1
|
Tang M, Wu Y, Olnood CG, Gao Y, Wang F, Zhang Z, Peng C, Zhou X, Huang C, Xiong X, Yin Y. Effects of peroxidized lipids on intestinal morphology, antioxidant capacity and gut microbiome in piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:430-443. [PMID: 40034456 PMCID: PMC11875184 DOI: 10.1016/j.aninu.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 03/05/2025]
Abstract
This study investigated the effect of peroxidized lipids on piglets' growth performance, intestinal morphology, inflammatory reactions, oxidative stress in the liver, duodenum, jejunum, ileum, and colon, and ileal microbiota. Twenty piglets (Duroc × [Landrace × Yorkshire]; age = 21 d old, BW = 6.5 ± 1 kg) were randomly assigned to two groups with 10 replicates per group and one piglet per replicate. The control group was fed 6% fresh soybean oil and the peroxidized soybean oil (PSO) group fed 6% PSO. The experimental feeding period lasted 24 d. The study found no impact on ADFI, ADG and gain to feed ratio (P > 0.05). However, the PSO group increased the diarrhea index and the serum levels of lactate dehydrogenase triglycerides, cholesterol, low-density lipoprotein cholesterol, and high-density lipoprotein cholesterol (P < 0.05), along with decreased concentrations of alanine aminotransferase and blood urea nitrogen (P < 0.05). For oxidative enzymes, PSO increased the concentration of F2-isoprostane in urine (P = 0.032), malondialdehyde (MDA) in the duodenum (P = 0.001) and jejunum (P = 0.004), decreased thiobarbituric acid reactive substances (TBARS) in the liver (P = 0.001) but increased TBARS in duodenum (P = 0.001), and carbonylated proteins in the duodenum (P = 0.003). For antioxidant enzymes, PSO decreased superoxide dismutase (SOD) in the liver (P = 0.001), colon (P = 0.002), and jejunum (P = 0.015), along with glutathione peroxidase (GSH-Px) in the liver (P = 0.008) and NAD(P)H:quinone oxidoreductase 1 (NQO1) in ileum (P = 0.001). For inflammatory reactions, PSO increased interleukin (IL)-1β concentrations in the duodenum and colon, and IL-10 in the jejunum, while decreasing IL-4 concentration in the duodenum (P < 0.05). For intestinal morphology and ileal microbiota, PSO increased ileal crypt depth, while decreasing the crypt-to-villus ratio (P < 0.05). Peroxidized soybean oil increased the relative abundance of Prevotella, Clostridium_sensu_stricto_1, Clostridium_sensu_stricto_6, Pasteurella and Klebsiella (P < 0.05). In conclusion, this study revealed that PSO worsened diarrhea, increasing the ileal crypt depth and the relative abundance of harmful microbiota, and induced oxidative stress and inflammation in the intestines and liver, primarily in the jejunum and ileum.
Collapse
Affiliation(s)
- Mengxuan Tang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yuliang Wu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| | | | - Yundi Gao
- Sichuan Synlight Biotech Ltd., Chengdu 610041, China
| | - Fei Wang
- Sichuan Synlight Biotech Ltd., Chengdu 610041, China
| | - Zicheng Zhang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Can Peng
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| | - Xihong Zhou
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| | - Chunxia Huang
- School of Stomatology, Changsha Medical University, Changsha 410219, China
| | - Xia Xiong
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
- School of Stomatology, Changsha Medical University, Changsha 410219, China
| | - Yulong Yin
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| |
Collapse
|
2
|
Xu K, Zhang B, He Y, Wang Y, Liu Y, Shi G, Shen Y, Chen F, Mi B, Shi L, Zeng L, Liu X, Dang S, Yan H. Serum Lipidomic Signatures Mediate the Association Between Coarse Grain Preference and Central Obesity in Adults With Normal Weight and High Wheat Intake. Mol Nutr Food Res 2024:e202400515. [PMID: 39692176 DOI: 10.1002/mnfr.202400515] [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: 07/13/2024] [Revised: 10/11/2024] [Accepted: 11/29/2024] [Indexed: 12/19/2024]
Abstract
Little is known about the association between grain preference andabdominal fat accumulation, and mediating roles of circulating lipidomicsignatures. We quantified 1245 circulating lipids in 150 normal-weight centralobesity (NWCO) cases and 150 controls using targeted lipidomics. Grainpreference was determined by the highest intake frequency of grains (whiterice, wheat, or coarse grain). In our participants with high wheat intakefrequency, preferring coarse grain over rice was associated with a 60% lowerrisk of NWCO. Of the 585 lipids showing opposing associations with white riceand coarse grains, 46 were significantly linked to either (p < 0.05), predominantly alkylacyl phospholipids (PE-Os; n < 9) and alkenylacylphospholipids (PE-Ps; nx = 7). Network analysis identified a module primarilycomposed of PE-Os and PE-Ps, which was positively associated with coarse grain (p = 0.014). Another module, mainly consisting of triacylglycerols (TGs), was associatedwith white rice (p = 0.003) and mediated the association between white rice(mediation proportion: 20.30%; p = 0.027) or coarse grain preference (11.43%; p = 0.040) and NWCO. Specific lipids, such as TG(8:0_16:0_16:0) and TG(8:0_14:0_18:0), exhibited notable mediation effects. In normal-weight individuals with highwheat intake frequency, preferring coarse grain was inversely associated with NWCO, mediated by specific lipidomic signatures.
Collapse
Affiliation(s)
- Kun Xu
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Binyan Zhang
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Yifei He
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Yutong Wang
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Yezhou Liu
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Guoshuai Shi
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Yuan Shen
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Fangyao Chen
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Baibing Mi
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi' an, Shaanxi, China
| | - Lingxia Zeng
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Xin Liu
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shaonong Dang
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Hong Yan
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Nutrition and Food Safety Engineering Research Center of Shaanxi Province, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
3
|
Macri EV, Touceda V, Wiszniewski M, Cacciagiú LD, Zago V, Puntarulo S, Pellegrino N, Lifshitz F, Friedman SM, Miksztowicz V. Liver response to the consumption of fried sunflower oil. J Nutr Biochem 2024; 134:109734. [PMID: 39117077 DOI: 10.1016/j.jnutbio.2024.109734] [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: 05/28/2024] [Revised: 07/16/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Sunflower oil is one of the most commonly used fat sources in Argentina, and deep-fat frying is the popular food preparation process. The liver response of feeding a diet containing fried sunflower oil (SFOx) on growing rats was studied. Thirty-nine male weanling Wistar rats were randomly assigned to one of three diets for 8 wks: control (C), sunflower oil (SFO), and a diet containing SFOx, both of the sunflower diets were mixed with a commercial rat chow at weight ratio of 13% (w/w). Body weight and food consumption were recorded weekly. At t=8 wk, lipid profile and glycemia were measured. Visceral adiposity was registered. Liver was weighed and preserved for histological analysis, relative fatty acid profile, fibrosis markers and oxidative status. The three diets did not alter body weights; however, the SFOx fed rats showed increased energy intake and visceral fat; therefore, in liver saturated fat content, trans fatty acids, plus other unidentified minor components, such as hydroperoxides, hydroxides, epidioxides, hydroperoxy epidioxides, hydroxylepidioxides, and epoxides, were detected. The hepatosomatic index of SFOx rats was altered and showed hepatic steatosis. SFOx rats exhibited increased liver dichlorodihydrofluorescein-diacetate and thiobarbituric acid substance levels and oxidized-proteins content. Their livers had lower relative levels of monounsaturated, polyunsaturated fatty acids and catalase activity, but matrix metalloproteinase-9 activity was unchanged. Consumption of a diet rich in fried oil during growth could induce liver damage due to steatosis, excessive lipid toxicity and the accumulation of reactive oxygen species. Further progression could lead to hepatic fibrosis.
Collapse
Affiliation(s)
- Elisa V Macri
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Bioquímica General y Bucal, Buenos Aires, Argentina
| | - Vanessa Touceda
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Bioquímica General y Bucal, Buenos Aires, Argentina; Pontificia Universidad Católica Argentina, Facultad de Medicina, Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Buenos Aires, Argentina
| | - Morena Wiszniewski
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Bioquímica General y Bucal, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Laboratorio de Endocrinología Molecular (LEM), Buenos Aires, Argentina
| | - Leonardo D Cacciagiú
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Bioquímica General y Bucal, Buenos Aires, Argentina; Hospital General de Agudos Teodoro Álvarez. Laboratorio Central, Sección Bioquímica, Buenos Aires, Argentina
| | - Valeria Zago
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Laboratorio de Lípidos y Aterosclerosis, Hospital de Clínicas. INFIBIOC-UBA, Buenos Aires, Argentina
| | - Susana Puntarulo
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Fisicoquímica, Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)- Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, Argentina
| | - Néstor Pellegrino
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Bromatología, Buenos Aires, Argentina
| | - Fima Lifshitz
- Honorary Professor, State University of New York, Downstate Medical Center, College of Medicine, Brooklyn, Santa Barbara, CA, USA
| | - Silvia M Friedman
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Bioquímica General y Bucal, Buenos Aires, Argentina
| | - Verónica Miksztowicz
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Bioquímica General y Bucal, Buenos Aires, Argentina; Pontificia Universidad Católica Argentina, Facultad de Medicina, Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Buenos Aires, Argentina.
