1
|
Ji X, Ding H, Zhou F, Zhang F, Wu D. Taurine ameliorates deoxynivalenol-induced intestinal injury in piglets: Restoration of mitochondrial function linked to the PGC1α-NRF1/2 axis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117938. [PMID: 39986055 DOI: 10.1016/j.ecoenv.2025.117938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Deoxynivalenol (DON) is a prevalent foodborne contaminant present in crops, posing significant risks to food safety and public health worldwide. Mitochondria, as the primary target of DON, play a crucial role in DON-mediated gastrointestinal toxicity. Taurine, a multifunctional nutrient, has been reported to exert antioxidant and anti-inflammatory effects by modulating mitochondrial function. However, whether taurine could alleviate intestinal injury by restoring mitochondrial function under DON exposure remains unclear. To address this knowledge gap, this study systematically investigated the potential protective effects of taurine on DON-induced intestinal damage in a piglet model. Twenty-four piglets were randomly assigned to four groups for 24 days: BD group (basal diet), DON group (3 mg/kg DON-contaminated diet), DON+LT group (DON diet with 0.3 % taurine), and DON+HT group (DON diet with 0.6 % taurine). Serum samples were collected for biochemical analysis, while jejunal tissues were examined for histology, barrier function, oxidative stress, inflammation, apoptosis, mitochondrial function, as well as related gene and protein expression. The results revealed that taurine effectively restored jejunal morphology disrupted by DON, as evidenced by increases in villus height/width and the villus height to crypt depth ratio. It preserved intestinal barrier integrity, reflected by reductions in diamine oxidase and D-lactate levels, alongside increased expression of genes and proteins related to intestinal mucus and mechanical barrier function. Furthermore, taurine mitigated intestinal oxidative stress by reducing reactive oxygen species, 8-hydroxydeoxyguanosine, and malondialdehyde levels, while enhancing antioxidant defenses. It also alleviated inflammation by suppressing pro-inflammatory cytokines and attenuated intestinal epithelial apoptosis through mitochondrial caspase-dependent and apoptosis-inducing factor-mediated pathways. Intriguingly, taurine improved the damaged mitochondrial structure and functionality within the intestinal epithelium irritated by DON. This improvement included enhanced respiratory chain complex activity, increased ATP levels, and mtDNA copy number. Additionally, taurine regulated gene expression related to mitochondrial respiration, fusion, fission, and autophagy. Simultaneously, taurine reversed the DON-induced inhibition of the peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1α)-nuclear respiratory factor 1/2 (NRF1/2) axis, a critical pathway regulating mitochondrial biogenesis, respiratory function, and oxidative stress responses. Correlation analysis revealed significant associations between the PGC1α-NRF1/2 axis and mitochondrial function, as well as correlations with intestinal health parameters, including barrier function, redox status, inflammation, and apoptosis. In summary, this study provides the first evidence that dietary taurine supplementation effectively alleviates intestinal injury in DON-challenged piglets through mitochondrial restoration, which is strongly associated with the reactivation of the PGC1α-NRF1/2 axis. Our findings highlight the potential of mitochondrial-targeted therapies to mitigate gastrointestinal toxicity caused by the foodborne contaminant DON in both humans and animals.
Collapse
Affiliation(s)
- Xu Ji
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Hongyan Ding
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Fen Zhou
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Feng Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou 233100, China; Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou 233100, China.
| | - Dong Wu
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China.
