
Affiliation(s)
- Behnaz Abiri
- Obesity Research Center, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Shirin Amini
- Department of NutritionShoushtar Faculty of Medical SciencesShoushtarIran
| | - Mahdi Hejazi
- Department of Nutrition, School of Public HealthIran University of Medical SciencesTehranIran
| | - Farhad Hosseinpanah
- Obesity Research Center, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Afshin Zarghi
- Department of Pharmaceutical Chemistry, School of PharmacyShahid Beheshti University of Medical SciencesTehranIran
| | - Faeze Abbaspour
- Obesity Research Center, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Majid Valizadeh
- Obesity Research Center, Research Institute for Endocrine SciencesShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
6
|
Lai Z, Zhan X, Lin L, Zhang J, Qi W, Yang H, Mao S, Jin W. High-grain diet feeding alters ileal microbiota and disrupts bile acid metabolism in lactating dairy cows. J Anim Sci 2023; 101:skad278. [PMID: 37606090 PMCID: PMC10494876 DOI: 10.1093/jas/skad278] [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: 06/05/2023] [Accepted: 08/18/2023] [Indexed: 08/23/2023] Open
Abstract
Bile acids (BAs) play an important role in the regulation of lipid metabolic homeostasis, but little is known about their metabolism in dairy cows fed a high-grain (HG) diet. In the present study, we examined the bacterial community, BA profile, and the FXR/FGF19 signaling pathway in the ileum and liver to investigate the gut microbe-BA metabolism interactions response to HG diet and the changes in the subsequent enterohepatic circulation of dairy cows. The results showed that the ileal bacterial community was altered, with an increase of Paraclostridium, Anaerobutyricum, Shuttleworthia, and Stomatobaculum in the relative abundance in the HG group. Moreover, real-time polymerase chain reaction (PCR) showed that the abundance of total bacteria and bacterial bile-salt hydrolase (BSH) genes was increased in the ileal digesta in the HG group. Meanwhile, HG feeding also decreased the total BA content in the digesta of jejunum and ileum and in feces. HG feeding altered the BA profile in the ileal digesta by increasing unconjugated BAs and decreasing conjugated BAs. In addition, the intestinal FXR/FGF19 signaling pathway was activated. The expression of CYP7A1 (cholesterol 7α-hydroxylase) was depressed, which inhibited BAs synthesis in the liver of cows fed HG. Overall, HG feeding altered the ileal bacterial community and BA profile, and activated FXR/FGF19 signaling pathway, resulting in a decrease of BA level in the ileal digesta via the inhibition of hepatic BA synthesis. The findings provided novel insights into understanding the relationship between gut microbiota and the homeostasis of BAs in dairy cows fed a HG diet.
Collapse
Affiliation(s)
- Zheng Lai
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, the National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoxiu Zhan
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, the National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Limei Lin
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, the National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiyou Zhang
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, the National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weibiao Qi
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, the National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Huisheng Yang
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, the National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shengyong Mao
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, the National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wei Jin
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, the National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Anhui Academy of Agricultural Sciences, Hefei 230041, China
| |
Collapse
|
7
|
Effects of diets containing proteins from fish muscles or fish by-products on the circulating cholesterol concentration in rodents: a systematic review and meta-analysis. Br J Nutr 2022:1-22. [PMID: 36268726 DOI: 10.1017/s000711452200349x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A high circulating cholesterol concentration is considered an important risk factor for the development of CVD. Since lean fish intake and fish protein supplementation have been associated with lower cholesterol concentration in some but not all clinical studies, the main aim of this study was to investigate the effect of diets containing proteins from fish muscles and fish by-products on the serum/plasma total cholesterol (TC) concentration in rodents. A systematic literature search was performed using the databases PubMed, Web of Science and Embase, structured around the population (rodents), intervention (type of fish and fraction, protein dose and duration), comparator (casein) and the primary outcome (circulating TC). Articles were assessed for risk of bias using the SYRCLE's tool. A meta-analysis was conducted in Review Manager v. 5·4·1 (the Cochrane Collaboration) to determine the effectiveness of proteins from fish on the circulating TC concentration. Thirty-nine articles were included in the systematic review and meta-analysis, with data from 935 rodents. The risk of bias is unclear since few of the entries in the SYRCLE's tool were addressed. Consumption of proteins from fish resulted in a significantly lower circulating TC concentration when compared with control groups (mean difference -0·24 mmol/l, 95 % CI - 0·34, -0·15, P < 0·00001), with high statistical heterogeneity (I2 = 71 %). To conclude, proteins from fish muscles and by-products show promise as a functional dietary ingredient or supplement by preventing high cholesterol concentration in rodents, thus reducing one of the most important risk factors for developing CVD.
Collapse
|
8
|
Jung Y, Koo BK, Jang SY, Kim D, Lee H, Lee DH, Joo SK, Jung YJ, Park JH, Yoo T, Choi M, Lee MK, Kang SW, Chang MS, Kim W, Hwang GS. Association between circulating bile acid alterations and nonalcoholic steatohepatitis independent of obesity and diabetes mellitus. Liver Int 2021; 41:2892-2902. [PMID: 34358397 DOI: 10.1111/liv.15030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/22/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Bile acid (BA) dysregulation is related to not only metabolic diseases but also nonalcoholic fatty liver disease (NAFLD). We investigated whether circulating BA levels are altered according to the histological severity of NAFLD independent of metabolic derangements. METHODS Global metabolic profiling and targeted BA analysis using sera collected from biopsy-proven no-NAFLD (n = 67), nonalcoholic fatty liver (NAFL) (n = 99), and nonalcoholic steatohepatitis (NASH, n = 75) subjects were performed sequentially. Circulating metabolome analysis integrated with the hepatic transcriptome was performed to elucidate the mechanistic basis of altered circulating BA profiles after stratification by obesity (body mass index ≤ 25 kg/m2 ). Circulating BA alterations were also validated in an independent validation cohort (29 no-NAFLD, 70 NAFL and 37 NASH). RESULTS Global profiling analysis showed that BA was the metabolite significantly altered in NASH compared to NAFL. Targeted BA analysis demonstrated that glyco-/tauro-conjugated primary BAs were commonly increased in nonobese and obese NASH, while unconjugated primary BAs increased only in nonobese NASH. These characteristic primary BA level changes were maintained even after stratification according to diabetes status and were replicated in the independent validation cohort. Compared to nonobese NAFL patients, nonobese NASH patients exhibited upregulated hepatic expression of CYP8B1. CONCLUSIONS BA metabolism is dysregulated as the histological severity of NAFLD worsens, independent of obesity and diabetes status; dysregulation is more prominent in nonobese NAFLD patients. Metabolome-driven omics approach provides new insight into our understanding of altered BA metabolism associated with individual phenotypes of NAFLD.
Collapse
Affiliation(s)
- Youngae Jung
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea.,Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Bo Kyung Koo
- Division of Endocrinology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Seo Young Jang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea.,Department of Chemistry & Nanoscience, Ewha Womans University, Seoul, Republic of Korea
| | - Dain Kim
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea.,Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Heeyeon Lee
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea.,Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Dong Hyeon Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Sae Kyung Joo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Yong Jin Jung
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Jeong Hwan Park
- Department of Pathology, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Taekyeong Yoo
- Department of Biochemical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Murim Choi
- Department of Biochemical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Min Kyung Lee
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea.,Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Sang Won Kang
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Mee Soo Chang
- Department of Pathology, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Won Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea.,Department of Chemistry & Nanoscience, Ewha Womans University, Seoul, Republic of Korea
| | | |
Collapse
|
9
|
No effect of salmon fish protein on 2-h glucose in adults with increased risk of type 2 diabetes: a randomised controlled trial. Br J Nutr 2021; 126:1304-1313. [PMID: 33413727 DOI: 10.1017/s0007114521000040] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The association between fish consumption and decreased risk of CVD is well documented. However, studies on health effects of fish consumption suggest that other components than n-3 PUFA have beneficial cardiometabolic effects, including effects on glucose metabolism. The aim of the present study was to investigate effects of salmon fish protein on cardiometabolic risk markers in a double-blind, randomised controlled parallel trial. We hypothesised that daily intake of a salmon fish protein supplement for 8 weeks would improve glucose tolerance in persons with increased risk of type 2 diabetes mellitus (T2DM). Our primary outcome measure was serum glucose (s-glucose) 2 h after a standardised oral glucose tolerance test. In total, eighty-eight adults with elevated s-glucose levels were randomised to 7·5 g of salmon fish protein/d or placebo, and seventy-four participants were included in the analysis. We found no significant effect of salmon fish protein supplementation on our primary outcome or other markers related to glucose tolerance, serum lipids, weight or blood pressure compared with placebo. The present study does not support the hypothesis that daily intake of a salmon fish protein supplement for 8 weeks improves glucose tolerance in persons with increased risk of T2DM.
Collapse
|
10
|
Frazier K, Frith M, Harris D, Leone VA. Mediators of Host–Microbe Circadian Rhythms in Immunity and Metabolism. BIOLOGY 2020; 9:biology9120417. [PMID: 33255707 PMCID: PMC7761326 DOI: 10.3390/biology9120417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/20/2020] [Indexed: 12/27/2022]
Abstract
Simple Summary Circadian rhythms serve as the body’s internal metronome, driving responses to environmental cues over a 24-h period. Essential to nearly all life forms, the core circadian clock gene network drives physiological outputs associated with metabolic and immune responses. Modern-day disruptions to host circadian rhythms, such as shift work and jet lag, result in aberrant metabolic responses and development of complex diseases, including obesity and Type 2 Diabetes. These complex diseases are also impacted by interactions between gut microbes and the host immune system, driving a chronic low-grade inflammatory response. Gut microbes exhibit circadian dynamics that are closely tied to host circadian networks and disrupting microbial rhythmicity contributes to metabolic diseases. The underlying mediators that drive communication between host metabolism, the immune system, gut microbes, and circadian networks remain unknown, particularly in humans. Here, we explore the current state of knowledge regarding the transkingdom control of circadian networks and discuss gaps and challenges to overcome to push the field forward from the preclinical to clinical setting. Abstract Circadian rhythms are essential for nearly all life forms, mediated by a core molecular gene network that drives downstream molecular processes involved in immune function and metabolic regulation. These biological rhythms serve as the body’s metronome in response to the 24-h light:dark cycle and other timed stimuli. Disrupted circadian rhythms due to drastic lifestyle and environmental shifts appear to contribute to the pathogenesis of metabolic diseases, although the mechanisms remain elusive. Gut microbiota membership and function are also key mediators of metabolism and are highly sensitive to environmental perturbations. Recent evidence suggests rhythmicity of gut microbes is essential for host metabolic health. The key molecular mediators that transmit rhythmic signals between microbes and host metabolic networks remain unclear, but studies suggest the host immune system may serve as a conduit between these two systems, providing homeostatic signals to maintain overall metabolic health. Despite this knowledge, the precise mechanism and communication modalities that drive these rhythms remain unclear, especially in humans. Here, we review the current literature examining circadian dynamics of gut microbes, the immune system, and metabolism in the context of metabolic dysregulation and provide insights into gaps and challenges that remain.
Collapse
Affiliation(s)
- Katya Frazier
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (K.F.); (M.F.); (D.H.)
| | - Mary Frith
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (K.F.); (M.F.); (D.H.)
- Medical Scientist Training Program, University of Chicago, Chicago, IL 60637, USA
| | - Dylan Harris
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (K.F.); (M.F.); (D.H.)
| | - Vanessa A. Leone
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (K.F.); (M.F.); (D.H.)