| |
Collapse
|
4
|
Zhang G, Huang J, Sun Z, Guo Y, Lin G, Zhang Z, Zhao J. Effects of Trace Mineral Source on Growth Performance, Antioxidant Activity, and Meat Quality of Pigs Fed an Oxidized Soy Oil Supplemented Diet. Antioxidants (Basel) 2024; 13:1227. [PMID: 39456480 PMCID: PMC11505604 DOI: 10.3390/antiox13101227] [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: 09/08/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigates the effects of oil quality and trace mineral source on the growth performance, antioxidant activity, and meat quality of growing-finishing pigs. A total of 180 crossbred pigs (Duroc × Landrace × Large White [64.4 ± 1.95]) were randomly allocated five dietary treatments based on body weight (BW) and sex in a 30 d trial. Pigs were fed five diets: (i) fresh soy oil + inorganic trace minerals (ITMs) + inorganic selenium (FISI), (ii) oxidized soy oil + ITMs + inorganic selenium (OISI), (iii) fresh soy oil + ITMs + selenium yeast (FISY), (iv) oxidized soy oil + ITMs + selenium yeast (OISY), and (v) oxidized soy oil + organic trace minerals (OTMs) + selenium yeast (OOSY). Each dietary treatment included six replicates and six pigs per replicate (three barrows and three gilts). Feeding OISI resulted in lower average daily gain (ADG) and dressing percentage (p < 0.05). The OOSY group had a higher dressing percentage and activities of serum CAT and GSH-Px in growing-finishing pigs (p < 0.05). In addition, the relative abundance of Campylobacterota in the colonic digesta varied with the quality of soy oil and source of trace minerals (p < 0.05), but no significant differences in short-chain fatty acid concentrations were observed among all dietary groups. In conclusion, adding oxidized soy oil to the diet negatively impacted the ADG and dressing percentage of growing-finishing pigs, and replacing ITMs with OTMs and SY alleviated these negative impacts. A combination of OTMs and SY can support antioxidant capacity to mitigate the negative impacts of oxidized oil on the growth performance and dressing percentage of growing-finishing pigs.
Collapse
Affiliation(s)
- Ge Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (G.Z.); (J.H.); (Z.S.); (Z.Z.)
| | - Jingyi Huang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (G.Z.); (J.H.); (Z.S.); (Z.Z.)
| | - Zhiqiang Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (G.Z.); (J.H.); (Z.S.); (Z.Z.)
| | - Yuhan Guo
- Beijing Alltech Biological Products (China) Co., Ltd., Beijing 100600, China; (Y.G.); (G.L.)
| | - Gang Lin
- Beijing Alltech Biological Products (China) Co., Ltd., Beijing 100600, China; (Y.G.); (G.L.)
| | - Zeyu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (G.Z.); (J.H.); (Z.S.); (Z.Z.)
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (G.Z.); (J.H.); (Z.S.); (Z.Z.)
| |
Collapse
|
5
|
Hu L, Huang L, Fang Z, Wang C, Luo J, Deng Q, Xu D, Sun L, Gooneratne R. Fried Soybean Oil Causes Systemic Low-Grade Inflammation by Disrupting the Balance of Gut Microbiota in Mice. Microorganisms 2024; 12:1210. [PMID: 38930592 PMCID: PMC11205791 DOI: 10.3390/microorganisms12061210] [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: 05/07/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Previous reports have mainly investigated the long-term effects (>30 d), such as gut microbiota dysbiosis and systemic low-grade inflammation, in mice fed fried oil. However, short-term intake of deep-fried oil is more likely to occur in daily life, and such studies are lacking. This study aimed to investigate the short-term effects of fried oil intake on systemic low-grade inflammation. Male Kunming mice were fed non-fried soybean oil or low (25%), medium (50%), or high (100%)-fried oil at 4.4 g/kg for 6 d. Serum and fecal samples were collected on day 7. In all groups fed fried oil, the serum levels of tumor necrosis factor (TNF-α) were significantly elevated 2-4-fold. Among the gut microbiota, the abundance of Alloprevotella significantly decreased by up to 76%, while Lactobacilli significantly increased by up to 385%. The fecal valeric acid content was significantly increased and positively correlated with TNF-α levels. Both valeric acid and TNF-α levels were positively correlated with the abundance of Lactobacilli and negatively correlated with that of Alloprevotella. In summary, a short-term ingestion of even low doses of fried oil alters the gut microbiota Alloprevotella and Lactobacilli and increases fecal valeric acid content, which correlates with increased serum TNF-α levels.
Collapse
Affiliation(s)
- Lianhua Hu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (L.H.); (L.H.); (C.W.); (J.L.); (Q.D.); (D.X.); (L.S.)
| | - Ling Huang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (L.H.); (L.H.); (C.W.); (J.L.); (Q.D.); (D.X.); (L.S.)
| | - Zhijia Fang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (L.H.); (L.H.); (C.W.); (J.L.); (Q.D.); (D.X.); (L.S.)
| | - Chen Wang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (L.H.); (L.H.); (C.W.); (J.L.); (Q.D.); (D.X.); (L.S.)
| | - Jinjin Luo
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (L.H.); (L.H.); (C.W.); (J.L.); (Q.D.); (D.X.); (L.S.)
| | - Qi Deng
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (L.H.); (L.H.); (C.W.); (J.L.); (Q.D.); (D.X.); (L.S.)
| | - Defeng Xu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (L.H.); (L.H.); (C.W.); (J.L.); (Q.D.); (D.X.); (L.S.)
| | - Lijun Sun
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (L.H.); (L.H.); (C.W.); (J.L.); (Q.D.); (D.X.); (L.S.)
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, P.O. Box 85084, Lincoln 7647, New Zealand;
| |
Collapse
|
6
|
Guo X, Ren H, Sun P, Ding E, Fang J, Fang K, Ma X, Li C, Li C, Xu Y, Cao K, Lin EZ, Guo P, Pollitt KJG, Tong S, Tang S, Shi X. Personal exposure to airborne organic pollutants and lung function changes among healthy older adults. ENVIRONMENTAL RESEARCH 2024; 258:119411. [PMID: 38876423 DOI: 10.1016/j.envres.2024.119411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Epidemiological evidence on the impact of airborne organic pollutants on lung function among the elderly is limited, and their underlying biological mechanisms remain largely unexplored. Herein, a longitudinal panel study was conducted in Jinan, Shandong Province, China, involving 76 healthy older adults monitored over a span of five months repetitively. We systematically evaluated personal exposure to a diverse range of airborne organic pollutants using a wearable passive sampler and their effects on lung function. Participants' pulmonary function indicators were assessed, complemented by comprehensive multi-omics analyses of blood and urine samples. Leveraging the power of interaction analysis, causal inference test (CIT), and integrative pathway analysis (IPA), we explored intricate relationships between specific organic pollutants, biomolecules, and lung function deterioration, elucidating the biological mechanisms underpinning the adverse impacts of these pollutants. We observed that bis (2-chloro-1-methylethyl) ether (BCIE) was significantly associated with negative changes in the forced vital capacity (FVC), with glycerolipids mitigating this adverse effect. Additionally, 31 canonical pathways [e.g., high mobility group box 1 (HMGB1) signaling, phosphatidylinositol 3-kinase (PI3K)/AKT pathway, epithelial mesenchymal transition, and heme and nicotinamide adenine dinucleotide (NAD) biosynthesis] were identified as potential mechanisms. These findings may hold significant implications for developing effective strategies to prevent and mitigate respiratory health risks arising from exposure to such airborne pollutants. However, due to certain limitations of the study, our results should be interpreted with caution.
Collapse
Affiliation(s)
- Xiaojie Guo
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Huimin Ren
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, China Medical University, Shenyang, Liaoning 110001, China
| | - Peijie Sun
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, China Medical University, Shenyang, Liaoning 110001, China
| | - Enmin Ding
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianlong Fang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Ke Fang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Xiao Ma
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, Shandong University, Jinan, Shandong 250100, China
| | - Chenfeng Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Chenlong Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, Shandong University, Jinan, Shandong 250100, China
| | - Yibo Xu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, China Medical University, Shenyang, Liaoning 110001, China
| | - Kangning Cao
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Elizabeth Z Lin
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT 06510, USA
| | - Pengfei Guo
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT 06510, USA
| | - Krystal J Godri Pollitt
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT 06510, USA
| | - Shilu Tong
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; School of Public Health and Social Work, Queensland University of Technology, Brisbane 4001, Australia
| | - Song Tang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| | - Xiaoming Shi
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| |
Collapse
|
7
|
Balakrishnan J, Sugasini D, Shanmugam K. Long-term Supplementation of Deep-fried Oil Consumption Impairs Oxidative Stress, Colon Histology and Increases Neurodegeneration. Cell Biochem Biophys 2024; 82:1477-1488. [PMID: 38898335 DOI: 10.1007/s12013-024-01301-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Sesame oil and sunflower oil are popular cooking oils in southern India. Deep-frying is a frequent method of food preparation. Deep-frying at high temperatures has been linked with several disorders, including cancer, diabetes, and unknown metabolic problems. There have been no long-term investigations on the influence of deep-fried oils on PUFA metabolism and pathogenesis. As a result, the current study aimed to explore the effect of deep-fried frying oil on Wistar rats by continuous treatment. Furthermore, the pathophysiology of MSG-induced neurotoxicity in Wistar rats was investigated. METHODS Wistar rats weighing 200-260 g were used in this study. Female rats were divided into five groups fed with (1) standard chow (control group), (2) unheated sesame oil (UHSO) along with standard chow, and (3) reheated sesame oil (RHSO) along with standard chow, (4) unheated sunflower oil (UHSFO) along with standard chow, and (5) reheated sunflower oil (RHSFO) along with standard chow and continued up to F1 generation. Furthermore, F1 male rats were treated with MSG of 2 g/kg body weight for 10 alternative days and were sacrificed for major tissues. RESULTS We found that rats treated with RHSO and RHSFO showed increased body weight. Deep-fried oil-fed rats (RHSO and RHSFO) showed a significant increase in total cholesterol- 100 mg/dl, LDL- 23 mg/dl, & TAG-100 mg/dl, when compared to unheated oil rats. Liver function tests revealed that AST and ALT levels were significantly elevated in RHSO and RHSFO when compared to unheated oils and the control group. Inflammatory markers revealed that Hs-CRP (0.35 mg/dl) and LDH levels (6000 U/L) were significantly elevated in RHSO and RHSFO when compared to the unheated oils and control group. RT-PCR results showed significant elevation in the antioxidant genes SOD (twofold) and GPX (3-fold) when compared to UHSO and UHSFO groups. Liver and colon histology showed significant damage in the cell structure of RHSO and RHSFO-treated rats. Further, rats treated with unheated oils and MSG showed statistically significantly higher mRNA expression of neuroplasticity genes CREB, BDNF and reduced NMDA levels (UHSO, UHSFO) when compared to reheated oil groups (RHSO & RHSFO). Proinflammatory marker TNF-α expression was significantly elevated in RHSFO-treated rats when compared to control. Brain histology showed focal damage in glial cell degeneration in rats treated with RHSO and RHSFO when compared to other groups. CONCLUSION The results from the present study proved that continuous supplementation deep-fried reheated oil consumption increased serum TGL and oxidative stress markers. Impaired liver metabolism and the involvement of the gut-liver-brain axis increased the risk of neurodegeneration.