| |
Collapse
|
2
|
Lei Y, Ma XL, Liu T, Wang MJ, Kang JS, Yang J, Mi N. Lactucin ameliorates FFA-induced steatosis in HepG2 cells by modulating mitochondrial homeostasis through the SIRT1/PGC-1α signaling axis. Heliyon 2024; 10:e39890. [PMID: 39524853 PMCID: PMC11550614 DOI: 10.1016/j.heliyon.2024.e39890] [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: 05/08/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Nonalcoholic fatty liver disease is a complex disease involving abnormal liver metabolism. Its strong association with metabolic dysfunction has led to a change in nomenclature to metabolism dysfunction-associated fatty liver disease (MAFLD). MAFLD pathogenesis involves abnormal accumulation of hepatic lipids that lead to the production of excess free fatty acids (FFAs), which in turn cause an imbalance in hepatic mitochondrial function. Lactucin, a natural compound extracted from Cichorium glandulosum Boiss. et Huet, regulates liver metabolism and protects the liver. However, the potential mechanisms underlying the lactucin-mediated effects in MAFLD require further investigation. In the present study, HepG2 cells were treated with FFAs to establish an in vitro model of MAFLD. Parameters related to lipid accumulation and mitochondrial function, including triglycerides (TG), oil red O-stained lipid droplets, reactive oxygen species (ROS), mitochondrial membrane potential (JC-1), adenine triphosphate (ATP), and complex III were analysed. Morphology of the mitochondria were evaluated by transmission electron microscopy. Furthermore, key proteins in the sirtuin 1 (SIRT1)/peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) signalling axis and mitochondrial quality control were analysed. The SIRT1 inhibitor EX-527 was used to verify the key role of the SIRT1 signalling pathway. Western blotting showed that lactucin upregulated the expression of SIRT-1, PGC-1α, Nrf1, Tfam, Mfn2, and Opa1, and promoted mitochondrial biosynthesis and kinetics. The results suggest that lactucin restores mitochondrial dynamic homeostasis by upregulating the SIRT1/PGC-1α signalling axis, thereby reducing FFA-induced lipid accumulation in HepG2 cells.
Collapse
Affiliation(s)
- Yi Lei
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Xiao-li Ma
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Urumqi, 830000, China
| | - Tong Liu
- Basic Medical College, Xinjiang Medical University, Urumqi, 830011, China
| | - Meng-jiao Wang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Jin-sen Kang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Urumqi, 830000, China
| | - Jian Yang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Urumqi, 830000, China
| | - Na Mi
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Urumqi, 830000, China
- Clinical Medicine Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| |
Collapse
|
3
|
Zhu J, Nie G, Dai X, Wang D, Li S, Zhang C. Activating PPARβ/δ-Mediated Fatty Acid β-Oxidation Mitigates Mitochondrial Dysfunction Co-induced by Environmentally Relevant Levels of Molybdenum and Cadmium in Duck Kidneys. Biol Trace Elem Res 2024:10.1007/s12011-024-04450-8. [PMID: 39546187 DOI: 10.1007/s12011-024-04450-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024]
Abstract
Cadmium (Cd) and high molybdenum (Mo) pose deleterious effects on health. Prior studies have indicated that exposure to Mo and Cd leads to damage in duck kidneys, but limited studies have explored this damage from the perspective of fatty acid metabolism. In this study, 40 healthy 8-day-old ducks were randomly assigned to four groups and fed a basic diet containing Cd (4 mg/kg Cd) or Mo (100 mg/kg Mo) or both. Kidney tissues were harvested on the 16th week. Results demonstrated that Cd and/or Mo inhibited mitochondrial fatty acid β-oxidation and disrupted mitochondrial dynamics, along with significant suppression of peroxisome proliferator-activated receptor β/δ (PPARβ/δ) protein in duck kidneys. In vitro study, duck renal tubular epithelial cells were exposed for 12 h to either Mo (480 μM Mo), Cd (2.5 μM Cd), and GW0742 (0.3 μM, a potent agonist of PPARβ/δ) alone or in combination. The results demonstrated that Cd and/or Mo led to marked fatty acid oxidation deficiency and mitochondrial dysfunction and that PPARβ/δ protein was involved in the process. Altogether, this study found that activating PPARβ/δ-mediated fatty acid β-oxidation mitigates mitochondrial dysfunction co-induced by Mo and Cd in duck kidneys.