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
- Correspondence: ; Tel.: +1-608-262-5551
| |
Collapse
|
11
|
Zagoskin P, Erlykina E. Bile Acids as a New Type of Steroid Hormones Regulating Nonspecific Energy Expenditure of the Body (Review). Sovrem Tekhnologii Med 2020; 12:114-127. [PMID: 34796012 PMCID: PMC8596256 DOI: 10.17691/stm2020.12.5.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
The review is devoted to the systematization, classification, and generalization of the results of modern scientific research on the role of bile acids as a new class of steroid hormones. The paper presents the evidence for bile acid participation in the regulation of the body energy metabolism, body weight control, as well as the pathogenesis of obesity, diabetes mellitus, insulin resistance, and cardiovascular diseases. Particular attention is paid to the role of bile acids in the control of nonspecific energy expenditure of the body. The applied aspects of using the novel data about the membrane and intracellular receptors responsible for the development of hormonal regulatory effects of bile acids are analyzed. According to the authors, the modern data on the role of bile acids in the regulation of body functions allow a deeper understanding of the pathogenesis of body weight disorders and associated cardiovascular diseases. The review demonstrates promising directions in the search for specific methods of prevention and correction of these pathological conditions.
Collapse
Affiliation(s)
- P.P. Zagoskin
- Associate Professor, Department of Biochemistry named after G.Ya. Gorodisskaya; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - E.I. Erlykina
- Professor, Head of the Department of Biochemistry named after G.Ya. Gorodisskaya Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| |
Collapse
|
12
|
Fjære E, Myrmel LS, Dybing K, Kuda O, Holbech Jensen BA, Rossmeisl M, Frøyland L, Kristiansen K, Madsen L. The Anti-Obesogenic Effect of Lean Fish Species is Influenced by the Fatty Acid Composition in Fish Fillets. Nutrients 2020; 12:E3038. [PMID: 33022997 PMCID: PMC7600456 DOI: 10.3390/nu12103038] [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: 09/14/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 11/17/2022] Open
Abstract
Fillets from marine fish species contain n-3 polyunsaturated fatty acids (PUFAs) in the form of phospholipids (PLs). To investigate the importance of PL-bound n-3 PUFAs in mediating the anti-obesogenic effect of lean seafood, we compared the anti-obesogenic properties of fillets from cod with fillets from pangasius, a fresh water fish with a very low content of PL-bound n-3 PUFAs. We prepared high-fat/high-protein diets using chicken, cod and pangasius as the protein sources, and fed male C57BL/6J mice these diets for 12 weeks. Mice fed the diet containing cod gained less adipose tissue mass and had smaller white adipocytes than mice fed the chicken-containing diet, whereas mice fed the pangasius-containing diet were in between mice fed the chicken-containing diet and mice fed the cod-containing diet. Of note, mice fed the pangasius-containing diet exhibited reduced glucose tolerance compared to mice fed the cod-containing diet. Although the sum of marine n-3 PUFAs comprised less than 2% of the total fatty acids in the cod-containing diet, this was sufficient to significantly increase the levels of eicosapentaenoic acid (EPA) and docosahexaenoic acids (DHA) in mouse tissues and enhance production of n-3 PUFA-derived lipid mediators as compared with mice fed pangasius or chicken.
Collapse
Affiliation(s)
- Even Fjære
- Institute of Marine Research, NO-5817 Bergen, Norway; (E.F.); (L.S.M.); (K.D.); (L.F.)
| | - Lene Secher Myrmel
- Institute of Marine Research, NO-5817 Bergen, Norway; (E.F.); (L.S.M.); (K.D.); (L.F.)
| | - Karianne Dybing
- Institute of Marine Research, NO-5817 Bergen, Norway; (E.F.); (L.S.M.); (K.D.); (L.F.)
| | - Ondrej Kuda
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, 14220 Prague 4, Czech Republic; (O.K.); (M.R.)
| | - Benjamin Anderschou Holbech Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark;
| | - Martin Rossmeisl
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, 14220 Prague 4, Czech Republic; (O.K.); (M.R.)
| | - Livar Frøyland
- Institute of Marine Research, NO-5817 Bergen, Norway; (E.F.); (L.S.M.); (K.D.); (L.F.)
| | - Karsten Kristiansen
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen, Denmark;
| | - Lise Madsen
- Institute of Marine Research, NO-5817 Bergen, Norway; (E.F.); (L.S.M.); (K.D.); (L.F.)
| |
Collapse
|
13
|
Increasing the Hindgut Carbohydrate/Protein Ratio by Cecal Infusion of Corn Starch or Casein Hydrolysate Drives Gut Microbiota-Related Bile Acid Metabolism To Stimulate Colonic Barrier Function. mSystems 2020; 5:5/3/e00176-20. [PMID: 32487741 PMCID: PMC8534727 DOI: 10.1128/msystems.00176-20] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Dietary high protein and low carbohydrate levels compromise colonic microbiota and bile acid metabolism, which underlies a detrimental gut environment. However, it remains unclear if the diet-induced changes in colonic health are due to a change in hindgut nutrient availability and what key intermediates link the microbe-epithelium dialogue. To specifically alter the hindgut nutrient substrate availability, here we used a cecally cannulated pig model to infuse corn starch and casein hydrolysate directly into the cecum to generate a stepwise change of carbohydrate/nitrogenous compound (C/N) ratio. Pigs were cecally infused daily with either saline (Control), corn starch (Starch), or casein hydrolysate (Casein) (n = 8 per group), respectively, for 19 days. After infusion, C/N ratios in colonic digesta were 16.33, 12.56, and 8.54 for the starch, control, and casein groups, respectively (P < 0.05). Relative to the control group, casein infusion showed greater abundance of the bacteria (Eubacterium) capable of bile acid 7α-dehydroxylation (baiJ), higher levels of expression of bacterial genes encoding the baiJ enzyme, and higher levels of secondary bile acid (deoxycholic acid [DCA] and lithocholic acid [LCA]), while the starch infusion showed the opposite effect. Correspondingly, casein infusion downregulated expression of genes encoding tight junction proteins (ZO-1 and OCLD) and upregulated expression of genes encoding epidermal growth factor receptor (EGFR). The ratio of C/N was linearly related with the concentrations of DCA and LCA and gene expression levels of ZO-1, occludin, and EGFR. Caco-2 cell experiments further showed that DCA and LCA downregulated expression of genes involved in barrier function (ZO-1 and OCLD) and upregulated the gene expression of EGFR and Src. Inhibition of EGFR and Src could abolish DCA- and LCA-induced downregulation of ZO-1, indicating that DCA and LCA impair gut barrier function via enhancing the EGFR-Src pathway. These results suggest that the ratio of C/N in the large intestine is an important determinant of microbial metabolism and gut barrier function in the colon. The findings provide evidence that microbe-related secondary bile acid metabolism may mediate the interplay between microbes and gut barrier function. IMPORTANCE High-fiber or high-protein diets could alter gut microbiota and health in the large intestine, but factors involved in the effects remain unclear. The present study for the first time demonstrates that the starch- and casein-induced C/N ratio in the hindgut is an important factor. Using the cannulated pig model, we found that the distinct C/N ratio induced by cecal infusion of corn starch or casein hydrolysate was linearly correlated with microbial metabolites (secondary bile acids) and tight junction proteins (ZO-1 and OCLD). Cell culture study further demonstrates that the gut microbial metabolites (DCA and LCA) could impair the intestinal barrier function via the EGFR-Src pathway. These suggest that DCA and LCA were key metabolites mediating microbe-epithelium dialogue when the hindgut C/N ratios were altered by cecal infusion of corn starch or casein hydrolysate. These findings provide new insight into the impact of C/N ratio in the large intestine on colonic health and provide a new framework for therapeutic strategy in gut health through targeted manipulation of hindgut microbiota by increasing the carbohydrate level in the large intestine.
Collapse
|
14
|
Wang S, Pan MH, Hung WL, Tung YC, Ho CT. From white to beige adipocytes: therapeutic potential of dietary molecules against obesity and their molecular mechanisms. Food Funct 2019; 10:1263-1279. [PMID: 30735224 DOI: 10.1039/c8fo02154f] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The global incidence of obesity and its complications continue to rise along with a demand for novel therapeutic approaches. In addition to classic brown adipose tissue (BAT), the formation of brown-like adipocytes called beige adipocytes, within white adipose tissue (WAT), has attracted much attention as a therapeutic target due to its inducible features when stimulated, resulting in the dissipation of extra energy as heat. There are various dietary agents that are able to modulate the beige-development process by interacting with critical molecular signaling cascades, leading to the enhancement of thermogenesis. Although challenges still remain regarding the origin of the beige adipocytes, the crosstalk with activation of BAT and induction of the beiging of white fat may provide attractive potential strategies for management of obesity.
Collapse
Affiliation(s)
- Siyu Wang
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA.
| | | | | | | | | |
Collapse
|
15
|
Dale HF, Madsen L, Lied GA. Fish-derived proteins and their potential to improve human health. Nutr Rev 2019; 77:572-583. [PMID: 31124569 DOI: 10.1093/nutrit/nuz016] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Emerging evidence from studies evaluating the effect of lean fish consumption in humans suggests that proteins from fish have several beneficial metabolic effects. Rest, or waste, material from the fishing industry contains high-quality proteins, and utilization of this material offers novel possibilities for the development of protein-containing products that might be beneficial for human consumption. Fish-derived peptides containing bioactive amino acid sequences suggested to beneficially influence pathways involved in body composition, hypertension, lipid profile, and regulation of glucose metabolism are of particular interest, although the results of published studies are conflicting. This review aims to summarize current knowledge from animal studies and clinical interventions in humans evaluating the effects of lean fish, fish proteins, and fish-derived peptides on outcomes related to metabolic health. Fish proteins have a high content of taurine, and animal trials suggest that taurine mediates some of the beneficial effects observed thus far, although the mechanisms by which fish peptides exert their action are not yet elucidated. At this time, the literature is inconsistent, and there is insufficient mechanistic evidence to support a beneficial effect of fish-derived peptides on metabolic health.
Collapse
Affiliation(s)
- Hanna Fjeldheim Dale
- Department of Clinical Medicine, Centre for Nutrition; the Section of Gastroenterology, Department of Medicine
- National Centre of Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
| | - Lise Madsen
- Department of Clinical Medicine, Centre for Nutrition; the Section of Gastroenterology, Department of Medicine
- National Centre of Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
| | - Gülen Arslan Lied
- Institute of Marine Research, Bergen, Norway; and the Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Vildmyren I, Halstensen A, Oterhals Å, Gudbrandsen OA. Cod protein powder lowered serum nonesterified fatty acids and increased total bile acid concentrations in healthy, lean, physically active adults: a randomized double-blind study. Food Nutr Res 2019; 63:3437. [PMID: 30890903 PMCID: PMC6416186 DOI: 10.29219/fnr.v63.3437] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/11/2022] Open
Abstract
Background Fish fillet consumption is associated with beneficial health effects; however, little is known about whether consuming other parts of the fish such as head, backbone, skin, cut-offs, and entrails (collectively known as residuals) will provide comparable effects. Objective The aim of the study was to investigate if daily supplementation with cod residual protein powder would impact lipid metabolism in healthy adults. Methods Forty healthy, lean, physically active participants (18 women, 22 men) with normal body mass index consumed 8.1 g of proteins daily from cod residual protein powder (Cod-RP) or placebo (control) for 8 weeks. Results Cod residual protein powder supplementation lowered fasting serum nonesterified fatty acids and increased serum total bile acid concentrations significantly when compared with control supplementation. Fasting serum low-density lipoprotein cholesterol and apolipoprotein (Apo) B concentrations, as well as the total cholesterol:high-density lipoprotein (HDL) cholesterol and ApoB:ApoA1 ratios, were significantly decreased within the Cod-RP group, but these changes were not different from the control group. Fasting serum concentrations of triacylglycerol, total cholesterol, HDL cholesterol, and ApoA1 were not changed within or between groups. Conclusion Eight weeks of daily supplementation with 8.1 g Cod-RP seems to be sufficient to affect lipid metabolism in healthy, lean, physically active adults.