Collapse
Affiliation(s)
- Jeyakumar Balakrishnan
- Central Research Laboratory for Biomedical Research, Vinayaka Mission's Medical College and Hospital, Vinayaka Mission Research Foundation, Deemed to be University, Karaikal, 609602, Puducherry, India
- Department of Molecular Biology, School of Biological Sciences, Madurai Kamaraj, University, Madurai, 625021, Tamil Nadu, India
| | - Dhavamani Sugasini
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, Illinois, 60612, USA.
| | - Kathiresan Shanmugam
- Department of Biotechnology, School of Integrative Biology, Central University of Tamil Nadu, Neelakudi, Thiruvarur, 610 005, Tamil Nadu, India.
| |
Collapse
|
8
|
Fan X, Yu W, Wang Q, Yang H, Tan D, Yu B, He J, Zheng P, Yu J, Luo J, Luo Y, Yan H, Wang J, Wang H, Wang Q, Mao X. Protective effect of Broussonetia papyrifera leaf polysaccharides on intestinal integrity in a rat model of diet-induced oxidative stress. Int J Biol Macromol 2024; 268:131589. [PMID: 38643924 DOI: 10.1016/j.ijbiomac.2024.131589] [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: 08/09/2023] [Revised: 03/28/2024] [Accepted: 04/12/2024] [Indexed: 04/23/2024]
Abstract
This study aimed to investigate the effect of Broussonetia papyrifera polysaccharides (BPP) on the jejunal intestinal integrity of rats ingesting oxidized fish oil (OFO) induced oxidative stress. Polysaccharides (Mw 16,956 Da) containing carboxyl groups were extracted from Broussonetia papyrifera leaves. In vitro antioxidant assays showed that this polysaccharide possessed antioxidant capabilities. Thirty-two male weaned rats were allocated into two groups orally infused BPP solution and PBS for 26 days, respectively. From day 9 to day 26, half of the rats in each group were fed food containing OFO, where the lipid peroxidation can induce intestinal oxidative stress. OFO administration resulted in diarrhea, decreased growth performance (p < 0.01), impaired jejunal morphology (p < 0.05) and antioxidant capacity (p < 0.01), increased the levels of ROS and its related products, IL-1β and IL-17 (p < 0.01) of jejunum, as well as down-regulated Bcl-2/Bax (p < 0.01) and Nrf2 signaling (p < 0.01) of jejunum in rats. BPP gavage effectively alleviated the negative effects of OFO on growth performance, morphology, enterocyte apoptosis, antioxidant capacity and inflammation of jejunum (p < 0.05) in rats. In the oxidative stress model cell assay, the use of receptor inhibitors inhibited the enhancement of antioxidant capacity by BPP. These results suggested that BPP protected intestinal morphology, thus improving growth performance and reducing diarrhea in rats ingesting OFO. This protective effect may be attributed to scavenging free radicals and activating the Nrf2 pathway, which enhances antioxidant capacity, consequently reducing inflammation and mitigating intestinal cell death.
Collapse
Affiliation(s)
- Xiangqi Fan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Wei Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Qingxiang Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Heng Yang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Dayan Tan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Jianping Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Huifen Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Quyuan Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China.
| |
Collapse
|
9
|
Wu IT, Yeh WJ, Huang WC, Yang HY. Very low-carbohydrate diet with higher protein ratio improves lipid metabolism and inflammation in rats with diet-induced nonalcoholic fatty liver disease. J Nutr Biochem 2024; 126:109583. [PMID: 38244701 DOI: 10.1016/j.jnutbio.2024.109583] [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: 09/02/2023] [Revised: 11/27/2023] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is commonly associated with obesity, and it is mainly treated through lifestyle modifications. The very low-carbohydrate diet (VLCD) can help lose weight rapidly but the possible effects of extreme dietary patterns on lipid metabolism and inflammatory responses in individuals with NAFLD remain debatable. Moreover, VLCD protein content may affect its effectiveness in weight loss, steatosis, and inflammatory responses. Therefore, we investigated the effects of VLCDs with different protein contents in NAFLD rats and the mechanisms underlying these effects. After a 16-week inducing period, the rats received an isocaloric normal diet (NC group) or a VLCD with high or low protein content (NVLH vs. NVLL group, energy ratio:protein/carbohydrate/lipid=20/1/79 vs. 6/1/93) for the next 8 weeks experimental period. We noted that the body weight decreased in both the NVLH and NVLL groups; nevertheless, the NVLH group demonstrated improvements in ketosis. The NVLL group led to hepatic lipid accumulation, possibly by increasing very-low-density lipoprotein receptor (VLDLR) expression and elevating liver oxidative stress, subsequently activating the expression of Nrf2, and inflammation through the TLR4/TRIF/NLRP3 and TLR4/MyD88/NF-κB pathway. The NVLH was noted to prevent the changes in VLDLR and the TLR4-inflammasome pathway partially. The VLCD also reduced the diversity of gut microbiota and changed their composition. In conclusion, although low-protein VLCD consumption reduces BW, it may also lead to metabolic disorders and changes in microbiota composition; nevertheless, a VLCD with high protein content may partially alleviate these limitations.
Collapse
Affiliation(s)
- I-Ting Wu
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wan-Ju Yeh
- Graduate Program of Nutrition Science, National Taiwan Normal University, Taipei, Taiwan
| | - Wen-Chih Huang
- Department of Anatomical Pathology, Taipei Institute of Pathology, Taipei City, Taiwan
| | - Hsin-Yi Yang
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
10
|
Huang HC, Lee PN, Huang WC, Yang HY. Partial Replacement of Diet with Dehulled Adlay Ameliorates Hepatic Steatosis, Inflammation, Oxidative Stress, and Gut Dysbiosis in Rats with Nonalcoholic Fatty Liver Disease. Nutrients 2023; 15:4375. [PMID: 37892450 PMCID: PMC10610228 DOI: 10.3390/nu15204375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) has been increasing worldwide, and the average age at NAFLD diagnosis has been decreasing. Although some components of adlay can ameliorate lipid metabolism, oxidative stress, inflammatory response, and gut microbiota, few studies have explored the effects of the dietary intake of intact dehulled adlay on liver diseases. Therefore, in this study, we investigated the effects of the dietary intake of dehulled adlay on NAFLD progression and explored the potential underlying mechanisms. Rats were randomized into a control group; a high-fat, high-sucrose diet (60% total energy derived from fat and 9.4% from sucrose)-induced NAFLD group (N); or a high-fat, high-sucrose diet with dehulled adlay group (received the same amounts of dietary fiber and total energy as did the N group). The experimental duration was 16 weeks. The diet containing dehulled adlay mitigated hepatic fat accumulation, proinflammatory cytokine levels, and oxidative stress by regulating the AMPK-Nrf2-NLRP3 inflammasome pathway and ferroptosis. Additionally, the dietary intake of dehulled adlay modulated the composition of the gut microbiota. In conclusion, a diet containing dehulled adlay may decelerate the progression of NAFLD by ameliorating hepatic steatosis, inflammation, oxidative stress, and gut dysbiosis.