Collapse
Affiliation(s)
- Jiamei Zhu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Gaohui Nie
- Jiangxi Hongzhou Vocational College, Fengcheng, Jiangxi, China
| | - Xueyan Dai
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Dianyun Wang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - ShanXin Li
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China.
| |
Collapse
|
4
|
Zhao H, Li Z, Sun Y, Yan M, Wang Y, Li Y, Zhang Y, Zhu M. Supplementation of Chlorogenic Acid Alleviates the Effects of H 2O 2-Induced Oxidative Stress on Laying Performance, Egg Quality, Antioxidant Capacity, Hepatic Inflammation, Mitochondrial Dysfunction, and Lipid Accumulation in Laying Hens. Antioxidants (Basel) 2024; 13:1303. [PMID: 39594445 PMCID: PMC11591049 DOI: 10.3390/antiox13111303] [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: 09/11/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
This research examined the impact of chlorogenic acid (CGA) on laying performance, antioxidant capacity, egg quality, hepatic inflammation, mitochondrial function, and lipid metabolism in hens subjected to hydrogen peroxide (H2O2)-induced oxidative stress (OS). Three hundred sixty healthy 43-wk-old Hy-Line brown hens were randomly assigned to six treatments: a basal diet + 0 (control and H2O2), 600 (600 mg/kg CGA and 600 mg/kg CGA + H2O2), and 800 (800 mg/kg CGA and 800 mg/kg CGA + H2O2) mg/kg CGA for 84 d. On the 64th and 78th days of the trial, hens in groups H2O2, 600 mg/kg CGA + H2O2, and 800 mg/kg CGA + H2O2 were injected intraperitoneally with 10% H2O2. The results demonstrated that 600 and 800 mg/kg CGA significantly improved the egg production rate (EPR) and egg quality and reduced lipid peroxidation compared to the control group. The 800 mg/kg CGA showed greater improvements in the EPR and average egg weight (AEW) compared to the 600 mg/kg dose. Conversely, H2O2 exposure significantly decreased the EPR, AEW, and egg quality and increased feed conversion rate and average daily feed intake. H2O2 exposure significantly decreased serum T-AOC and increased serum MDA levels while reducing hepatic T-SOD, GSH-Px, and CAT activities. Meanwhile, H2O2 exposure significantly elevated liver reactive oxygen species levels, pathological damage, and NF-κB, TNFα, and IL-1β gene expression. Additionally, H2O2 treatment disrupted hepatocyte mitochondrial structure and significantly increased the expression of VDAC1 protein, and IP3R, GRP75, MCU, Fis1, and MFF genes, while downregulating the expression of MFN2 protein and PGC1α gene. Oil Red O staining demonstrated that H2O2 induced significant lipid accumulation in hepatocytes. Concurrently, H2O2 significantly increased serum triglycerides, total cholesterol, and liver triglycerides levels while decreasing serum hepatic lipase activity. This was primarily attributed to the significant upregulation of liver SREBP1, FASN, and ACC genes and the downregulation of the liver CPT1 gene induced by H2O2. Furthermore, CGA pretreatment effectively prevented the degeneration in laying performance and egg quality, as well as OS, liver inflammation, pathological damage, and mitochondrial dysfunction induced by H2O2. CGA inhibited H2O2-induced hepatic lipid accumulation by upregulating fatty acid oxidation-related gene expression and downregulating fatty acid synthesis-related gene expression. These findings indicate that the dietary addition of 800 mg/kg of CGA is the optimum supplementation dose. CGA can enhance laying performance and egg quality while alleviating OS, hepatic inflammation, mitochondrial dysfunction, and lipid accumulation in H2O2-challenged laying hens.