Collapse
Affiliation(s)
- Iselin Vildmyren
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, Bergen, Norway.,K. Halstensen AS, Bekkjarvik, Norway
| | - Alfred Halstensen
- K. Halstensen AS, Bekkjarvik, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Oddrun A Gudbrandsen
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
17
|
Sun L, Xu H, Ye J, Gaikwad NW. Comparative effect of black, green, oolong, and white tea intake on weight gain and bile acid metabolism. Nutrition 2019; 65:208-215. [PMID: 31031064 DOI: 10.1016/j.nut.2019.02.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 01/23/2019] [Accepted: 02/14/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The beneficial effects of tea on health, including obesity, are well known. However, the comparative effects of black, green, white, and oolong teas, which are prepared from the same fresh leaves, on weight gain and the potential mechanisms involved are not yet fully understood. Bile acids (BAs) are shown to be powerful regulators of metabolism; however, to our knowledge, no studies have investigated the effect of tea on BA metabolism. The aim of this study was to investigate the modulatory effects that green, black, white, and oolong teas that were prepared from the same raw tea leaves have on the plasma BA profile. METHODS Female rats were dosed with the aforementioned tea types as their sole source of drinking fluid for 28 d. We then investigated their weight and effect on BA metabolic profile using advanced ultra-performance liquid chromatography-tandem mass spectrometer (UPLC-MS/MS)-based metabolomics. RESULTS The UPLC-MS/MS analysis of the plasma show that the levels of murocholic acid, glycochenodeoxycholic acid, glycocholic acid, glycodeoxycholic acid, taurochenodeoxycholic acid, tauroursodeoxycholic acid, taurodeoxycholic acid, tauromuricholic acid, and taurocholic acid were increased; whereas levels of taurolithocholic acid and isolithocholic acid were decreased after drinking green, oolong, and white tea types compared with control. Surprisingly, oolong tea significantly influenced reduction in relative weight compared with control, black, and green tea; whereas black, green, and white teas had no effects on weight compared with control. CONCLUSIONS Green, black, oolong, and white teas altered the BA metabolism. This change in BA metabolism could be associated with the health benefit effects of tea. Oolong tea was most effective in reducing weight.
Collapse
Affiliation(s)
- Lili Sun
- Tea Research Institute, Zhejiang University, Hangzhou, China
| | - Hairong Xu
- Tea Research Institute, Zhejiang University, Hangzhou, China
| | - Jianhui Ye
- Tea Research Institute, Zhejiang University, Hangzhou, China
| | | |
Collapse
|
18
|
Small molecules for fat combustion: targeting obesity. Acta Pharm Sin B 2019; 9:220-236. [PMID: 30976490 PMCID: PMC6438825 DOI: 10.1016/j.apsb.2018.09.007] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/01/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022] Open
Abstract
Obesity is increasing in an alarming rate worldwide, which causes higher risks of some diseases, such as type 2 diabetes, cardiovascular diseases, and cancer. Current therapeutic approaches, either pancreatic lipase inhibitors or appetite suppressors, are generally of limited effectiveness. Brown adipose tissue (BAT) and beige cells dissipate fatty acids as heat to maintain body temperature, termed non-shivering thermogenesis; the activity and mass of BAT and beige cells are negatively correlated with overweight and obesity. The existence of BAT and beige cells in human adults provides an effective weight reduction therapy, a process likely to be amenable to pharmacological intervention. Herein, we combed through the physiology of thermogenesis and the role of BAT and beige cells in combating with obesity. We summarized the thermogenic regulators identified in the past decades, targeting G protein-coupled receptors, transient receptor potential channels, nuclear receptors and miscellaneous pathways. Advances in clinical trials were also presented. The main purpose of this review is to provide a comprehensive and up-to-date knowledge from the biological importance of thermogenesis in energy homeostasis to the representative thermogenic regulators for treating obesity. Thermogenic regulators might have a large potential for further investigations to be developed as lead compounds in fighting obesity.
Collapse
Key Words
- AKT, protein kinase B
- ALDH9, aldehyde dehydrogenase 9
- AMPK, AMP-activated protein kinase
- ATP, adenosine triphosphate
- BA, bile acids
- BAT, brown adipose tissue
- BMP8b, bone morphogenetic protein 8b
- Beige cells
- Brown adipose tissue
- C/EBPα, CCAAT/enhancer binding protein α
- CLA, cis-12 conjugated linoleic acid
- CRABP-II, cellular RA binding protein type II
- CRE, cAMP response element
- Cidea, cell death-inducing DNA fragmentation factor α-like effector A
- Dio2, iodothyronine deiodinase type 2
- ERE, estrogen response element
- ERs, estrogen receptors
- FAS, fatty acid synthase
- FGF21, fibroblast growth factor 21
- GPCRs, G protein-coupled receptors
- HFD, high fat diet
- LXR, liver X receptors
- MAPK, mitogen-activated protein kinase
- OXPHOS, oxidative phosphorylation
- Obesity
- PDEs, phosphodiesterases
- PET-CT, positron emission tomography combined with computed tomography
- PGC-1α, peroxisome proliferator-activated receptor γ coactivator 1-α
- PKA, protein kinase A
- PPARs, peroxisome proliferator-activated receptors
- PPREs, peroxisome proliferator response elements
- PRDM16, PR domain containing 16
- PTP1B, protein-tyrosine phosphatase 1B
- PXR, pregnane X receptor
- RA, retinoic acid
- RAR, RA receptor
- RARE, RA response element
- RMR, resting metabolic rate
- RXR, retinoid X receptor
- SIRT1, silent mating type information regulation 2 homolog 1
- SNS, sympathetic nervous system
- TFAM, mitochondrial transcription factor A
- TMEM26, transmembrane protein 26
- TRPs, transient receptor potential cation channels
- Thermogenesis
- UCP1, uncoupling protein 1
- Uncoupling protein 1
- VDR, vitamin D receptor
- VDRE, VDR response elements
- WAT, white adipose tissue
- cAMP, cyclic adenosine monophosphate
- cGMP, cyclic guanosine monophosphate
- β3-AR, β3-adrenergic receptor
Collapse
|
19
|
Abstract
We provide an overview of studies on seafood intake in relation to obesity, insulin resistance and type 2 diabetes. Overweight and obesity development is for most individuals the result of years of positive energy balance. Evidence from intervention trials and animal studies suggests that frequent intake of lean seafood, as compared with intake of terrestrial meats, reduces energy intake by 4–9 %, sufficient to prevent a positive energy balance and obesity. At equal energy intake, lean seafood reduces fasting and postprandial risk markers of insulin resistance, and improves insulin sensitivity in insulin-resistant adults. Energy restriction combined with intake of lean and fatty seafood seems to increase weight loss. Marine n-3 PUFA are probably of importance through n-3 PUFA-derived lipid mediators such as endocannabinoids and oxylipins, but other constituents of seafood such as the fish protein per se, trace elements or vitamins also seem to play a largely neglected role. A high intake of fatty seafood increases circulating levels of the insulin-sensitising hormone adiponectin. As compared with a high meat intake, high intake of seafood has been reported to reduce plasma levels of the hepatic acute-phase protein C-reactive protein level in some, but not all studies. More studies are needed to confirm the dietary effects on energy intake, obesity and insulin resistance. Future studies should be designed to elucidate the potential contribution of trace elements, vitamins and undesirables present in seafood, and we argue that stratification into responders and non-responders in randomised controlled trials may improve the understanding of health effects from intake of seafood.
Collapse
|
20
|
Chicken Protein Hydrolysates Have Anti-Inflammatory Effects on High-Fat Diet Induced Obesity in Mice. MEDICINES 2018; 6:medicines6010005. [PMID: 30597839 PMCID: PMC6473722 DOI: 10.3390/medicines6010005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 11/30/2022]
Abstract
Background: Studies have shown that dietary source of protein and peptides can affect energy metabolism and influence obesity-associated diseases. This study aimed to investigate the impact of different chicken protein hydrolysates (CPHs) generated from chicken rest raw materials in a mouse obesity model. Methods: Male C57BL/6 mice were fed a high-fat, high-sucrose diet with casein or CPHs generated using Papain + Bromelain, Alcalase, Corolase PP, or Protamex for 12 weeks (n = 12). Body weight, feed intake, and intraperitoneal glucose tolerance was determined, and plasma and liver and adipose tissues were collected at sacrifice. Results: The average feed intake and body weight did not differ between the groups and white adipose tissue depots were unchanged, except for a reduction in the subcutaneous depot in mice fed the Protamex CPH diet. Moreover, the CPH diets did not prevent increased fasting glucose and insulin levels. Interestingly, the hepatic mitochondrial fatty acid β-oxidation was increased in mice fed Alcalase and Corolase PP CPHs. All CPH diets reduced plasma interleukine (IL)-1β, interferon-γ, tumor necrosis factor α, and monocyte chemotactic protein 1 compared to control, indicating anti-inflammatory effects. In addition, Corolase PP and Protamex CPHs significantly reduced plasma levels of IL-1α, IL-2, IL-6, IL-10, and granulocyte macrophage colony-stimulating factor. Conclusions: CPH diets were not able to counteract obesity and glucose intolerance in a mouse obesity model, but strongly reduced inflammatory parameters associated with obesity. Alcalase and Corolase PP CPHs also stimulated mitochondrial fatty acid β-oxidation. The possibility that hydrolysates from chicken rest raw materials could alleviate obesity-associated metabolic disease should be investigated further.
Collapse
|
21
|
Mao L, Lei J, Schoemaker MH, Ma B, Zhong Y, Lambers TT, Van Tol EAF, Zhou Y, Nie T, Wu D. Long-chain polyunsaturated fatty acids and extensively hydrolyzed casein-induced browning in a Ucp-1 reporter mouse model of obesity. Food Funct 2018; 9:2362-2373. [PMID: 29589625 DOI: 10.1039/c7fo01835e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Browning in adipose tissues, which can be affected by diet, may mitigate the detrimental effects of adiposity and improve longer-term metabolic health. Here, browning-inducing effects of long-chain polyunsaturated fatty acids, e.g., arachidonic acid (ARA)/docosahexaenoic acid (DHA) and extensively hydrolyzed casein (eHC) were investigated in uncoupling protein 1 (Ucp-1) reporter mice. To address the overall functionality, their potential role in supporting a healthy metabolic profile under obesogenic dietary challenges later in life was evaluated. At weaning Ucp1+/LUC reporter mice were fed a control low fat diet (LFD) with or without ARA + DHA, eHC or eHC + ARA + DHA for 8 weeks until week 12 after which interventions continued for another 12 weeks under a high-fat diet (HFD) challenge. Serology (metabolic responses and inflammation) and in vivo and ex vivo luciferase activity were determined; in the meantime browning-related proteins UCP-1 and the genes peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), PR domain containing 16 (PRDM16) and Ucp-1 were examined. ARA + DHA, eHC or their combination reduced body weight gain and adipose tissue weight compared to the HFD mice. The interventions induced Ucp-1 expression in adipose tissues prior to and during the HFD exposure. Ucp-1 induction was accompanied by higher PGC1a and PRDM16 expression. Glucose tolerance and insulin sensitivity were improved coinciding with lower serum cholesterol, triglycerides, free fatty acids, insulin, leptin, resistin, fibroblast growth factor 21, alanine aminotransferase, aspartate aminotransferase and higher adiponectin than the HFD group. HFD-associated increased systemic (IL-1β and TNF-α) and adipose tissue inflammation (F4/80, IL-1β, TNF-α, IL-6) was reduced. Studies in a Ucp-1 reporter mouse model revealed that early intervention with ARA/DHA and eHC improves metabolic flexibility and attenuates obesity during HFD challenge later in life. Increased browning is suggested as, at least, part of the underlying mechanism.