Collapse
Affiliation(s)
- Hsuan-Chih Huang
- Department of Nutritional Science, Fu Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang District, New Taipei City 24205, Taiwan
| | - Pei-Ni Lee
- Department of Nutrition, Taipei Hospital, Ministry of Health and Welfare, No. 127, Siyuan Rd., Xinzhuang District, New Taipei City 24250, Taiwan;
| | - Wen-Chih Huang
- Department of Anatomical Pathology, Taipei Institute of Pathology, No. 146, Sec. 3, Chongqing N. Rd., Datong District, Taipei City 10374, Taiwan
| | - Hsin-Yi Yang
- Department of Nutritional Science, Fu Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang District, New Taipei City 24205, Taiwan
| |
Collapse
|
11
|
Gao Y, Meng Q, Qin J, Zhao Q, Shi B. Resveratrol alleviates oxidative stress induced by oxidized soybean oil and improves gut function via changing gut microbiota in weaned piglets. J Anim Sci Biotechnol 2023; 14:54. [PMID: 37029412 PMCID: PMC10080898 DOI: 10.1186/s40104-023-00851-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/12/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Oxidized soybean oil (OSO) has been shown to impair growth and exacerbate inflammation, leading to intestinal barrier injury in animals. Recent evidence suggests important roles for resveratrol (RES) in the promoting growth performance, antioxidant capacity, anti-inflammatory, and regulate intestinal barriers in animals. Therefore, The objectives of this study are to investigate the effects of dietary RES (purity 98%) supplementation on the growth performance, antioxidant capacity, inflammatory state, and intestinal function of weaned piglets challenged with OSO. METHODS A total of 28 castrated weaned male piglets with a similar body weight of 10.19 ± 0.10 kg were randomly assigned to 4 dietary treatments for 28-d feeding trial with 7 replications per treatment and 1 piglet per replicate. Treatments were arranged as a 2 × 2 factorial with oil type [3% fresh soybean oil (FSO) vs. 3% OSO] and dietary RES (0 vs. 300 mg/kg). RESULTS The results showed that relative to the FSO group, OSO stress tended to decrease the average daily feed intake (ADFI), and decreased the activity levels of lipase, villus/crypt ratio (VCR), the mRNA expression of FABP1, SOD2, IL-10 and ZO-1 in the jejunum, and SOD2, GPX1, occludin and ZO-1 in the colon, the levels of acetic acid in the colonic digesta, whereas up-regulated the mRNA expression of IL-1β and TNF-α in the jejunum (P < 0.05). Moreover, dietary supplementation with RES increased ether extract (EE), the activity levels of sucrase, lipase, α-amylase, villus height (VH) and VCR, the mRNA expression of FABP1, SOD2, IL-10 and occludin in the jejunum, and FABP1, PPAR-γ, GPX1, occludin and ZO-1 in the colon, and the abundance of Firmicutes, acetic and propionic acid, but decreased the levels of D-lactic acid in the plasma, the abundance of Bacteroidetes in the colonic digesta of weaned piglets compared to the non-RES group (P < 0.05). Meanwhile, in the interaction effect analysis, relative to the OSO group, dietary RES supplementation in the diets supplemented with OSO increased the activity levels of trypsin, VH in the jejunum, the abundance of Actinobacteria, the levels of butyric acid of weaned piglets, but failed to influence the activity levels of trypsin and VH, Actinobacteria abundance, the levels of butyric acid when diets were supplemented with FSO (interaction, P < 0.05). Relative to the OSO group, dietary RES supplementation in the diets supplemented with OSO decreased the activity levels of DAO in the plasma of weaned piglets but failed to influence the activity levels of DAO when diets were supplemented with FSO (interaction, P < 0.05). Relative to the FSO group, dietary RES supplementation in the diets supplemented with FSO decreased the level of propionic acid, whereas RES supplementation failed to influence the level of propionic acid when the diet was supplemented with OSO (interaction, P < 0.01). CONCLUSIONS Inclusion of OSO intensified inflammatory states and impaired the intestinal health characteristics of weaned piglets. Dietary RES supplementation improved the antioxidant capacity, anti-inflammatory activity, and intestinal morphology. Further studies showed that the protective effects of RES on gut health could be linked to the decreased abundance of Prevotella_1, Clostridium_sensu_stricto_6, and Prevotellaceae_UCG003 and increased levels of acetic and propionic acid.
Collapse
Affiliation(s)
- Yanan Gao
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin, China
| | - Qingwei Meng
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin, China
| | - Jianwei Qin
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin, China
| | - Qianqian Zhao
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin, China
| | - Baoming Shi
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin, China.
| |
Collapse
|
12
|
Effect of Lactobacillus plantarum on folliculogenesis in deep frying oil-fed rats. Reprod Toxicol 2023; 115:157-162. [PMID: 36572232 DOI: 10.1016/j.reprotox.2022.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Today, the tendency towards Western World diet characterized by a high consumption of Deep Frying Oil (DFO), as well as female infertility has increased. On the other hand, probiotics are living microorganisms that can benefit human health. Therefore, this study aimed to investigate the effect of a probiotic treatment (Lactobacillus plantarum) on the process of follicular growth in rats fed with DFO. Twenty adult female Wistar rats were divided into four groups: control, DFO treatment, DFO treatment group receiving probiotic and the healthy group receiving probiotic for one month. After blood sampling and dissection, ovarian tissue was examined for the number of ovarian follicles at different stages. In addition, the expression of Bmp15 (Gdf-9b) and Gdf9 genes was assessed by the real-time PCR method. The ELISA test was also used to measure hormonal changes (LH and FSH). Data showed that rats treated with DFO had a significant decrease in follicle numbers, hormone levels and Bmp15 and Gdf9 gene expression. Moreover, the number of atretic follicles was increased. Treatment of rats with the probiotic reduced the observed side effects of DFO. Thus, treatments of rats with the probiotic mitigated some of the observed side effects of DFO. An increase in primordial follicles and a reduction of atretic follicles was indicated compared to the DFO group (P ≤ 0.001). Lactobacillus plantarum could reduce the detrimental effects of DFO on folliculogenesis through its beneficial effects.
Collapse
|
13
|
FABP5 controls macrophage alternative activation and allergic asthma by selectively programming long-chain unsaturated fatty acid metabolism. Cell Rep 2022; 41:111668. [DOI: 10.1016/j.celrep.2022.111668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/13/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
|
14
|
Zhu H, Jiang W, Liu C, Wang C, Hu B, Guo Y, Cheng Y, Qian H. Ameliorative effects of chlorogenic acid on alcoholic liver injury in mice via gut microbiota informatics. Eur J Pharmacol 2022; 928:175096. [PMID: 35697148 DOI: 10.1016/j.ejphar.2022.175096] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/22/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022]
Abstract
Chlorogenic acid (CGA) is a functional phenolic acid widely used in food and medicine-related fields. It has been proved to be effective in the treatment of alcoholic liver disease (ALD). However, the exact mechanism by which CGA prevents ALD, especially from the crosstalk between gut and liver, has not been previously reported. This work was aimed to explore the protective effects of CGA against ALD and its relationships to gut-liver axis abnormalities. Experimental results showed the increased (p < 0.05) serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), low density lipoprotein (LDL), total cholesterol (TC) and triglyceride (TG) levels of mice fed with ethanol were ameliorated by supplementing with CGA. Moreover, CGA promoted the production of n-butyric acid by nearly 3 times (1.78 vs 0.62 nM, p < 0.01), a short-chain fatty acid that helps maintain the integrity of the intestinal barrier. Furthermore, CGA alleviated microbial dysbiosis, evidenced by the increased relative abundances of beneficial bacteria Muribaculaceae, Bacteroides, Alloprevotella, and Parabacteroides, and decreased that of opportunistic pathogens Eubacterium_nodatum, Eubacterium_ruminantium, and Anaerotruncus. Correlation analysis further elucidated the microbiota altered after CGA intervention was positively correlated with short-chain fatty acids and antioxidant indexes, while negatively correlated with inflammatory cytokines. In summary, these findings suggested the hepatoprotective effect of CGA was ascribed to the modulation of gut-liver axis homeostasis.
Collapse
Affiliation(s)
- Hongkang Zhu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; Synergetic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Wenhao Jiang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; Synergetic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Chang Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; Synergetic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Cheng Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; Synergetic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Bin Hu
- School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Yahui Guo
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; Synergetic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Yuliang Cheng
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; Synergetic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China.
| | - He Qian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; Synergetic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
15
|
Li X, Nian BB, Tan CP, Liu YF, Xu YJ. Deep-frying oil induces cytotoxicity, inflammation and apoptosis on intestinal epithelial cells. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:3160-3168. [PMID: 34786719 DOI: 10.1002/jsfa.11659] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 10/05/2021] [Accepted: 11/17/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Deep-frying oil has been found to cause inflammatory bowel disease (IBD). However, the molecular mechanism of the effect of deep-frying palm oil on IBD still remains undetermined. RESULTS In the present study, bioinformatics and cell biology were used to investigate the functions and signal pathway enrichments of differentially expressed genes. The bioinformatics analysis of three original microarray datasets (GSE73661, GSE75214 and GSE126124) in the NCBI-Gene Expression Omnibus database showed 17 down-regulated genes (logFC < 0) and 2 up-regulated genes (logFC > 0) existed in the enteritis tissue. Meanwhile, pathway enrichment and protein-protein interaction network analysis suggested that IBD is relevant to cytotoxicity, inflammation and apoptosis. Furthermore, Caco-2 cells were treated with the main oxidation products of deep-frying oil-total polar compounds (TPC) and its components (polymerized triglyceride, oxidized triglycerides and triglyceride degradation products) isolated from deep-frying oil. The flow cytometry experiment revealed that TPC and its components could induce apoptosis, especially for oxidized triglyceride. A quantitative polymerase chain reaction analysis demonstrated that TPC and its component could induce Caco-2 cell apoptosis through AQP8/CXCL1/TNIP3/IL-1. CONCLUSION The present study provides fundamental knowledge for understanding the effects of deep-frying oils on the cytotoxic and inflammatory of Caco-2 cells, in addition to clarifying the molecular function mechanism of deep-frying oil in IBD. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xue Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, China
| | - Bin-Bin Nian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, China
| | - Chin-Ping Tan
- Department of Food Technology, Faculty of Food Science and Technology, University Putra Malaysia, Seri Kembangan, Malaysia
| | - Yuan-Fa Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, China
| |
Collapse
|
16
|
Wu F, Zhuang P, Zhan C, Shen X, Jiao J, Zhang Y. Egg and Dietary Cholesterol Consumption and the Prevalence of Metabolic Syndrome: Findings from a Population-Based Nationwide Cohort. J Acad Nutr Diet 2022; 122:758-770.e5. [PMID: 34506988 DOI: 10.1016/j.jand.2021.09.010] [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] [Received: 09/10/2020] [Revised: 07/29/2021] [Accepted: 09/02/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Although the Chinese Dietary Guidelines (2016) removed restrictions on dietary cholesterol intake, evidence of egg and dietary cholesterol intake and cardiometabolic diseases is inconsistent. Associations between egg and cholesterol consumption and metabolic syndrome (MetS) in non-Western populations are still poorly documented. OBJECTIVE Our aim was to assess egg and dietary cholesterol intake in relation to the prevalence of MetS among participants in a Chinese nationwide study. DESIGN This cross-sectional study used data from the China Health and Nutrition Survey (1991-2009). PARTICIPANTS/SETTING The sample consisted of 8,241 healthy Chinese adults (20 years and older). MAIN OUTCOME MEASURES MetS cases were defined according to the National Cholesterol Education Program Adult Treatment Panel III criteria. STATISTICAL ANALYSIS Cumulative means of egg and cholesterol consumption were calculated in accordance with 3 consecutive 24-hour dietary recalls in each examination cycle. Logistic regression models were conducted to assess the associations with prevalent MetS. RESULTS Overall, 2,580 (31.3%) participants were identified as MetS cases in 2009. After multivariate adjustment, total egg consumption (>1 egg/d) was associated with 20% higher odds of MetS (odds ratio [OR] 1.20, 95% CI 1.06 to 1.37; P trend = .001) compared with consumption of ≤1/2 egg/d. Examining cooking methods, a positive association was observed between fried egg consumption and MetS odds (OR comparing the highest category [>1/2 egg/d] with the lowest category [≤1/7 egg/d] 1.22, 95% CI 1.08 to 1.39; P trend = .001), and nonfried egg intake was not associated with MetS odds (P trend = .08). Total dietary intake and egg-sourced cholesterol intake were both positively correlated with MetS odds (OR 1.31, 95% CI 1.12 to 1.53; P trend = .005) comparing the highest consumption (>371 mg · 2,000 kcal-1 · d-1) with the lowest consumption (≤132 mg · 2,000 kcal-1 · d-1) for total dietary cholesterol (OR 1.36; 95% CI 1.17 to 1.58; P trend < .001) and comparing the highest consumption (>232 mg · 2,000 kcal-1 · d-1) with the lowest consumption (≤46 mg · 2,000 kcal-1 · d-1) for egg-sourced cholesterol; similar associations were not observed for non-egg-sourced cholesterol consumption (P trend = .83). Substituting eggs and fried eggs for other protein sources, including low-fat and whole-fat dairy products; nuts and legumes; total red meat; processed meat; poultry meat; or seafood, was still associated with higher odds of MetS. CONCLUSIONS Consumption of >1 egg/d and >1/2 fried egg/d was associated with a higher prevalence of MetS than consumption of ≤1/2 egg/d and ≤1/7 fried egg/d. Future longitudinal cohort studies and randomized controlled trials are needed to further investigate the relationship between egg consumption and MetS and explore possible mechanisms of action.