Collapse
Affiliation(s)
- Liufeng Mao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Effects of supplemented isoenergetic diets varying in cereal fiber and protein content on the bile acid metabolic signature and relation to insulin resistance. Nutr Diabetes 2018; 8:11. [PMID: 29549243 PMCID: PMC5856807 DOI: 10.1038/s41387-018-0020-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/16/2018] [Accepted: 01/21/2018] [Indexed: 12/13/2022] Open
Abstract
Bile acids (BA) are potent metabolic regulators influenced by diet. We studied effects of isoenergetic increases in the dietary protein and cereal-fiber contents on circulating BA and insulin resistance (IR) in overweight and obese adults. Randomized controlled nutritional intervention (18 weeks) in 72 non-diabetic participants (overweight/obese: 29/43) with at least one further metabolic risk factor. Participants were group-matched and allocated to four isoenergetic supplemented diets: control; high cereal fiber (HCF); high-protein (HP); or moderately increased cereal fiber and protein (MIX). Whole-body IR and insulin-mediated suppression of hepatic endogenous glucose production were measured using euglycaemic–hyperinsulinemic clamps with [6-62H2] glucose infusion. Circulating BA, metabolic biomarkers, and IR were measured at 0, 6, and 18 weeks. Under isoenergetic conditions, HP-intake worsened IR in obese participants after 6 weeks (M-value: 3.77 ± 0.58 vs. 3.07 ± 0.44 mg/kg/min, p = 0.038), with partial improvement back to baseline levels after 18 weeks (3.25 ± 0.45 mg/kg/min, p = 0.089). No deleterious effects of HP-intake on IR were observed in overweight participants. HCF-diet improved IR in overweight participants after 6 weeks (M-value 4.25 ± 0.35 vs. 4.81 ± 0.31 mg/kg/min, p = 0.016), but did not influence IR in obese participants. Control and MIX diets did not influence IR. HP-induced, but not HCF-induced changes in IR strongly correlated with changes of BA profiles. MIX-diet significantly increased most BA at 18 weeks in obese, but not in overweight participants. BA remained unchanged in controls. Pooled BA concentrations correlated with fasting fibroblast growth factor-19 (FGF-19) plasma levels (r = 0.37; p = 0.003). Higher milk protein intake was the only significant dietary predictor for raised total and primary BA in regression analyses (total BA, p = 0.017; primary BA, p = 0.011). Combined increased intake of dietary protein and cereal fibers markedly increased serum BA concentrations in obese, but not in overweight participants. Possible mechanisms explaining this effect may include compensatory increases of the BA pool in the insulin resistant, obese state; or defective BA transport.
Collapse
|
23
|
Yao S, Agyei D, Udenigwe CC. Structural Basis of Bioactivity of Food Peptides in Promoting Metabolic Health. ADVANCES IN FOOD AND NUTRITION RESEARCH 2018; 84:145-181. [PMID: 29555068 DOI: 10.1016/bs.afnr.2017.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Bioactive peptides have many structural features that enable them to become functional in controlling several biological processes in the body, especially those related to metabolic health. This chapter provides an overview of the multiple targets of food-derived peptides against metabolic health problems (e.g., hypertension, dyslipidemia, hyperglycemia, oxidative stress) and discusses the importance of structural chemistry in determining the bioactivities of peptides and protein hydrolysates.
Collapse
Affiliation(s)
- Shixiang Yao
- Southwest University, Chongqing, PR China; University of Ottawa, Ottawa, ON, Canada
| | | | | |
Collapse
|
24
|
Supplementation with an insoluble fiber obtained from carob pod (Ceratonia siliqua L.) rich in polyphenols prevents dyslipidemia in rabbits through SIRT1/PGC-1α pathway. Eur J Nutr 2017; 58:357-366. [DOI: 10.1007/s00394-017-1599-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/14/2017] [Indexed: 02/07/2023]
|
25
|
Madsen L, Myrmel LS, Fjære E, Liaset B, Kristiansen K. Links between Dietary Protein Sources, the Gut Microbiota, and Obesity. Front Physiol 2017; 8:1047. [PMID: 29311977 PMCID: PMC5742165 DOI: 10.3389/fphys.2017.01047] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/30/2017] [Indexed: 12/22/2022] Open
Abstract
The association between the gut microbiota and obesity is well documented in both humans and in animal models. It is also demonstrated that dietary factors can change the gut microbiota composition and obesity development. However, knowledge of how diet, metabolism and gut microbiota mutually interact and modulate energy metabolism and obesity development is still limited. Epidemiological studies indicate an association between intake of certain dietary protein sources and obesity. Animal studies confirm that different protein sources vary in their ability to either prevent or induce obesity. Different sources of protein such as beans, vegetables, dairy, seafood, and meat differ in amino acid composition. Further, the type and level of other factors, such as fatty acids and persistent organic pollutants (POPs) vary between dietary protein sources. All these factors can modulate the composition of the gut microbiota and may thereby influence their obesogenic properties. This review summarizes evidence of how different protein sources affect energy efficiency, obesity development, and the gut microbiota, linking protein-dependent changes in the gut microbiota with obesity.
Collapse
Affiliation(s)
- Lise Madsen
- National Institute of Nutrition and Seafood Research, Bergen, Norway.,Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark.,BGI-Shenzhen, Shenzhen, China
| | - Lene S Myrmel
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Even Fjære
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Bjørn Liaset
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark.,BGI-Shenzhen, Shenzhen, China
| |
Collapse
|
26
|
Mazidi M, de Caravatto PPP, Speakman JR, Cohen RV. Mechanisms of Action of Surgical Interventions on Weight-Related Diseases: the Potential Role of Bile Acids. Obes Surg 2017; 27:826-836. [PMID: 28091894 DOI: 10.1007/s11695-017-2549-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Surgical interventions for weight-related diseases (SWRD) may have substantial and sustainable effect on weight reduction, also leading to a higher remission rate of type 2 diabetes (T2D) mellitus than any other medical treatment or lifestyle intervention. The resolution of T2D after Roux-en-Y gastric bypass (RYGB) typically occurs too quickly to be accounted for by weight loss alone, suggesting that these operations have a direct impact on glucose homeostasis. The mechanisms underlying these beneficial effects however remain unclear. Recent research suggests that changes in the concentrations of plasma bile acids might contribute to these metabolic changes after surgery. In this review, we aimed to outline the potential role of bile acids in SWRD. We systematically reviewed MEDLINE, SCOPUS, and Web of Science for articles reporting the effect of SWRD on outcomes published between 1969 and 2016. We found that changes in circulating bile acids after surgery may play a major role through activation of the farnesoid X receptor A (FXRA), the fibroblast growth factor 19 (FGF19), and the G protein-coupled bile acid receptor (TGR5). Bile acid concentration increased significantly after RYGB. Some studies suggest that a transitory decrease occurs at 1 week post-surgery, followed by a gradual increase. Most studies have shown the increase to be proportionate by all bile acid subtypes. Bile acids can regulate glucose metabolism through the expression of TGR5 receptor in L cells, resulting in a release of glucagon-like peptide 1 (GLP-1). It may also induce the synthesis and secretion of FGF19 in ileal cells, thereby improving insulin sensitivity and regulating glucose metabolism. All the present SWRD are involved with changes in food stimulation to the stomach. This implies that discovering and developing the antagonists to TGR5 and FXRA may effectively control metabolic syndrome and the elucidation of the mechanisms underlying the physiological effects related to weight loss and T2D remission after surgery may help to identify new drug targets.
Collapse
Affiliation(s)
- Mohsen Mazidi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China.,University of the Chinese Academy of Sciences, Huairou, Beijing, China
| | - Pedro Paulo P de Caravatto
- The Center for Obesity and Diabetes, Oswaldo Cruz German Hospital, Rua Cincinato Braga, 37 5o. andar, São Paulo, São Paulo, Brazil
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China.,Institute of Biological and Environmental Science, University of Aberdeen, Aberdeen, Scotland, UK
| | - Ricardo V Cohen
- The Center for Obesity and Diabetes, Oswaldo Cruz German Hospital, Rua Cincinato Braga, 37 5o. andar, São Paulo, São Paulo, Brazil.
| |
Collapse
|
27
|
Diet-induced obesity and weight loss alter bile acid concentrations and bile acid-sensitive gene expression in insulin target tissues of C57BL/6J mice. Nutr Res 2017; 46:11-21. [PMID: 29173647 DOI: 10.1016/j.nutres.2017.07.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/13/2017] [Accepted: 07/20/2017] [Indexed: 12/31/2022]
Abstract
Bile acids (BAs) influence the metabolism of glucose, lipids, and energy expenditure. We hypothesized that BA concentrations and related gene expression would be altered in lean (low-fat diet fed; LFD) vs diet-induced obese (high-fat diet fed; HFD) groups of mice and that some detected changes would remain after weight loss in an HFD group switched to the LFD (SW). Taurine conjugates dominated the bile acid composition of the liver, epididymal white adipose tissue (eWAT), and hypothalamus, with the latter having lower levels (~95%, ~95%, and ~80%, respectively; P<.05). Plasma conjugated bile acids were elevated in the HFD relative to the LFD and SW animals. Total hepatic BA concentrations decreased in obese mice fed HFD, and levels returned to preobese levels in the SW group. Subtle changes in unconjugated bile acids were detected in the eWAT, hypothalamus, and muscle. Liver expression of a variety of enzymes involved in BA synthesis (eg, Cyp27a1, Acox2), BA transport (eg, Slc22a8), and BA-sensitive receptors (Fxr, Tgr5) were unchanged by HFD feeding but decreased with SW. Other hepatic enzymes were induced in the SW group (eg, Amacr and Bal). In eWAT, Cyp27a1 and Acox2 also declined in the SW group, whereas the HFD group showed reduced expression of BA transporters (eg, Abcc3), and changes in Fxr and Tgr5 were unclear. Therefore, although most detectable changes in BA metabolism associated with diet-induced obesity are reversed by diet-induced weight loss, some effects on BA composition, concentrations, and gene expression can persist after weight loss.
Collapse
|
28
|
Vargas-Castillo A, Fuentes-Romero R, Rodriguez-Lopez LA, Torres N, Tovar AR. Understanding the Biology of Thermogenic Fat: Is Browning A New Approach to the Treatment of Obesity? Arch Med Res 2017; 48:401-413. [DOI: 10.1016/j.arcmed.2017.10.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/17/2017] [Indexed: 12/18/2022]
|
29
|
Bonet ML, Mercader J, Palou A. A nutritional perspective on UCP1-dependent thermogenesis. Biochimie 2017; 134:99-117. [DOI: 10.1016/j.biochi.2016.12.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 12/23/2016] [Indexed: 12/16/2022]
|
30
|
Shi L, Wang J, Wang Y, Feng Y. MDG-1, an Ophiopogon polysaccharide, alleviates hyperlipidemia in mice based on metabolic profile of bile acids. Carbohydr Polym 2016; 150:74-81. [PMID: 27312615 DOI: 10.1016/j.carbpol.2016.05.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/01/2016] [Accepted: 05/04/2016] [Indexed: 01/15/2023]
Abstract
Hyperlipidemia is a chronic metabolic disorder with systemic complications that is prevalent worldwide. MDG-1, a water-soluble β-d-fructan polysaccharide from Ophiopogon japonicas has potent hypolipidemic and weight-control effects. The present study aimed to investigate the effects of MDG-1 on lipid metabolic disorders in diet-induced obese mice based on the metabolic profile of bile acids. C57BL/6 mice were treated with a low-fat diet, high-fat diet or high fat mixed with 1‰ (w/w) MDG-1 diet for 12 weeks. The results showed that MDG-1 inhibited body weight gain and lowered serum and liver total cholesterol contents in obese mice. In addition, MDG-1 could adsorb bile acids in the gut lumen and reduce their reabsorption, thus promoting cholesterol catabolism. Furthermore, MDG-1 inhibited the expression of the farnesoid X receptor, but activated the liver X receptor. Our findings shed new light on the mechanism of MDG-1 in the control of lipids.
Collapse
Affiliation(s)
- Linlin Shi
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Jie Wang
- Institute of Chinese Material Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Yuan Wang
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China.
| | - Yi Feng
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China.
| |
Collapse
|
31
|
Aadland EK, Graff IE, Lavigne C, Eng Ø, Paquette M, Holthe A, Mellgren G, Madsen L, Jacques H, Liaset B. Lean Seafood Intake Reduces Postprandial C-peptide and Lactate Concentrations in Healthy Adults in a Randomized Controlled Trial with a Crossover Design. J Nutr 2016; 146:1027-34. [PMID: 27099232 DOI: 10.3945/jn.115.229278] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/07/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Recently we showed that lean seafood consumption reduced circulating triacylglycerol (TG) and VLDL concentrations and prevented an elevated total-to-HDL-cholesterol ratio relative to intake of a nonseafood diet. OBJECTIVE We aimed to elucidate whether diet-induced altered carbohydrate metabolism could be a contributing factor to the previously observed different lipoprotein patterns. METHODS This was a secondary outcome and explorative randomized controlled trial with a crossover design in 20 healthy adults (7 men and 13 women) that were 50.6 ± 3.4 (mean ± SEM) y old, weighed 75.7 ± 2.5 kg, and had a body mass index (BMI, in kg/m(2)) of 25.6 ± 0.7. After a 3-wk run-in period and separated by a 5-wk wash-out period, the participants consumed 2 balanced diets [in percentage of energy (energy%); 29% fat, 52% carbohydrates, 19% protein] for 4 wk. The diets varied in the main protein sources; 60 energy% of total protein was from either lean seafood or nonseafood sources. On the first and last day of each diet period, fasting and postprandial blood samples were collected before and after consumption of test meals (in energy%; 28% fat, 52% carbohydrates, 20% protein) with cod or lean beef. RESULTS The diets did not alter serum insulin and glucose concentrations. However, relative to the nonseafood diet period, the lean seafood diet period reduced postprandial C-peptide (P = 0.04) and lactate (P = 0.012) concentrations and fasting and postprandial TG/HDL-cholesterol ratios (P = 0.002). Hence, different postprandial lactate levels occurred at equal glucose concentrations. CONCLUSIONS Even though the diets did not alter serum insulin and glucose concentrations, intake of the lean seafood compared with the nonseafood diet reduced postprandial concentrations of C-peptide and lactate and the TG/HDL-cholesterol ratio in healthy adults in a manner that may affect the long-term development of insulin resistance, type 2 diabetes, and cardiovascular disease. This trial was registered at www.clinicaltrials.gov as NCT01708681.
Collapse
Affiliation(s)
- Eli K Aadland
- National Institute of Nutrition and Seafood Research, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway; Faculty of Education, Bergen University College, Bergen, Norway
| | - Ingvild E Graff
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Charles Lavigne
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Øyvin Eng
- Haukeland University Hospital, Hormone Laboratory, Bergen, Norway
| | | | - Asle Holthe
- Faculty of Education, Bergen University College, Bergen, Norway
| | - Gunnar Mellgren
- Department of Clinical Science, University of Bergen, Bergen, Norway; Haukeland University Hospital, Hormone Laboratory, Bergen, Norway
| | - Lise Madsen
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Hélène Jacques
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bjørn Liaset
- National Institute of Nutrition and Seafood Research, Bergen, Norway;
| |
Collapse
|
32
|
Holm JB, Rønnevik A, Tastesen HS, Fjære E, Fauske KR, Liisberg U, Madsen L, Kristiansen K, Liaset B. Diet-induced obesity, energy metabolism and gut microbiota in C57BL/6J mice fed Western diets based on lean seafood or lean meat mixtures. J Nutr Biochem 2016; 31:127-36. [DOI: 10.1016/j.jnutbio.2015.12.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 11/27/2015] [Accepted: 12/22/2015] [Indexed: 01/21/2023]
|
33
|
Liisberg U, Myrmel LS, Fjære E, Rønnevik AK, Bjelland S, Fauske KR, Holm JB, Basse AL, Hansen JB, Liaset B, Kristiansen K, Madsen L. The protein source determines the potential of high protein diets to attenuate obesity development in C57BL/6J mice. Adipocyte 2016; 5:196-211. [PMID: 27386160 DOI: 10.1080/21623945.2015.1122855] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 10/28/2015] [Accepted: 11/12/2015] [Indexed: 10/22/2022] Open
Abstract
The notion that the obesogenic potential of high fat diets in rodents is attenuated when the protein:carbohydrate ratio is increased is largely based on studies using casein or whey as the protein source. We fed C57BL/6J mice high fat-high protein diets using casein, soy, cod, beef, chicken or pork as protein sources. Casein stood out as the most efficient in preventing weight gain and accretion of adipose mass. By contrast, mice fed diets based on pork or chicken, and to a lesser extent mice fed cod or beef protein, had increased adipose tissue mass gain relative to casein fed mice. Decreasing the protein:carbohydrate ratio in diets with casein or pork as protein sources led to accentuated fat mass accumulation. Pork fed mice were more obese than casein fed mice, and relative to casein, the pork-based feed induced substantial accumulation of fat in classic interscapular brown adipose tissue accompanied by decreased UCP1 expression. Furthermore, intake of a low fat diet with casein, but not pork, as a protein source reversed diet-induced obesity. Compared to pork, casein seems unique in maintaining the classical brown morphology in interscapular brown adipose tissue with high UCP1 expression. This was accompanied by increased expression of genes involved in a futile cycling of fatty acids. Our results demonstrate that intake of high protein diets based on other protein sources may not have similar effects, and hence, the obesity protective effect of high protein diets is clearly modulated by protein source.
Collapse
Affiliation(s)
- Ulrike Liisberg
- National Institute of Nutrition and Seafood Research, Bergen, Norway
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Lene Secher Myrmel
- National Institute of Nutrition and Seafood Research, Bergen, Norway
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Even Fjære
- National Institute of Nutrition and Seafood Research, Bergen, Norway
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Alexander K. Rønnevik
- National Institute of Nutrition and Seafood Research, Bergen, Norway
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Bjelland
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | | | - Jacob Bak Holm
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Jacob B. Hansen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Bjørn Liaset
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | | | - Lise Madsen
- National Institute of Nutrition and Seafood Research, Bergen, Norway
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Jensen IJ, Walquist M, Liaset B, Elvevoll EO, Eilertsen KE. Dietary intake of cod and scallop reduces atherosclerotic burden in female apolipoprotein E-deficient mice fed a Western-type high fat diet for 13 weeks. Nutr Metab (Lond) 2016; 13:8. [PMID: 26839578 PMCID: PMC4735963 DOI: 10.1186/s12986-016-0068-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/21/2016] [Indexed: 01/25/2023] Open
Abstract
Background It is now increasingly recognized that the beneficial effects of seafood consumption is not limited to lipids and fatty acid, but that the protein part, i.e., peptides and amino acids, together with vitamins and even unknown bioactive constituents also are important for disease prevention. This study was designed to evaluate the putative anti-atherogenic effects of different protein sources (a lean seafood and a nonseafood) in apolipoprotein E-deficient (apoE−/−) mice. Methods Twenty-four 5-week-old female apoE−/− mice were fed Western type diets containing chicken or a combination of cod and scallops as dietary protein sources for 13 weeks. Atherosclerotic plaque burden, weight, serum levels of leptin, glucose and LDL cholesterol as well as gene expressions from liver and heart were evaluated. Statistical analyses were performed using SPSS. Differences between the variables were evaluated using independent t-test or Mann–Whitney U test for normally and non-normally distributed variables, respectively. Normality was defined by the Shapiro-Wilk test. Results The mice fed cod-scallop had a 24 % (p < 0.05) reduced total aorta atherosclerotic plaque burden compared to the chicken fed group, whereas the reduction in plaque in the less lesion prone thoracic and abdominal parts of the descending aorta were 46 % (p < 0.05) and 56 % (p < 0.05), respectively. In addition, mice fed cod-scallop gained less weight, and had lower serum levels of leptin, glucose and LDL cholesterol, compared to those fed chicken. Analysis of expression of the genes from liver and heart showed that hepatic endogenous antioxidant paraoxonase 2 (Pon2 gene) and the vascular cell adhesion molecule VCAM-1 (Vcam1 gene) were down regulated in mice fed cod-scallop compared to mice fed chicken. Conclusion The present study revealed a metabolic beneficial effect of lean seafood compared to chicken, as atherosclerotic plaque burden, serum glucose, leptin and LDL cholesterol levels were reduced in mice fed cod-scallop. Electronic supplementary material The online version of this article (doi:10.1186/s12986-016-0068-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ida-Johanne Jensen
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UIT - The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Mari Walquist
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UIT - The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Bjørn Liaset
- National Institute of Nutrition and Seafood Research, 5004 Bergen, Norway
| | - Edel O Elvevoll
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UIT - The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Karl-Erik Eilertsen
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UIT - The Arctic University of Norway, N-9037 Tromsø, Norway
| |
Collapse
|
35
|
Carr RM, Reid AE. FXR agonists as therapeutic agents for non-alcoholic fatty liver disease. Curr Atheroscler Rep 2015; 17:500. [PMID: 25690590 DOI: 10.1007/s11883-015-0500-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome and a risk factor for both cardiovascular and hepatic related morbidity and mortality. The increasing prevalence of this disease requires novel therapeutic approaches to prevent disease progression. Farnesoid X receptors are bile acid receptors with roles in lipid, glucose, and energy homeostasis. Synthetic farnesoid X receptor (FXR) agonists have been developed to specifically target these receptors for therapeutic use in NAFLD patients. Here, we present a review of bile acid physiology and how agonism of FXR receptors has been examined in pre-clinical and clinical NAFLD. Early evidence suggests a potential role for synthetic FXR agonists in the management of NAFLD; however, additional studies are needed to clarify their effects on lipid and glucose parameters in humans.
Collapse
Affiliation(s)
- Rotonya M Carr
- Division of Gastroenterology, University of Pennsylvania, 421 Curie Boulevard, 907 Biomedical Research Building, Philadelphia, PA, 19104, USA,
| | | |
Collapse
|
36
|
Stadlbauer V, Leber B, Lemesch S, Trajanoski S, Bashir M, Horvath A, Tawdrous M, Stojakovic T, Fauler G, Fickert P, Högenauer C, Klymiuk I, Stiegler P, Lamprecht M, Pieber TR, Tripolt NJ, Sourij H. Lactobacillus casei Shirota Supplementation Does Not Restore Gut Microbiota Composition and Gut Barrier in Metabolic Syndrome: A Randomized Pilot Study. PLoS One 2015; 10:e0141399. [PMID: 26509793 PMCID: PMC4625062 DOI: 10.1371/journal.pone.0141399] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/05/2015] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Metabolic syndrome is associated with disturbances in gut microbiota composition. We aimed to investigate the effect of Lactobacillus casei Shirota (LcS) on gut microbiota composition, gut barrier integrity, intestinal inflammation and serum bile acid profile in metabolic syndrome. In a single-centre, prospective, randomised controlled pilot study, 28 subjects with metabolic syndrome received either LcS for 12 weeks (n = 13) or no LcS (n = 15). Data were compared to healthy controls (n = 16). Gut microbiota composition was characterised from stool using 454 pyrosequencing of 16S rRNA genes. Serum bile acids were quantified by tandem mass spectrometry. Zonulin and calprotectin were measured in serum and stool by ELISA. Bacteroidetes/Firmicutes ratio was significantly higher in healthy controls compared to metabolic syndrome but was not influenced by LcS. LcS supplementation led to enrichment of Parabacteroides. Zonulin and calprotectin were increased in metabolic syndrome stool samples but not influenced by LcS supplementation. Serum bile acids were similar to controls and not influenced by LcS supplementation. Metabolic syndrome is associated with a higher Bacteroidetes/Firmicutes ratio and gut barrier dysfunction but LcS was not able to change this. LcS administration was associated with subtle microbiota changes at genus level. TRIAL REGISTRATION ClinicalTrials.gov NCT01182844.