Collapse
Affiliation(s)
- Fei Wu
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Pan Zhuang
- Zhejiang Key Laboratory for Agro-Food Processing, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chuchu Zhan
- Zhejiang Key Laboratory for Agro-Food Processing, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinyi Shen
- Zhejiang Key Laboratory for Agro-Food Processing, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingjing Jiao
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Yu Zhang
- Zhejiang Key Laboratory for Agro-Food Processing, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
17
|
Wang L, Chen W, Tian Y, Duan X, Yuan Y, Wang N, Xu C, Liu X, Liu Z. Preventive Effects of Sesamol on Deep‐frying Oil‐induced Liver Metabolism Disorders by Altering Gut Microbiota and Protecting Gut Barrier Integrity. Mol Nutr Food Res 2022; 66:e2101122. [DOI: 10.1002/mnfr.202101122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/07/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Lei Wang
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Weixuan Chen
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Yujie Tian
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Xiaorong Duan
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Yi Yuan
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Na Wang
- College of Food Science and Technology Henan Agricultural University Zhengzhou 450002 China
- Zhengzhou Key Laboratory of Nutrition and Health Food Zhengzhou 450002 China
| | - Chao Xu
- College of Food Science and Technology Henan Agricultural University Zhengzhou 450002 China
- Zhengzhou Key Laboratory of Nutrition and Health Food Zhengzhou 450002 China
| | - Xuebo Liu
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Zhigang Liu
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
- Department of Food Science Cornell University Ithaca New York 14853 United States
| |
Collapse
|
18
|
Kommentar. DIABETOL STOFFWECHS 2021. [DOI: 10.1055/a-1687-3383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
19
|
Gao J, Guo X, Wei W, Li R, Hu K, Liu X, Jiang W, Liu S, Wang W, Sun H, Wu H, Zhang Y, Gu W, Li Y, Sun C, Han T. The Association of Fried Meat Consumption With the Gut Microbiota and Fecal Metabolites and Its Impact on Glucose Homoeostasis, Intestinal Endotoxin Levels, and Systemic Inflammation: A Randomized Controlled-Feeding Trial. Diabetes Care 2021; 44:1970-1979. [PMID: 34253560 DOI: 10.2337/dc21-0099] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/03/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE This randomized controlled-feeding trial aimed to determine the impact of fried meat intake on the gut microbiota and fecal cometabolites and whether such impacts influenced host glucose homoeostasis, intestinal endotoxin levels, and systemic inflammation. RESEARCH DESIGN AND METHODS A total of 117 overweight adults were randomized into two groups. Fifty-nine participants were provided fried meat four times per week, and 58 participants were restricted from fried meat intake, while holding food group and nutrient compositions constant, for 4 weeks. The gut microbiota was analyzed by 16S rRNA sequencing. Glucose and insulin concentrations at 0, 30, 60, and 120 min of an oral glucose tolerance test, fecal microbiota-host cometabolite levels, and intestinal endotoxin and inflammation serum biomarker levels were measured. The area under the curve (AUC) for insulin, insulinogenic index (IGI), and muscle insulin resistance index (MIRI) were calculated. RESULTS The participants who consumed fried meat had lower IGI values than the control subjects, but they had higher MIRI and AUC values of insulin and lipopolysaccharide (LPS), TNF-α, IL-10, and IL-1β levels (P < 0.05). Fried meat intake lowered microbial community richness and decreased Lachnospiraceae and Flavonifractor abundances while increasing Dialister, Dorea, and Veillonella abundances (P FDR <0.05), provoking a significant shift in the fecal cometabolite profile, with lower 3-indolepropionic acid, valeric acid, and butyric acid concentrations and higher carnitine and methylglutaric acid concentrations (P FDR <0.05). Changes in these cometabolite levels were significantly associated with changes in IGI and MIRI values and LPS, FGF21, TNF-α, IL-1β, and IL-10 levels (P < 0.05). CONCLUSIONS Fried meat intake impaired glucose homoeostasis and increased intestinal endotoxin and systemic inflammation levels by influencing the gut microbiota and microbial-host cometabolites.
Collapse
Affiliation(s)
- Jian Gao
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Xiaoyu Guo
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Wei Wei
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Ran Li
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China.,Harbin Center for Disease Control and Prevention, Harbin, China
| | - Ke Hu
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Xin Liu
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Wenbo Jiang
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Siyao Liu
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Memphis, TN.,Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Weiqi Wang
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Hu Sun
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Huanyu Wu
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Yuntao Zhang
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Wenbo Gu
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Ying Li
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Harbin Medical University, Ministry of Education, Harbin, China.,NHC Key Laboratory of Cell Translation, Harbin Medical University, Harbin, China
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Tianshu Han
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China .,Department of Endocrinology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
20
|
Qi L. Fried Foods, Gut Microbiota, and Glucose Metabolism. Diabetes Care 2021; 44:1907-1909. [PMID: 34417280 PMCID: PMC8740929 DOI: 10.2337/dci21-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, and Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
21
|
|
22
|
Ruan M, Bu Y, Wu F, Zhang S, Chen R, Li N, Liu Z, Wang H. Chronic consumption of thermally processed palm oil or canola oil modified gut microflora of rats. FOOD SCIENCE AND HUMAN WELLNESS 2021. [DOI: 10.1016/j.fshw.2020.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
23
|
Li X, Tan CP, Liu YF, Xu YJ. Interactions between Food Hazards and Intestinal Barrier: Impact on Foodborne Diseases. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:14728-14738. [PMID: 33289375 DOI: 10.1021/acs.jafc.0c07378] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The intestine is an important digestive organ of the human body, and its barrier is the guardian of the body from the external environment. The impairment of the intestinal barrier is believed to be an important determinant in various foodborne diseases. Food hazards can lead to the occurrence of many foodborne diseases represented by inflammation. Therefore, understanding the mechanisms of the impact of the food hazards on intestinal barriers is essential for promoting human health. This review examined the relationship between food hazards and the intestinal barrier in three aspects: apoptosis, imbalance of gut microbiota, and pro-inflammatory cytokines. The mechanism of dysfunctional gut microbiota caused by food hazards was also discussed. This review discusses the interaction among food hazards, intestinal barrier, and foodborne diseases and, thus, offers a new thought to deal with foodborne disease.
Collapse
Affiliation(s)
- Xue Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, People's Republic of China
| | - Chin-Ping Tan
- Department of Food Technology, Faculty of Food Science and Technology, University Putra Malaysia, Selangor 410500, Malaysia
| | - Yuan-Fa Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, People's Republic of China
| |
Collapse
|
24
|
Xue LJ, Han JQ, Zhou YC, Peng HY, Yin TF, Li KM, Yao SK. Untargeted metabolomics characteristics of nonobese nonalcoholic fatty liver disease induced by high-temperature-processed feed in Sprague-Dawley rats. World J Gastroenterol 2020; 26:7299-7311. [PMID: 33362385 PMCID: PMC7739162 DOI: 10.3748/wjg.v26.i46.7299] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/27/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) has become one of the most common chronic liver diseases in the world. In our early clinical data and questionnaire analysis of NAFLD, it was found that the body mass index of some patients did not meet the diagnostic criteria for overweight or obesity. The consumption of high-temperature-processed foods such as fried food, hot pot and barbecue is closely related to the occurrence of nonobese NAFLD. Reducing the intake of this kind of food can reduce disease severity and improve prognosis.
AIM To explore the untargeted metabolomics characteristics of nonobese nonalcoholic fatty liver disease in Sprague-Dawley rats induced by high-temperature-processed feed.
METHODS Fifty-four male Sprague-Dawley rats were divided into three groups: The control group received a standard diet; the nonfried soybeans (NDFS) group received 60% NDFS and 40% basic feed and the dry-fried soybeans (DFS) group received 60% DFS and 40% basic feed. Six rats were sacrificed at week 4, 8, and 12 in each group. The food intake, body weight, Lee’s index, liver index, serological index and hepatic histopathology were assessed. Untargeted metabolomics characteristics were used to analyze the changes in liver metabolites of rats at week 12. Correlations between metabolites and pathology scores between the DFS and control groups and between the DFS and NDFS groups were analyzed. We selected some of the metabolites, both within the pathway and outside of the pathway, to explain preliminarily the difference in liver pathology in the three groups of rats.