Collapse
Affiliation(s)
- Vanessa Stadlbauer
- Medical University of Graz, Division of Gastroenterology and Hepatology, Graz, Austria
| | - Bettina Leber
- Medical University of Graz, Division of Transplantation Surgery, Graz, Austria
| | - Sandra Lemesch
- Medical University of Graz, Division of Gastroenterology and Hepatology, Graz, Austria
| | | | - Mina Bashir
- Medical University of Graz, Division of Endocrinology and Metabolism, Graz, Austria
| | - Angela Horvath
- Medical University of Graz, Division of Gastroenterology and Hepatology, Graz, Austria
| | - Monika Tawdrous
- Medical University of Graz, Division of Gastroenterology and Hepatology, Graz, Austria
| | - Tatjana Stojakovic
- Medical University of Graz, Clinical Institute of Medical and Chemical Laboratory Diagnostics, Graz, Austria
| | - Günter Fauler
- Medical University of Graz, Clinical Institute of Medical and Chemical Laboratory Diagnostics, Graz, Austria
| | - Peter Fickert
- Medical University of Graz, Division of Gastroenterology and Hepatology, Graz, Austria
| | - Christoph Högenauer
- Medical University of Graz, Division of Gastroenterology and Hepatology, Graz, Austria
| | | | - Philipp Stiegler
- Medical University of Graz, Division of Transplantation Surgery, Graz, Austria
| | - Manfred Lamprecht
- Medical University of Graz, Institute of Physiological Chemistry, Centre for Physiological Medicine, Graz, Austria
| | - Thomas R. Pieber
- Medical University of Graz, Division of Endocrinology and Metabolism, Graz, Austria
- Centre for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Norbert J. Tripolt
- Medical University of Graz, Division of Endocrinology and Metabolism, Graz, Austria
| | - Harald Sourij
- Medical University of Graz, Division of Endocrinology and Metabolism, Graz, Austria
- Centre for Biomarker Research in Medicine (CBmed), Graz, Austria
| |
Collapse
|
37
|
Myrmel LS, Fjære E, Midtbø LK, Bernhard A, Petersen RK, Sonne SB, Mortensen A, Hao Q, Brattelid T, Liaset B, Kristiansen K, Madsen L. Macronutrient composition determines accumulation of persistent organic pollutants from dietary exposure in adipose tissue of mice. J Nutr Biochem 2015; 27:307-16. [PMID: 26507541 DOI: 10.1016/j.jnutbio.2015.09.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 09/08/2015] [Accepted: 09/22/2015] [Indexed: 01/27/2023]
Abstract
Accumulation of persistent organic pollutants (POPs) has been linked to adipose tissue expansion. As different nutrients modulate adipose tissue development, we investigated the influence of dietary composition on POP accumulation, obesity development and related disorders. Lifespan was determined in mice fed fish-oil-based high fat diets during a long-term feeding trial and accumulation of POPs was measured after 3, 6 and 18months of feeding. Further, we performed dose-response experiments using four abundant POPs found in marine sources, PCB-153, PCB-138, PCB-118 and pp'-DDE as single congeners or as mixtures in combination with different diets: one low fat diet and two high fat diets with different protein:sucrose ratios. We measured accumulation of POPs in adipose tissue and liver and determined obesity development, glucose tolerance, insulin sensitivity and hepatic expression of genes involved in metabolism of xenobiotics. Compared with mice fed diets with a low protein:sucrose ratio, mice fed diets with a high protein:sucrose ratio had significantly lower total burden of POPs in adipose tissue, were protected from obesity development and exhibited enhanced hepatic expression of genes involved in metabolism and elimination of xenobiotics. Exposure to POPs, either as single compounds or mixtures, had no effect on obesity development, glucose tolerance or insulin sensitivity. In conclusion, this study demonstrates that the dietary composition of macronutrients profoundly modulates POP accumulation in adipose tissues adding an additional parameter to be included in future studies. Our results indicate that alterations in macronutrient composition might be an additional route for reducing total body burden of POPs.
Collapse
Affiliation(s)
- Lene Secher Myrmel
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark; National Institute of Nutrition and Seafood Research, 5817 Bergen, Norway
| | - Even Fjære
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark; National Institute of Nutrition and Seafood Research, 5817 Bergen, Norway
| | - Lisa Kolden Midtbø
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark; National Institute of Nutrition and Seafood Research, 5817 Bergen, Norway
| | - Annette Bernhard
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark; National Institute of Nutrition and Seafood Research, 5817 Bergen, Norway
| | - Rasmus Koefoed Petersen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Si Brask Sonne
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Alicja Mortensen
- Division of Risk Assessment and Nutrition, National Food Institute, Technical University of Denmark, 2800 Copenhagen, Denmark
| | - Qin Hao
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Trond Brattelid
- National Institute of Nutrition and Seafood Research, 5817 Bergen, Norway
| | - Bjørn Liaset
- National Institute of Nutrition and Seafood Research, 5817 Bergen, Norway
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark.
| | - Lise Madsen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark; National Institute of Nutrition and Seafood Research, 5817 Bergen, Norway.
| |
Collapse
|
38
|
Chevrier G, Mitchell PL, Rioux LE, Hasan F, Jin T, Roblet CR, Doyen A, Pilon G, St-Pierre P, Lavigne C, Bazinet L, Jacques H, Gill T, McLeod RS, Marette A. Low-Molecular-Weight Peptides from Salmon Protein Prevent Obesity-Linked Glucose Intolerance, Inflammation, and Dyslipidemia in LDLR-/-/ApoB100/100 Mice. J Nutr 2015; 145:1415-22. [PMID: 25995281 DOI: 10.3945/jn.114.208215] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/22/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND We previously reported that fish proteins can alleviate metabolic syndrome (MetS) in obese animals and human subjects. OBJECTIVES We tested whether a salmon peptide fraction (SPF) could improve MetS in mice and explored potential mechanisms of action. METHODS ApoB(100) only, LDL receptor knockout male mice (LDLR(-/-)/ApoB(100/100)) were fed a high-fat and -sucrose (HFS) diet (25 g/kg sucrose). Two groups were fed 10 g/kg casein hydrolysate (HFS), and 1 group was additionally fed 4.35 g/kg fish oil (FO; HFS+FO). Two other groups were fed 10 g SPF/kg (HFS+SPF), and 1 group was additionally fed 4.35 g FO/kg (HFS+SPF+FO). A fifth (reference) group was fed a standard feed pellet diet. We assessed the impact of dietary treatments on glucose tolerance, adipose tissue inflammation, lipid homeostasis, and hepatic insulin signaling. The effects of SPF on glucose uptake, hepatic glucose production, and inducible nitric oxide synthase activity were further studied in vitro with the use of L6 myocytes, FAO hepatocytes, and J774 macrophages. RESULTS Mice fed HFS+SPF or HFS+SPF+FO diets had lower body weight (protein effect, P = 0.024), feed efficiency (protein effect, P = 0.018), and liver weight (protein effect, P = 0.003) as well as lower concentrations of adipose tissue cytokines and chemokines (protein effect, P ≤ 0.003) compared with HFS and HFS+FO groups. They also had greater glucose tolerance (protein effect, P < 0.001), lower activation of the mammalian target of rapamycin complex 1/S6 kinase 1/insulin receptor substrate 1 (mTORC1/S6K1/IRS1) pathway, and increased insulin signaling in liver compared with the HFS and HFS+FO groups. The HFS+FO, HFS+SPF, and HFS+SPF+FO groups had lower plasma triglycerides (protein effect, P = 0.003; lipid effect, P = 0.002) than did the HFS group. SPF increased glucose uptake and decreased HGP and iNOS activation in vitro. CONCLUSIONS SPF reduces obesity-linked MetS features in LDLR(-/-)/ApoB(100/100) mice. The anti-inflammatory and glucoregulatory properties of SPF were confirmed in L6 myocytes, FAO hepatocytes, and J774 macrophages.
Collapse
Affiliation(s)
- Geneviève Chevrier
- Department of Medicine, Quebec Heart and Lung Institute, Institute of Nutrition and Functional Foods, and
| | - Patricia L Mitchell
- Department of Medicine, Quebec Heart and Lung Institute, Institute of Nutrition and Functional Foods, and
| | - Laurie-Eve Rioux
- Department of Medicine, Quebec Heart and Lung Institute, Institute of Nutrition and Functional Foods, and
| | - Fida Hasan
- Departments of Process Engineering and Applied Science and
| | - Tianyi Jin
- Departments of Process Engineering and Applied Science and
| | - Cyril Roland Roblet
- Institute of Nutrition and Functional Foods, and Department of Food Sciences, Laval University, Quebec City, Canada; and
| | - Alain Doyen
- Institute of Nutrition and Functional Foods, and Department of Food Sciences, Laval University, Quebec City, Canada; and
| | - Geneviève Pilon
- Department of Medicine, Quebec Heart and Lung Institute, Institute of Nutrition and Functional Foods, and
| | - Philippe St-Pierre
- Department of Medicine, Quebec Heart and Lung Institute, Institute of Nutrition and Functional Foods, and
| | - Charles Lavigne
- Department of Medicine, Quebec Heart and Lung Institute, Institute of Nutrition and Functional Foods, and
| | - Laurent Bazinet
- Institute of Nutrition and Functional Foods, and Department of Food Sciences, Laval University, Quebec City, Canada; and
| | - Hélène Jacques
- Institute of Nutrition and Functional Foods, and Department of Food Sciences, Laval University, Quebec City, Canada; and
| | - Tom Gill
- Departments of Process Engineering and Applied Science and
| | - Roger S McLeod
- Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - André Marette
- Department of Medicine, Quebec Heart and Lung Institute, Institute of Nutrition and Functional Foods, and
| |
Collapse
|
39
|
Lysne V, Bjørndal B, Vik R, Nordrehaug JE, Skorve J, Nygård O, Berge RK. A Protein Extract from Chicken Reduces Plasma Homocysteine in Rats. Nutrients 2015; 7:4498-511. [PMID: 26053618 PMCID: PMC4488798 DOI: 10.3390/nu7064498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/27/2015] [Accepted: 05/29/2015] [Indexed: 11/16/2022] Open
Abstract
The present study aimed to evaluate effects of a water-soluble protein fraction of chicken (CP), with a low methionine/glycine ratio, on plasma homocysteine and metabolites related to homocysteine metabolism. Male Wistar rats were fed either a control diet with 20% w/w casein as the protein source, or an experimental diet where 6, 14 or 20% w/w of the casein was replaced with the same amount of CP for four weeks. Rats fed CP had reduced plasma total homocysteine level and markedly increased levels of the choline pathway metabolites betaine, dimethylglycine, sarcosine, glycine and serine, as well as the transsulfuration pathway metabolites cystathionine and cysteine. Hepatic mRNA level of enzymes involved in homocysteine remethylation, methionine synthase and betaine-homocysteine S-methyltransferase, were unchanged, whereas cystathionine gamma-lyase of the transsulfuration pathway was increased in the CP treated rats. Plasma concentrations of vitamin B2, folate, cobalamin, and the B-6 catabolite pyridoxic acid were increased in the 20% CP-treated rats. In conclusion, the CP diet was associated with lower plasma homocysteine concentration and higher levels of serine, choline oxidation and transsulfuration metabolites compared to a casein diet. The status of related B-vitamins was also affected by CP.
Collapse
Affiliation(s)
- Vegard Lysne
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway.
- Department of Heart Disease, Haukeland University Hospital, 5021 Bergen, Norway.
| | - Bodil Bjørndal
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway.
| | - Rita Vik
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway.
| | | | - Jon Skorve
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway.
| | - Ottar Nygård
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway.
- Department of Heart Disease, Haukeland University Hospital, 5021 Bergen, Norway.
- KG Jebsen Centre for Diabetes Research, University of Bergen, 5020 Bergen, Norway.
| | - Rolf K Berge
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway.
- Department of Heart Disease, Haukeland University Hospital, 5021 Bergen, Norway.
| |
Collapse
|
40
|
Scallop protein with endogenous high taurine and glycine content prevents high-fat, high-sucrose-induced obesity and improves plasma lipid profile in male C57BL/6J mice. Amino Acids 2015; 46:1659-71. [PMID: 24658997 PMCID: PMC4055845 DOI: 10.1007/s00726-014-1715-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/25/2014] [Indexed: 01/13/2023]
Abstract
High-protein diets induce alterations in metabolism that may prevent diet-induced obesity. However, little is known as to whether different protein sources consumed at normal levels may affect diet-induced obesity and associated co-morbidities. We fed obesity-prone male C57BL/6J mice high-fat, high-sucrose diets with protein sources of increasing endogenous taurine content, i.e., chicken, cod, crab and scallop, for 6 weeks. The energy intake was lower in crab and scallop-fed mice than in chicken and cod-fed mice, but only scallop-fed mice gained less body and fat mass. Liver mass was reduced in scallop-fed mice, but otherwise no changes in lean body mass were observed between the groups. Feed efficiency and apparent nitrogen digestibility were reduced in scallop-fed mice suggesting alterations in energy utilization and metabolism. Overnight fasted plasma triacylglyceride, non-esterified fatty acids, glycerol and hydroxy-butyrate levels were significantly reduced, indicating reduced lipid mobilization in scallop-fed mice. The plasma HDL-to-total-cholesterol ratio was higher, suggesting increased reverse cholesterol transport or cholesterol clearance in scallop-fed mice in both fasted and non-fasted states. Dietary intake of taurine and glycine correlated negatively with body mass gain and total fat mass, while intake of all other amino acids correlated positively. Furthermore taurine and glycine intake correlated positively with improved plasma lipid profile, i.e., lower levels of plasma lipids and higher HDL-to-total-cholesterol ratio. In conclusion, dietary scallop protein completely prevents high-fat, high-sucrose-induced obesity whilst maintaining lean body mass and improving the plasma lipid profile in male C57BL/6J mice.