RESULTS There were no statistically significant differences in the food intake, body weight, Lee's index or serological index between the DFS group and the control group (P > 0.05). At week 8 and week 12, the steatosis scores in the DFS group were significantly higher than those in the other two groups (P < 0.05). At week 12, the liver index of the DFS group was the lowest (NDFS group vs DFS group, P < 0.05). The fibrosis score in the DFS group was significantly higher than those in the other two groups (P < 0.05). The correlation analysis of the liver pathology score and differential metabolites in the DFS and NDFS groups showed that there were 10 strongly correlated substances: Five positively correlated substances and five negatively correlated substances. The positively correlated substances included taurochenodeoxycholate-3-sulfate, acetylcarnitine, 20a,22b-dihydroxycholesterol, 13E-tetranor-16-carboxy-LTE4 and taurocholic acid. The negatively correlated substances included choline, cholesterane-3,7,12,25-tetrol-3-glucuronide, nicotinamide adenine dinucleotide phosphate, lysoPC [16:1 (9Z)] and glycerol 3-phosphate. The correlation analysis of the liver pathology score and differential metabolites in the DFS and control groups showed that there were 13 strongly correlated substances: Four positively correlated substances and 9 negatively correlated substances. The positively correlated substances included 4-hydroxy-6-eicosanone, 3-phosphoglyceric acid, 13-hydroxy-9-methoxy-10-oxo-11-octadecenoic acid and taurochenodeoxycholate-3-sulfate. The negatively correlated substances included lysoPC [16:1(9Z)], S-(9-hydroxy-PGA1)-glutathione, lysoPC [20:5 (5Z, 8Z, 11Z, 14Z, 17Z)], SM (d18:1/14:0), nicotinamide adenine dinucleotide phosphate, 5,10-methylene-THF, folinic acid, N-lactoyl-glycine and 6-hydroxy-5-methoxyindole glucuronide.
CONCLUSION We successfully induced liver damage in rats by using a specially prepared high-temperature-processed feed and explored the untargeted metabolomics characteristics.
Collapse
Affiliation(s)
- Li-Jun Xue
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Ju-Qiang Han
- Department of Hepatology, The Seventh Medical Center of PLA General Hospital, Beijing 100700, China
| | - Yuan-Chen Zhou
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Hong-Ye Peng
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Teng-Fei Yin
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Kai-Min Li
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Shu-Kun Yao
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
25
|
Rayhan MA, Islam MK, Khatun MA, Islam D, Rahman MN. Remedial role of exercise training to deep-fried oil-induced metabolic and histological changes in Wistar rats. J Food Biochem 2020; 44:e13458. [PMID: 32869884 DOI: 10.1111/jfbc.13458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/30/2020] [Accepted: 08/11/2020] [Indexed: 11/29/2022]
Abstract
Exercise training is a well-known lifestyle to maintain good health. The present study was conducted to explore the effect of regular exercise training (for 15 min) on biochemical, physiological, and histological changes in fried oil intake (5 g/kg body weight, for 5 weeks) with or without raw oil supplementation. Liver disease and heart muscle injury were accounted for by significant (p < .05) increase in liver and heart biomarker enzymes in serum. Creatinine and urea level were also significantly increased in the fried oil-fed group as a sign of kidney injury. But all the biological markers including triglycerides, low-density lipoprotein cholesterol (LDL-C) was decreased significantly (p < .05) in the exercise-trained rat group. These metabolic changes were substantiated by the histological study of respective organs. Therefore, people should avoid repeatedly deep-fried oil consumption rather than fresh ones though regular exercise training has been found to resolve these metabolic abnormalities. PRACTICAL APPLICATIONS: Exercise training is a common practice to reduce the onset of some metabolic abnormalities for example cardiovascular disease, non-alcoholic fatty liver disease, chronic kidney disease, and also prevents various cell and tissue damages. Repeated frying of vegetable oil not only reduces its nutritional value but also produce free fatty acids and other toxic compounds. The present study revealed that repeatedly heated vegetable oil consumption causes injuries in the heart, liver, kidney, and small intestine by the oxidative products. As a result, the level of biomarkers of these responsive organs was found to be elevated. But regular exercise training ameliorated these detrimental effects. So, in terms of public health concern, regular exercise is one of the best ways to keep the body fit especially those who are consuming deep-fried oil.
Collapse
Affiliation(s)
- Md Abu Rayhan
- Laboratory of Lipid Research and Exercise Biochemistry, Department of Biochemistry and Molecular Biology, Jahangirnagar University, Dhaka, Bangladesh
| | - Md Kamrul Islam
- Laboratory of Lipid Research and Exercise Biochemistry, Department of Biochemistry and Molecular Biology, Jahangirnagar University, Dhaka, Bangladesh
| | - Mst Afifa Khatun
- Food Safety and Quality Analysis Division, Institute of Food and Radiation Biology, Atomic Energy Research Establishment, Savar, Dhaka, Bangladesh
| | - Dipa Islam
- Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research, Dhaka, Bangladesh
| | - Md Nazibur Rahman
- Laboratory of Lipid Research and Exercise Biochemistry, Department of Biochemistry and Molecular Biology, Jahangirnagar University, Dhaka, Bangladesh
| |
Collapse
|
26
|
Kianmehr P, Azarbayjani MA, Peeri M, Farzanegi P. Synergic effects of exercise training and octopamine on peroxisome proliferator-activated receptor-gamma coactivator -1a and uncoupling protein 1 mRNA in heart tissue of rat treated with deep frying oil. Biochem Biophys Rep 2020; 22:100735. [PMID: 32140572 PMCID: PMC7047140 DOI: 10.1016/j.bbrep.2020.100735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/18/2020] [Accepted: 01/21/2020] [Indexed: 11/06/2022] Open
Abstract
Octopamine (OCT) have an adverse effect on heart function. One of the positive effects of exercise training is improving cardiac function and cardiomyocytes signaling, which along with herbal supplements can have better effects on the heart tissue. Therefore, the aim of this study was to evaluate the effects of exercise training and OCT on changes of PGC1α and UCP1 expression in heart tissue of rat treated with deep frying oil (DFO). In this study, 45 male wistar rats were divided into 5 groups (n = 9 in each): I) control (Co), II) DFO, III) DFO + exercise, IV) DFO + OCT, and V) DFO + OCT + exercise. The quantification of apoptotic effects of DFO in heart tissue was assessed by TUNEL assay. Masson's trichrome stain applied to study cardiomyocytic fibers. Moreover, PGC1α and UCP1 genes and proteins expression in all groups were investigated using quantitative real-time PCR and immunohistochemical method. A significant increase in apoptotic cells was observed in the DFO-treated group (p < 0.05). In Masson's Trichrome stain study, more cardiomyocytic fibers were observed and some lymphocytic cells were present in some fibers. Also, the expression of PGC1α and UCP1 was significantly increase in DFO + exercise group, DFO + OCT group, and DFO + OCT + exercise group compare to DFO group (p < 0.05). Based on these findings, exercise and octopamine can be considered as factors affecting the expression of PGC1α genes and UCP1 as well as drug poisoning.
Exercise training with octopamine can be upregulate expression of PGC1α. Exercise training with octopamine can be upregulate expression of UCP1. Increase the PGC1α and UCP1 can improve the mitochondria disfunction induced by DFO.
Collapse
Affiliation(s)
- Pantea Kianmehr
- -Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Ali Azarbayjani
- -Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maghsoud Peeri
- -Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Parvin Farzanegi
- -Department of Exercise Physiology, Sari Branch, Islamic Azad University, Sari, Iran
| |
Collapse
|
27
|
Food matrix and the microbiome: considerations for preclinical chronic disease studies. Nutr Res 2020; 78:1-10. [PMID: 32247914 DOI: 10.1016/j.nutres.2020.02.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/05/2020] [Accepted: 02/25/2020] [Indexed: 01/05/2023]
Abstract
Animal models of chronic disease are continuously being refined and have evolved with the goal of increasing the translation of results to human populations. Examples of this progress include transgenic models and germ-free animals conventionalized with human microbiota. The gut microbiome is involved in the etiology of several chronic diseases. Therefore, consideration of the experimental conditions that may affect the gut microbiome in preclinical disease is very important. Of note, diet plays a large role in shaping the gut microbiome and can be a source of variation between animal models and human populations. Traditionally, nutrition researchers have focused on manipulating the macronutrient profile of experimental diets to model diseases such as metabolic syndrome. However, other dietary components found in human foods, but not in animal diets, can have sizable effects on the composition and metabolic capacity of the gut microbiome and, as a consequence, manifestation of the chronic disease being modeled. The purpose of this review is to describe how food matrix food components, including diverse fiber sources, oxidation products from cooking, and dietary fat emulsifiers, shape the composition of the gut microbiome and influence gut health.
Collapse
|
28
|
Short-chain fructo-oligosaccharides alleviates oxidized oil-induced intestinal dysfunction in piglets associated with the modulation of gut microbiota. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
29
|
Park SO, Zammit VA. In vivo monitoring of hepatic glycolipid distribution of n-6 ∕ n-3 in jugular-vein-cannulated rats as a nutritional research model for monogastric animal. Arch Anim Breed 2019; 62:437-446. [PMID: 31807655 PMCID: PMC6852779 DOI: 10.5194/aab-62-437-2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 06/04/2019] [Indexed: 12/13/2022] Open
Abstract
The metabolic distribution via blood from liver of glycerolipids by omega-6
to omega-3 fatty acid (n-6 / n-3) ratio in monogastric animal nutrition is
very important. In vivo monitoring technique using jugular-vein-cannulated
rats as a nutritional model for monogastric animal can yield important
insights into animal nutrition. This study was conducted to determine the
effect of different n-6 / n-3 ratios (71:1, 4:1, 15:1, 30:1) on
metabolic distribution of glycerolipids newly synthesized and secreted in
the liver of the rats and explore the mechanism involved. Regarding
14CO2 released from oxidation of glycerolipid metabolism, it was
the highest (22.5 %) in groups with a n-6 / n-3 ratio of 4:1 (P<0.05).