Collapse
|
41
|
Song Y, Xu C, Shao S, Liu J, Xing W, Xu J, Qin C, Li C, Hu B, Yi S, Xia X, Zhang H, Zhang X, Wang T, Pan W, Yu C, Wang Q, Lin X, Wang L, Gao L, Zhao J. Thyroid-stimulating hormone regulates hepatic bile acid homeostasis via SREBP-2/HNF-4α/CYP7A1 axis. J Hepatol 2015; 62:1171-9. [PMID: 25533663 DOI: 10.1016/j.jhep.2014.12.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/03/2014] [Accepted: 12/03/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Bile acids (BAs) play a crucial role in dietary fat digestion and in the regulation of lipid, glucose, and energy metabolism. Thyroid-stimulating hormone (TSH) is a hormone produced by the anterior pituitary gland that directly regulates several metabolic pathways. However, the impact of TSH on BA homeostasis remains largely unknown. METHODS We analyzed serum BA and TSH levels in healthy volunteers under strict control of caloric intake. Thyroidectomized rats were administered thyroxine and injected with different doses of TSH. Tshr(-/-) mice were supplemented with thyroxine, and C57BL/6 mice were injected with Tshr-siRNA via the tail vein. The serum BA levels, BA pool size, and fecal BA excretion rate were measured. The regulation of SREBP-2, HNF-4α, and CYP7A1 by TSH were analyzed using luciferase reporter, RNAi, EMSA, and CHIP assays. RESULTS A negative correlation was observed between the serum levels of TSH and the serum BA levels in healthy volunteers. TSH administration led to a decrease in BA content and CYP7A1 activity in thyroidectomized rats supplemented with thyroxine. When Tshr was silenced in mice, the BA pool size, fecal BA excretion rate, and serum BA levels all increased. Additionally, we found that HNF-4α acts as a critical molecule through which TSH represses CYP7A1 activity. We further confirmed that the accumulation of mature SREBP-2 protein could impair the capacity of nuclear HNF-4α to bind to the CYP7A1 promoter, a mechanism that appears to mediate the effects of TSH. CONCLUSIONS TSH represses hepatic BA synthesis via a SREBP-2/HNF-4α/CYP7A1 signaling pathway. This finding strongly supports the notion that TSH is an important pathophysiological regulator of liver BA homeostasis independently of thyroid hormones.
Collapse
Affiliation(s)
- Yongfeng Song
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Chao Xu
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Shanshan Shao
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Jun Liu
- Department of Organ Transplantation Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Wanjia Xing
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Jin Xu
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Chengkun Qin
- Department of General Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Chunyou Li
- Department of Organ Transplantation Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Baoxiang Hu
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Shounan Yi
- Center for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Sydney, Australia
| | - Xuefeng Xia
- Genomic Medicine and Center for Diabetes Research, The Methodist Hospital Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA
| | - Haiqing Zhang
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Xiujuan Zhang
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Tingting Wang
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Wenfei Pan
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Chunxiao Yu
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Qiangxiu Wang
- Department of Pathology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Xiaoyan Lin
- Department of Pathology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Laicheng Wang
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Ling Gao
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China.
| | - Jiajun Zhao
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China.
| |
Collapse
|
42
|
The Role of Food Peptides in Lipid Metabolism during Dyslipidemia and Associated Health Conditions. Int J Mol Sci 2015; 16:9303-13. [PMID: 25918936 PMCID: PMC4463589 DOI: 10.3390/ijms16059303] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 04/10/2015] [Accepted: 04/20/2015] [Indexed: 01/17/2023] Open
Abstract
Animal and human clinical studies have demonstrated the ability of dietary food proteins to modulate endogenous lipid levels during abnormal lipid metabolism (dyslipidemia). Considering the susceptibility of proteins to gastric proteolytic activities, the hypolipidemic functions of proteins are possibly due, in part, to their peptide fragments. Food-derived peptides may directly modulate abnormal lipid metabolism in cell cultures and animal models of dyslipidemia. The peptides are thought to act by perturbing intestinal absorption of dietary cholesterol and enterohepatic bile acid circulation, and by inhibiting lipogenic enzymatic activities and gene expression in hepatocytes and adipocytes. Recent evidence indicates that the hypolipidemic activities of some peptides are due to activation of hepatic lipogenic transcription factors. However, detailed molecular mechanisms and structural requirements of peptides for these activities are yet to be elucidated. As hypolipidemic peptides can be released during enzymatic food processing, future studies can explore the prospects of combating metabolic syndrome and associated complications using peptide-rich functional food and nutraceutical products.
Collapse
|
43
|
Clausen MR, Zhang X, Yde CC, Ditlev DB, Lillefosse HH, Madsen L, Kristiansen K, Liaset B, Bertram HC. Intake of hydrolyzed casein is associated with reduced body fat accretion and enhanced phase II metabolism in obesity prone C57BL/6J mice. PLoS One 2015; 10:e0118895. [PMID: 25738501 PMCID: PMC4349863 DOI: 10.1371/journal.pone.0118895] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 01/08/2015] [Indexed: 01/12/2023] Open
Abstract
The amount and form of dietary casein have been shown to affect energy metabolism and lipid accumulation in mice, but the underlying mechanisms are not fully understood. We investigated 48 hrs urinary metabolome, hepatic lipid composition and gene expression in male C57BL/6J mice fed Western diets with 16 or 32 energy% protein in the form of extensively hydrolyzed or intact casein. LC-MS based metabolomics revealed a very strong impact of casein form on the urinary metabolome. Evaluation of the discriminatory metabolites using tandem mass spectrometry indicated that intake of extensively hydrolyzed casein modulated Phase II metabolism associated with an elevated urinary excretion of glucuronic acid- and sulphate conjugated molecules, whereas glycine conjugated molecules were more abundant in urine from mice fed the intact casein diets. Despite the differences in the urinary metabolome, we observed no differences in hepatic expression of genes involved in Phase II metabolism, but it was observed that expression of Abcc3 encoding ATP binding cassette c3 (transporter of glucuronic acid conjugates) was increased in livers of mice fed hydrolyzed casein. As glucuronic acid is derived from glucose and sulphate is derived from cysteine, our metabolomic data provided evidence for changes in carbohydrate and amino acid metabolism and we propose that this modulation of metabolism was associated with the reduced glucose and lipid levels observed in mice fed the extensively hydrolyzed casein diets.
Collapse
Affiliation(s)
| | - Xumin Zhang
- Department of Food Science, Aarhus University, Aarslev, Denmark
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | | | - Ditte B. Ditlev
- Department of Food Science, Aarhus University, Aarslev, Denmark
| | - Haldis H. Lillefosse
- National Institute of Nutrition and Seafood Research, Bergen, Norway
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Lise Madsen
- National Institute of Nutrition and Seafood Research, Bergen, Norway
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Bjørn Liaset
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | | |
Collapse
|
44
|
Lonardo A, Ballestri S, Marchesini G, Angulo P, Loria P. Nonalcoholic fatty liver disease: a precursor of the metabolic syndrome. Dig Liver Dis 2015; 47:181-90. [PMID: 25739820 DOI: 10.1016/j.dld.2014.09.020] [Citation(s) in RCA: 483] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 09/19/2014] [Accepted: 09/21/2014] [Indexed: 02/07/2023]
Abstract
The conventional paradigm of nonalcoholic fatty liver disease representing the "hepatic manifestation of the metabolic syndrome" is outdated. We identified and summarized longitudinal studies that, supporting the association of nonalcoholic fatty liver disease with either type 2 diabetes mellitus or metabolic syndrome, suggest that nonalcoholic fatty liver disease precedes the development of both conditions. Online Medical databases were searched, relevant articles were identified, their references were further assessed and tabulated data were checked. Although several cross-sectional studies linked nonalcoholic fatty liver disease to either diabetes and other components of the metabolic syndrome, we focused on 28 longitudinal studies which provided evidence for nonalcoholic fatty liver disease as a risk factor for the future development of diabetes. Moreover, additional 19 longitudinal reported that nonalcoholic fatty liver disease precedes and is a risk factor for the future development of the metabolic syndrome. Finally, molecular and genetic studies are discussed supporting the view that aetiology of steatosis and lipid intra-hepatocytic compartmentation are a major determinant of whether fatty liver is/is not associated with insulin resistance and metabolic syndrome. Data support the novel paradigm of nonalcoholic fatty liver disease as a strong determinant for the development of the metabolic syndrome, which has potentially relevant clinical implications for diagnosing, preventing and treating metabolic syndrome.
Collapse
Affiliation(s)
- Amedeo Lonardo
- AUSL Modena and University of Modena and Reggio Emilia, Department of Biomedical, Metabolic and Neural Sciences, Division of Internal Medicine, NOCSAE - Baggiovara, Modena, Italy.
| | - Stefano Ballestri
- AUSL Modena, Department of Internal Medicine, Division of Internal Medicine, Hospital of Pavullo, Pavullo nel Frignano, Italy
| | - Giulio Marchesini
- "Alma Mater Studiorum" University, Unit of Metabolic Diseases and Clinical Dietetics, Bologna, Italy
| | - Paul Angulo
- University of Kentucky, Division of Digestive Diseases & Nutrition, Section of Hepatology, Medical Center, Lexington, KY, USA
| | - Paola Loria
- AUSL Modena and University of Modena and Reggio Emilia, Department of Biomedical, Metabolic and Neural Sciences, Division of Internal Medicine, NOCSAE - Baggiovara, Modena, Italy
| |
Collapse
|
45
|
Tastesen HS, Rønnevik AK, Borkowski K, Madsen L, Kristiansen K, Liaset B. A mixture of cod and scallop protein reduces adiposity and improves glucose tolerance in high-fat fed male C57BL/6J mice. PLoS One 2014; 9:e112859. [PMID: 25390887 PMCID: PMC4229262 DOI: 10.1371/journal.pone.0112859] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/15/2014] [Indexed: 02/02/2023] Open
Abstract
Low-protein and high-protein diets regulate energy metabolism in animals and humans. To evaluate whether different dietary protein sources modulate energy balance when ingested at average levels obesity-prone male C57BL/6J mice were pair-fed high-fat diets (67 energy percent fat, 18 energy percent sucrose and 15 energy percent protein) with either casein, chicken filet or a mixture of cod and scallop (1∶1 on amino acid content) as protein sources. At equal energy intake, casein and cod/scallop fed mice had lower feed efficiency than chicken fed mice, which translated into reduced adipose tissue masses after seven weeks of feeding. Chicken fed mice had elevated hepatic triglyceride relative to casein and cod/scallop fed mice and elevated 4 h fasted plasma cholesterol concentrations compared to low-fat and casein fed mice. In casein fed mice the reduced adiposity was likely related to the observed three percent lower apparent fat digestibility compared to low-fat, chicken and cod/scallop fed mice. After six weeks of feeding an oral glucose tolerance test revealed that despite their lean phenotype, casein fed mice had reduced glucose tolerance compared to low-fat, chicken and cod/scallop fed mice. In a separate set of mice, effects on metabolism were evaluated by indirect calorimetry before onset of diet-induced obesity. Spontaneous locomotor activity decreased in casein and chicken fed mice when shifting from low-fat to high-fat diets, but cod/scallop feeding tended (P = 0.06) to attenuate this decrease. Moreover, at this shift, energy expenditure decreased in all groups, but was decreased to a greater extent in casein fed than in cod/scallop fed mice, indicating that protein sources regulated energy expenditure differently. In conclusion, protein from different sources modulates energy balance in C57BL/6J mice when given at normal levels. Ingestion of a cod/scallop-mixture prevented diet-induced obesity compared to intake of chicken filet and preserved glucose tolerance compared to casein intake.