The control group showed the highest total glycerolipid level, followed by the
30:1, 15:1, and 4:1 groups in order (P<0.05). When secreted
triacylglycerol level of each group was compared with that of the control
group, the 4:1, 15:1, and 30:1 groups were decreased by 36.3 %, 20.9 %, and
13.3 %, respectively (P<0.05). Regarding the distribution of
phospholipid against total glycerolipid compared to the control group, the 4:1, 15:1, and 30:1 groups were 1.38, 1.29, and 1.17 times higher, respectively
(P<0.05). In the comparison of 14CO2 emission against
total glycerolipid compared with the control group, the 4:1, 15:1, and 30:1
groups were 1.61, 1.52, and 1.29 times higher, respectively
(P<0.05). These results demonstrate that a dietary n-6 / n-3 fatty acid
ratio of 4:1 could significantly decrease harmful lipid levels in the blood
by controlling the mechanism of metabolic distribution via blood from
triglyceride and phospholipid newly synthesized in the liver of cannulated
rat.
Collapse
Affiliation(s)
- Sang-O Park
- College of Animal Life Science, Kangwon National University, Chuncheon, Gangwon-do, 24419 Republic of Korea
| | - Victor A Zammit
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
30
|
Boukandoul S, Santos CS, Casal S, Zaidi F. Oxidation delay of sunflower oil under frying by moringa oil addition: more than just a blend. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2019; 99:5483-5490. [PMID: 31087351 DOI: 10.1002/jsfa.9809] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 05/05/2019] [Accepted: 05/09/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Blending Moringa oleifera oil (MOO) with other oils of recognized lower stability under prolonged frying results in oxidation delay. The present study aimed to detail the probable molecular interactions supporting these observations, using a small amount of MOO (20%) and sunflower oil (SFO; 80%) under domestic deep-frying conditions (intermittent frying of fresh potatoes, 180 °C, 2 × 90 min day-1 , 5 days). RESULTS Blending 20% MOO with SFO resulted in a significantly lower formation of polymers (<43 to 85%) and oxidized triglycerides (<20 to 60%), a 25-60% reduction in p-anisidine value and total volatile aldehydes, particularly alkadienals, and a better performance than the one predicted from the oils' mass ratio. Blending was particularly effective in vitamin E and antioxidant activity preservation, probably from interaction with some MOO components such as sterols and vitamin E, while carotenoids and phenolics do not seem to be implicated. CONCLUSIONS These results provide an interesting use for MOO, improving the thermo-oxidative performance of SFO while providing nutritional benefits and lowering the formation of toxic compounds during prolonged deep-frying. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Silia Boukandoul
- Département des Sciences Alimentaires, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
- LAQV@REQUIMTE/Laboratory of Bromatology and Hydrology, Faculty of Pharmacy, Porto University, Porto, Portugal
| | - Carla Sp Santos
- LAQV@REQUIMTE/Laboratory of Bromatology and Hydrology, Faculty of Pharmacy, Porto University, Porto, Portugal
| | - Susana Casal
- LAQV@REQUIMTE/Laboratory of Bromatology and Hydrology, Faculty of Pharmacy, Porto University, Porto, Portugal
| | - Farid Zaidi
- Département des Sciences Alimentaires, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
| |
Collapse
|
31
|
Larson ED, Magno JPM, Steritz MJ, Llanes EGDV, Cardwell J, Pedro M, Roberts TB, Einarsdottir E, Rosanes RAQ, Greenlee C, Santos RAP, Yousaf A, Streubel SO, Santos ATR, Ruiz AG, Lagrana-Villagracia SM, Ray D, Yarza TKL, Scholes MA, Anderson CB, Acharya A, Gubbels SP, Bamshad MJ, Cass SP, Lee NR, Shaikh RS, Nickerson DA, Mohlke KL, Prager JD, Cruz TLG, Yoon PJ, Abes GT, Schwartz DA, Chan AL, Wine TM, Cutiongco-de la Paz EM, Friedman N, Kechris K, Kere J, Leal SM, Yang IV, Patel JA, Tantoco MLC, Riazuddin S, Chan KH, Mattila PS, Reyes-Quintos MRT, Ahmed ZM, Jenkins HA, Chonmaitree T, Hafrén L, Chiong CM, Santos-Cortez RLP. A2ML1 and otitis media: novel variants, differential expression, and relevant pathways. Hum Mutat 2019; 40:1156-1171. [PMID: 31009165 DOI: 10.1002/humu.23769] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/26/2019] [Accepted: 04/18/2019] [Indexed: 12/16/2022]
Abstract
A genetic basis for otitis media is established, however, the role of rare variants in disease etiology is largely unknown. Previously a duplication variant within A2ML1 was identified as a significant risk factor for otitis media in an indigenous Filipino population and in US children. In this report exome and Sanger sequencing was performed using DNA samples from the indigenous Filipino population, Filipino cochlear implantees, US probands, Finnish, and Pakistani families with otitis media. Sixteen novel, damaging A2ML1 variants identified in otitis media patients were rare or low-frequency in population-matched controls. In the indigenous population, both gingivitis and A2ML1 variants including the known duplication variant and the novel splice variant c.4061 + 1 G>C were independently associated with otitis media. Sequencing of salivary RNA samples from indigenous Filipinos demonstrated lower A2ML1 expression according to the carriage of A2ML1 variants. Sequencing of additional salivary RNA samples from US patients with otitis media revealed differentially expressed genes that are highly correlated with A2ML1 expression levels. In particular, RND3 is upregulated in both A2ML1 variant carriers and high-A2ML1 expressors. These findings support a role for A2ML1 in keratinocyte differentiation within the middle ear as part of otitis media pathology and the potential application of ROCK inhibition in otitis media.
Collapse
Affiliation(s)
- Eric D Larson
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Jose Pedrito M Magno
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines
| | - Matthew J Steritz
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Erasmo Gonzalo D V Llanes
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Jonathan Cardwell
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Melquiadesa Pedro
- Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Tori Bootpetch Roberts
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Elisabet Einarsdottir
- Folkhälsan Institute of Genetics and Molecular Neurology Research Program, University of Helsinki, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Rose Anne Q Rosanes
- Department of Community Dentistry, College of Dentistry, University of the Philippines Manila, Manila, Philippines
| | - Christopher Greenlee
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | | | - Ayesha Yousaf
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, Punjab, Pakistan
| | - Sven-Olrik Streubel
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | | | - Amanda G Ruiz
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Sheryl Mae Lagrana-Villagracia
- Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Dylan Ray
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Talitha Karisse L Yarza
- Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines.,Newborn Hearing Screening Reference Center, University of the Philippines Manila - National Institutes of Health (NIH), Manila, Philippines
| | - Melissa A Scholes
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Catherine B Anderson
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Anushree Acharya
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | | | - Samuel P Gubbels
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Michael J Bamshad
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | - Stephen P Cass
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Nanette R Lee
- USC-Office of Population Studies Foundation, Inc. and Department of Anthropology, Sociology and History, University of San Carlos, Cebu, Philippines
| | - Rehan S Shaikh
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, Punjab, Pakistan
| | - Deborah A Nickerson
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Jeremy D Prager
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Teresa Luisa G Cruz
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Patricia J Yoon
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Generoso T Abes
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - David A Schwartz
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Abner L Chan
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Todd M Wine
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Eva Maria Cutiongco-de la Paz
- Philippine Genome Center, University of the Philippines, Quezon City, Philippines.,University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Norman Friedman
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Katerina Kechris
- Department of Biostatistics and Bioinformatics, Colorado School of Public Health, Aurora, Colorado
| | - Juha Kere
- Folkhälsan Institute of Genetics and Molecular Neurology Research Program, University of Helsinki, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Suzanne M Leal
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Ivana V Yang
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Janak A Patel
- Division of Infectious Diseases, Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas
| | - Ma Leah C Tantoco
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Saima Riazuddin
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kenny H Chan
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Petri S Mattila
- Department of Otorhinolaryngology, Head & Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Maria Rina T Reyes-Quintos
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines.,Newborn Hearing Screening Reference Center, University of the Philippines Manila - National Institutes of Health (NIH), Manila, Philippines.,University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Zubair M Ahmed
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Herman A Jenkins
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Tasnee Chonmaitree
- Division of Infectious Diseases, Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas
| | - Lena Hafrén
- Department of Otorhinolaryngology, Head & Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Charlotte M Chiong
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines.,Newborn Hearing Screening Reference Center, University of the Philippines Manila - National Institutes of Health (NIH), Manila, Philippines
| | - Regie Lyn P Santos-Cortez
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines.,Center for Children's Surgery, Children's Hospital Colorado, Aurora, Colorado
| |
Collapse
|
32
|
Zhang J, Lu Y, Yang X, Zhao Y. Supplementation of okra seed oil ameliorates ethanol-induced liver injury and modulates gut microbiota dysbiosis in mice. Food Funct 2019; 10:6385-6398. [DOI: 10.1039/c9fo00189a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study assesses the possible effects of dietary okra seed oil (OSO) consumption on attenuation of alcohol-induced liver damage and gut microbiota dysbiosis, and associated mechanisms in mice.
Collapse
Affiliation(s)
- Jin Zhang
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry
- College of Life Sciences
- Shaanxi Normal University
- Xi'an 710062
- China
| | - Yalong Lu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control
- College of Food Engineering and Nutritional Science
- Shaanxi Normal University
- Xi'an 710062
- China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control
- College of Food Engineering and Nutritional Science
- Shaanxi Normal University
- Xi'an 710062
- China
| | - Yan Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry
- College of Life Sciences
- Shaanxi Normal University
- Xi'an 710062
- China
| |
Collapse
|
33
|
Idris CAC, Sundram K, Razis AFA. Effect of Consumption Heated Oils with or without Dietary Cholesterol on the Development of Atherosclerosis. Nutrients 2018; 10:nu10101527. [PMID: 30336600 PMCID: PMC6213572 DOI: 10.3390/nu10101527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/10/2018] [Accepted: 10/13/2018] [Indexed: 01/22/2023] Open
Abstract
Heating oils and fats for a considerable length of time results in chemical reactions, leading to the aggravation of a free radical processes, which ultimately contributes to atherosclerosis. Our study focused on elucidating the effect of feeding heated oils with or without dietary cholesterol on the development of atherosclerosis in rabbits. We heated palm olein and corn oil at 180 °C for 18 h and 9 h per day, respectively, for two consecutive days. Next, 20 male rabbits were divided into four groups and fed the following diet for 12 weeks: (i) heated palm olein (HPO); (ii) HPO with cholesterol (HPOC); (iii) heated corn oil (HCO); and (iv) HCO with cholesterol (HCOC). Plasma total cholesterol (TC) was significantly lower in the HCO group compared to the HCOC group. Atherosclerotic lesion scores for both fatty plaques and fatty streaks were significantly higher in the HCO and HCOC groups as compared to the HPO and HPOC groups. Additionally, fibrous plaque scores were also higher in the HCO and HCOC groups as compared to the HPO and HPOC groups. These results suggest that heated palm oil confers protection against the onset of atherosclerosis compared to heated polyunsaturated oils in a rabbit model.
Collapse
Affiliation(s)
- Che Anishas Che Idris
- Malaysian Palm Oil Board, No. 6, Persiaran Institusi, Bandar Baru Bangi, Kajang 43000, Selangor, Malaysia.
| | - Kalyana Sundram
- Malaysian Palm Oil Council, 2nd Floor, Wisma Sawit, Lot 6, SS6, Jalan Perbandaran, Kelana Jaya 47301, Selangor, Malaysia.
| | - Ahmad Faizal Abdull Razis
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia.
- Laboratory of Food Safety and Food Integrity, Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia.
- Faculty of Food Science and Technology, Universiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia.
| |
Collapse
|
34
|
Danchin A. Bacteria in the ageing gut: did the taming of fire promote a long human lifespan? Environ Microbiol 2018; 20:1966-1987. [PMID: 29727052 DOI: 10.1111/1462-2920.14255] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Unique among animals as they evolved towards Homo sapiens, hominins progressively cooked their food on a routine basis. Cooked products are characterized by singular chemical compounds, derived from the pervasive Maillard reaction. This same reaction is omnipresent in normal metabolism involving carbonyls and amines, and its products accumulate with age. The gut microbiota acts as a first line of defence against the toxicity of cooked Maillard compounds, that also selectively shape the microbial flora, letting specific metabolites to reach the blood stream. Positive selection of metabolic functions allowed the body of hominins who tamed fire to use and dispose of these age-related compounds. I propose here that, as a hopeful accidental consequence, this resulted in extending human lifespan far beyond that of our great ape cousins. The limited data exploring the role of taming fire on the human genetic setup and on its microbiota is discussed in relation with ageing.
Collapse
Affiliation(s)
- Antoine Danchin
- Integromics, Institute of Cardiometabolism and Nutrition, Hôpital de la Pitié-Salpêtrière, 47 Boulevard de l'Hôpital, Paris, 75013, France.,School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, Hong Kong University, 21 Sassoon Road, Pokfulam, Hong Kong
| |
Collapse
|
35
|
Wang L, Yao D, Urriola PE, Hanson AR, Saqui-Salces M, Kerr BJ, Shurson GC, Chen C. Identification of activation of tryptophan-NAD + pathway as a prominent metabolic response to thermally oxidized oil through metabolomics-guided biochemical analysis. J Nutr Biochem 2018; 57:255-267. [PMID: 29800812 DOI: 10.1016/j.jnutbio.2018.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 02/08/2023]
Abstract
Consumption of thermally oxidized oil is associated with metabolic disorders, but oxidized oil-elicited changes in the metabolome are not well defined. In this study, C57BL/6 mice were fed the diets containing either control soybean oil or heated soybean oil (HSO) for 4 weeks. HSO-responsive metabolic events were examined through untargeted metabolomics-guided biochemical analysis. HSO directly contributed to the presence of new HSO-derived metabolites in urine and the decrease of polyunsaturated fatty acid-containing phospholipids in serum and the liver. HSO disrupted redox balance by decreasing hepatic glutathione and ascorbic acid. HSO also activated peroxisome proliferator-activated receptors, leading to the decrease of serum triacylglycerols and the changes of cofactors and products in fatty acid oxidation pathways. Most importantly, multiple metabolic changes, including the decrease of tryptophan in serum; the increase of NAD+ in the liver; the increases of kynurenic acid, nicotinamide and nicotinamide N-oxide in urine; and the decreases of the metabolites from pyridine nucleotide degradation in the liver indicated that HSO activated tryptophan-NAD+ metabolic pathway, which was further confirmed by the upregulation of gene expression in this pathway. Because NAD+ and its metabolites are essential cofactors in many HSO-induced metabolic events, the activation of tryptophan-NAD+ pathway should be considered as a central metabolic response to the exposure of HSO.
Collapse
Affiliation(s)
- Lei Wang
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN 55108, USA
| | - Dan Yao
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN 55108, USA
| | - Pedro E Urriola
- Department of Animal Science, University of Minnesota, St. Paul, MN 55108, USA
| | - Andrea R Hanson
- Department of Animal Science, University of Minnesota, St. Paul, MN 55108, USA
| | - Milena Saqui-Salces
- Department of Animal Science, University of Minnesota, St. Paul, MN 55108, USA
| | - Brian J Kerr
- USDA-ARS-National Laboratory for Agriculture and the Environment, USDA, Ames, IA 50011, USA
| | - Gerald C Shurson
- Department of Animal Science, University of Minnesota, St. Paul, MN 55108, USA
| | - Chi Chen
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN 55108, USA; Department of Animal Science, University of Minnesota, St. Paul, MN 55108, USA.
| |
Collapse
|
36
|
The Effects of Heated Oils Used in Fast Food Restaurants on Metabolic, Inflammatory and Oxidative Stress Markers, Blood Pressure, and Liver Histology in Sprague-Dawley Rats. IRANIAN RED CRESCENT MEDICAL JOURNAL 2018. [DOI: 10.5812/ircmj.59696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
37
|
Zhang X, Wang H, Yin P, Fan H, Sun L, Liu Y. Flaxseed oil ameliorates alcoholic liver disease via anti-inflammation and modulating gut microbiota in mice. Lipids Health Dis 2017; 16:44. [PMID: 28228158 PMCID: PMC5322643 DOI: 10.1186/s12944-017-0431-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 02/13/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Alcoholic liver disease (ALD) represents a chronic wide-spectrum of liver injury caused by consistently excessive alcohol intake. Few satisfactory advances have been made in management of ALD. Thus, novel and more practical treatment options are urgently needed. Flaxseed oil (FO) is rich in α-linolenic acid (ALA), a plant-derived n-3 polyunsaturated fatty acids (PUFAs). However, the impact of dietary FO on chronic alcohol consumption remains unknown. METHODS In this study, we assessed possible effects of dietary FO on attenuation of ALD and associated mechanisms in mice. Firstly, mice were randomly allocated into four groups: pair-fed (PF) with corn oil (CO) group (PF/CO); alcohol-fed (AF) with CO group (AF/CO); PF with FO group (PF/FO); AF with FO group (AF/FO). Each group was fed modified Lieber-DeCarli liquid diets containing isocaloric maltose dextrin a control or alcohol with corn oil and flaxseed oil, respectively. After 6 weeks feeding, mice were euthanized and associated indications were investigated. RESULTS Body weight (BW) was significantly elevated in AF/FO group compared with AF/CO group. Dietary FO reduced the abnormal elevated aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels in chronic ethanol consumption. Amelioration of these parameters as well as liver injury via HE staining in dietary FO supplementation in ALD demonstrated that dietary FO can effectively benefit for the protection against ALD. To further understand the underlying mechanisms, we investigated the inflammatory cytokine levels and gut microbiota. A series of inflammatory cytokines, including TNF-α, IL-1β, IL-6 and IL-10, were determined. As a result, TNF-α, IL-1β and IL-6 were decreased in AF/FO group compared with control group; IL-10 showed no significant alteration between AF/CO and AF/FO groups (p > 0.05). Sequencing and analysis of gut microbiota gene indicated that a reduction of Porphyromonadaceae and Parasutterella, as well as an increase in Firmicutes and Parabacteroides, were seen in AF group compared with PF control. Furthermore, dietary FO in ethanol consumption group induced a significant reduction in Proteobacteria and Porphyromonadaceae compared with AF/CO group. CONCLUSION Dietary FO ameliorates alcoholic liver disease via anti-inflammation and modulating gut microbiota, thus can potentially serve as an inexpensive interventions for the prevention and treatment of ALD.
Collapse
Affiliation(s)
- Xiaoxia Zhang
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Qinghua Donglu No35, Haidian District, Beijing, 100083, China.,Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Hao Wang
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Peipei Yin
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Qinghua Donglu No35, Haidian District, Beijing, 100083, China
| | - Hang Fan
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Qinghua Donglu No35, Haidian District, Beijing, 100083, China
| | - Liwei Sun
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Qinghua Donglu No35, Haidian District, Beijing, 100083, China
| | - Yujun Liu
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Qinghua Donglu No35, Haidian District, Beijing, 100083, China.
| |
Collapse
|