Collapse
Affiliation(s)
- Hanne Sørup Tastesen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Alexander Krokedal Rønnevik
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Kamil Borkowski
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Lise Madsen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Karsten Kristiansen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- * E-mail: (KK); (BL)
| | - Bjørn Liaset
- National Institute of Nutrition and Seafood Research, Bergen, Norway
- * E-mail: (KK); (BL)
| |
Collapse
|
46
|
Bile acids, obesity, and the metabolic syndrome. Best Pract Res Clin Gastroenterol 2014; 28:573-83. [PMID: 25194176 PMCID: PMC4159616 DOI: 10.1016/j.bpg.2014.07.004] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 06/30/2014] [Accepted: 07/05/2014] [Indexed: 01/31/2023]
Abstract
Bile acids are increasingly recognized as key regulators of systemic metabolism. While bile acids have long been known to play important and direct roles in nutrient absorption, bile acids also serve as signalling molecules. Bile acid interactions with the nuclear hormone receptor farnesoid X receptor (FXR) and the membrane receptor G-protein-coupled bile acid receptor 5 (TGR5) can regulate incretin hormone and fibroblast growth factor 19 (FGF19) secretion, cholesterol metabolism, and systemic energy expenditure. Bile acid levels and distribution are altered in type 2 diabetes and increased following bariatric procedures, in parallel with reduced body weight and improved insulin sensitivity and glycaemic control. Thus, modulation of bile acid levels and signalling, using bile acid binding resins, TGR5 agonists, and FXR agonists, may serve as a potent therapeutic approach for the treatment of obesity, type 2 diabetes, and other components of the metabolic syndrome in humans.
Collapse
|
47
|
Fjære E, Aune UL, Røen K, Keenan AH, Ma T, Borkowski K, Kristensen DM, Novotny GW, Mandrup-Poulsen T, Hudson BD, Milligan G, Xi Y, Newman JW, Haj FG, Liaset B, Kristiansen K, Madsen L. Indomethacin treatment prevents high fat diet-induced obesity and insulin resistance but not glucose intolerance in C57BL/6J mice. J Biol Chem 2014; 289:16032-45. [PMID: 24742673 DOI: 10.1074/jbc.m113.525220] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Chronic low grade inflammation is closely linked to obesity-associated insulin resistance. To examine how administration of the anti-inflammatory compound indomethacin, a general cyclooxygenase inhibitor, affected obesity development and insulin sensitivity, we fed obesity-prone male C57BL/6J mice a high fat/high sucrose (HF/HS) diet or a regular diet supplemented or not with indomethacin (±INDO) for 7 weeks. Development of obesity, insulin resistance, and glucose intolerance was monitored, and the effect of indomethacin on glucose-stimulated insulin secretion (GSIS) was measured in vivo and in vitro using MIN6 β-cells. We found that supplementation with indomethacin prevented HF/HS-induced obesity and diet-induced changes in systemic insulin sensitivity. Thus, HF/HS+INDO-fed mice remained insulin-sensitive. However, mice fed HF/HS+INDO exhibited pronounced glucose intolerance. Hepatic glucose output was significantly increased. Indomethacin had no effect on adipose tissue mass, glucose tolerance, or GSIS when included in a regular diet. Indomethacin administration to obese mice did not reduce adipose tissue mass, and the compensatory increase in GSIS observed in obese mice was not affected by treatment with indomethacin. We demonstrate that indomethacin did not inhibit GSIS per se, but activation of GPR40 in the presence of indomethacin inhibited glucose-dependent insulin secretion in MIN6 cells. We conclude that constitutive high hepatic glucose output combined with impaired GSIS in response to activation of GPR40-dependent signaling in the HF/HS+INDO-fed mice contributed to the impaired glucose clearance during a glucose challenge and that the resulting lower levels of plasma insulin prevented the obesogenic action of the HF/HS diet.
Collapse
Affiliation(s)
- Even Fjære
- From the Department of Biology, University of Copenhagen, 1165 Copenhagen, Denmark, the National Institute of Nutrition and Seafood Research, 5817 Bergen, Norway
| | - Ulrike L Aune
- From the Department of Biology, University of Copenhagen, 1165 Copenhagen, Denmark, the National Institute of Nutrition and Seafood Research, 5817 Bergen, Norway
| | - Kristin Røen
- From the Department of Biology, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Alison H Keenan
- From the Department of Biology, University of Copenhagen, 1165 Copenhagen, Denmark, the Departments of Nutrition and
| | - Tao Ma
- From the Department of Biology, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Kamil Borkowski
- From the Department of Biology, University of Copenhagen, 1165 Copenhagen, Denmark
| | - David M Kristensen
- the INSERM U1085-IRSET, Université de Rennes 1, Rennes, France, the Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Guy W Novotny
- the Section for Endocrinological Research, Department of Biomedical Sciences, University of 2200 Copenhagen, Copenhagen, Denmark
| | - Thomas Mandrup-Poulsen
- the Section for Endocrinological Research, Department of Biomedical Sciences, University of 2200 Copenhagen, Copenhagen, Denmark, the Department of Molecular Medicine and Surgery, Karolinska Institute, 171 77 Solna, Sweden
| | - Brian D Hudson
- the Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom, and
| | - Graeme Milligan
- the Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom, and
| | | | - John W Newman
- the Departments of Nutrition and the United States Department of Agriculture-Agricultural Research Service-Western Human Nutrition Research Center, Davis, California 95616
| | - Fawaz G Haj
- the Departments of Nutrition and Internal Medicine, University of California, Davis, California 95616
| | - Bjørn Liaset
- the National Institute of Nutrition and Seafood Research, 5817 Bergen, Norway
| | - Karsten Kristiansen
- From the Department of Biology, University of Copenhagen, 1165 Copenhagen, Denmark,
| | - Lise Madsen
- From the Department of Biology, University of Copenhagen, 1165 Copenhagen, Denmark, the National Institute of Nutrition and Seafood Research, 5817 Bergen, Norway,
| |
Collapse
|
48
|
Lillefosse HH, Clausen MR, Yde CC, Ditlev DB, Zhang X, Du ZY, Bertram HC, Madsen L, Kristiansen K, Liaset B. Urinary loss of tricarboxylic acid cycle intermediates as revealed by metabolomics studies: an underlying mechanism to reduce lipid accretion by whey protein ingestion? J Proteome Res 2014; 13:2560-70. [PMID: 24702026 PMCID: PMC4045150 DOI: 10.1021/pr500039t] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
![]()
Whey protein intake is associated
with the modulation of energy
metabolism and altered body composition both in human subjects and
in animals, but the underlying mechanisms are not yet elucidated.
We fed obesity-prone C57BL/6J mice high-fat diets with either casein
(HF casein) or whey (HF whey) for 6 weeks. At equal energy intake
and apparent fat and nitrogen digestibility, mice fed HF whey stored
less energy as lipids, evident both as lower white adipose tissue
mass and as reduced liver lipids, compared with HF-casein-fed mice.
Explorative analyses of 48 h urine, both by 1H NMR and
LC–MS metabolomic platforms, demonstrated higher urinary excretion
of tricarboxylic acid (TCA) cycle intermediates citric acid and succinic
acid (identified by both platforms), and cis-aconitic
acid and isocitric acid (identified by LC–MS platform) in the
HF whey, relative to in the HF-casein-fed mice. Targeted LC–MS
analyses revealed higher citric acid and cis-aconitic acid concentrations
in fed state plasma, but not in liver of HF-whey-fed mice. We propose
that enhanced urinary loss of TCA cycle metabolites drain available
substrates for anabolic processes, such as lipogenesis, thereby leading
to reduced lipid accretion in HF-whey-fed compared to HF-casein-fed
mice.
Collapse
Affiliation(s)
- Haldis H Lillefosse
- Department of Biology, University of Copenhagen , Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hanai M, Esashi T. Comparison of the effects of dietary protein on the sexual organ development of male mice and rats kept under constant darkness. J Nutr Sci Vitaminol (Tokyo) 2014; 59:552-9. [PMID: 24477253 DOI: 10.3177/jnsv.59.552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The purpose of this study was to determine whether the effects of dietary protein on sexual organ development were different in mice and rats kept under constant darkness. Four-week-old mice (ICR strain) and rats (F344 strain) were kept under constant darkness (D) or normal lighting (N; 12-h light/dark cycle) for 4 wk. The dietary protein level was 9% casein with the addition of 0.135% cystine (9PC) or without it (9P); other components of the diet were based on the AIN-93G diet. The testes and epididymides weights (g/100 g BW) of the rats given the 9P diet in the D-group were lower than those of the rats given the 9P diet in the N-group. In the mice, lighting conditions and diet did not affect testes or epididymides weights. Body weight and food intake in the rats were affected by diet, and these values were lower in the 9P diet group; however, body weight and food intake in the mice was not affected by diet. The serum albumin concentration in the rats was lower in the 9P diet group, while that of the mice was lower in the 9PC diet group. In the rats kept under constant darkness, a diet lacking in cystine accelerated the suppression of sexual organ development and decreased serum albumin concentration, but this diet had no such effects on the mice. The finding that the effects of dietary protein were different in mice and rats suggests that protein requirements of mice are different from those of rats.
Collapse
Affiliation(s)
- Miho Hanai
- Department of Nutrition and Life Science, Kanagawa Institute of Technology
| | | |
Collapse
|
50
|
Bjørndal B, Berge C, Ramsvik MS, Svardal A, Bohov P, Skorve J, Berge RK. A fish protein hydrolysate alters fatty acid composition in liver and adipose tissue and increases plasma carnitine levels in a mouse model of chronic inflammation. Lipids Health Dis 2013; 12:143. [PMID: 24098955 PMCID: PMC4021737 DOI: 10.1186/1476-511x-12-143] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/24/2013] [Indexed: 01/17/2023] Open
Abstract
Background There is growing evidence that fish protein hydrolysate (FPH) diets affect mitochondrial fatty acid metabolism in animals. The aim of the study was to determine if FPH could influence fatty acid metabolism and inflammation in transgene mice expressing human tumor necrosis factor alpha (hTNFα). Methods hTNFα mice (C57BL/6 hTNFα) were given a high-fat (23%, w/w) diet containing 20% casein (control group) or 15% FPH and 5% casein (FPH group) for two weeks. After an overnight fast, blood, adipose tissue, and liver samples were collected. Gene expression and enzyme activity was analysed in liver, fatty acid composition was analyzed in liver and ovarian white adipose tissue, and inflammatory parameters, carnitine, and acylcarnitines were analyzed in plasma. Results The n-3/n-6 fatty acid ratio was higher in mice fed the FPH diet than in mice fed the control diet in both adipose tissue and liver, and the FPH diet affected the gene expression of ∆6 and ∆9 desaturases. Mice fed this diet also demonstrated lower hepatic activity of fatty acid synthase. Concomitantly, a lower plasma INF-γ level was observed. Plasma carnitine and the carnitine precursor γ-butyrobetaine was higher in the FPH-group compared to control, as was plasma short-chained and medium-chained acylcarnitine esters. The higher level of plasma acetylcarnitine may reflect a stimulated mitochondrial and peroxisomal β-oxidation of fatty acids, as the hepatic activities of peroxisomal acyl-CoA oxidase 1 and mitochondrial carnitine palmitoyltransferase-II were higher in the FPH-fed mice. Conclusions The FPH diet was shown to influence hepatic fatty acid metabolism and fatty acid composition. This indicates that effects on fatty acid metabolism are important for the bioactivity of protein hydrolysates of marine origin.
Collapse
Affiliation(s)
- Bodil Bjørndal
- Institute of Clinical Science, University of Bergen, N-5020 Bergen, Norway.
| | | | | | | | | | | | | |
Collapse
|