Collapse
Affiliation(s)
- Haitong Zhao
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.Z.); (Z.L.); (Y.S.); (M.Y.); (Y.W.); (Y.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Zhuang Li
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.Z.); (Z.L.); (Y.S.); (M.Y.); (Y.W.); (Y.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yue Sun
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.Z.); (Z.L.); (Y.S.); (M.Y.); (Y.W.); (Y.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Ming Yan
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.Z.); (Z.L.); (Y.S.); (M.Y.); (Y.W.); (Y.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yingjie Wang
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.Z.); (Z.L.); (Y.S.); (M.Y.); (Y.W.); (Y.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yurong Li
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.Z.); (Z.L.); (Y.S.); (M.Y.); (Y.W.); (Y.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yeshun Zhang
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.Z.); (Z.L.); (Y.S.); (M.Y.); (Y.W.); (Y.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Mingkun Zhu
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.Z.); (Z.L.); (Y.S.); (M.Y.); (Y.W.); (Y.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| |
Collapse
|
5
|
Zhao J, Liu L, Cao YY, Gao X, Targher G, Byrne CD, Sun DQ, Zheng MH. MAFLD as part of systemic metabolic dysregulation. Hepatol Int 2024; 18:834-847. [PMID: 38594474 DOI: 10.1007/s12072-024-10660-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/11/2024] [Indexed: 04/11/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases worldwide. In recent years, a new terminology and definition of metabolic dysfunction-associated fatty liver disease (MAFLD) has been proposed. Compared to the NAFLD definition, MAFLD better emphasizes the pathogenic role of metabolic dysfunction in the development and progression of this highly prevalent condition. Metabolic disorders, including overweight/obesity, type 2 diabetes mellitus (T2DM), atherogenic dyslipidemia and hypertension, are often associated with systemic organ dysfunctions, thereby suggesting that multiple organ damage can occur in MAFLD. Substantial epidemiological evidence indicates that MAFLD is not only associated with an increased risk of liver-related complications, but also increases the risk of developing several extra-hepatic diseases, including new-onset T2DM, adverse cardiovascular and renal outcomes, and some common endocrine diseases. We have summarized the current literature on the adverse effect of MAFLD on the development of multiple extrahepatic (cardiometabolic and endocrine) complications and examined the role of different metabolic pathways and organ systems in the progression of MAFLD, thus providing new insights into the role of MAFLD as a multisystem metabolic disorder. Our narrative review aimed to provide insights into potential mechanisms underlying the known associations between MAFLD and extrahepatic diseases, as part of MAFLD as a multisystem disease, in order to help focus areas for future drug development targeting not only liver disease but also the risk of extrahepatic complications.
Collapse
Affiliation(s)
- Jing Zhao
- Urologic Nephrology Center, Jiangnan University Medical Center, Wuxi, China
- Affiliated Wuxi Clinical College of Nantong University, Wuxi, China
- Wuxi No. 2 People's Hospital, Wuxi, China
| | - Lu Liu
- Urologic Nephrology Center, Jiangnan University Medical Center, Wuxi, China
- Affiliated Wuxi Clinical College of Nantong University, Wuxi, China
- Wuxi No. 2 People's Hospital, Wuxi, China
| | - Ying-Ying Cao
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, Zhejiang, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Christopher D Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton, and University of Southampton, Southampton General Hospital, Southampton, UK
| | - Dan-Qin Sun
- Urologic Nephrology Center, Jiangnan University Medical Center, Wuxi, China.
- Affiliated Wuxi Clinical College of Nantong University, Wuxi, China.
- Wuxi No. 2 People's Hospital, Wuxi, China.
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, Zhejiang, China.
| |
Collapse
|
6
|
Li W, Zhang Y, Yang J, Xu H, Ye R, Wu J, Cao M, Zhao C, Yang B, Liu C, Li L. Effect of Bile Acids Supplementation in Fatty Liver Hemorrhagic Syndrome, Production Performance, Physiological and Quality Characteristics of Laying Hen Eggs. Animals (Basel) 2024; 14:1910. [PMID: 38998024 PMCID: PMC11240722 DOI: 10.3390/ani14131910] [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: 05/27/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
This study aimed to investigate the effects of bile acids (BAs) supplementation on fatty liver hemorrhagic syndrome (FLHS), production performance, and physiological and quality characteristics of laying hen eggs. Sixty Sanhuang laying hens, aged 28 weeks, were randomly allocated to six dietary treatments over a 4-week period, including the control (CON) group (feeding basal diet), the high-fat diet (HFD)-treated group (basal diet containing 10% soybean oil), and HFD supplemented with 0.01% and 0.02% of chenodeoxycholic acid (CDCA) or hyodeoxycholic acid (HDCA) groups. Production performance, egg quality, liver morphology, serum biochemical indexes, antioxidant capacity, proinflammatory cytokines, and intestinal microbiota were evaluated. The average body weight in 0.01% CDCA was larger than in the HFD group (p < 0.05). Eggshell Thickness in the CON group was greater than in the HFD, 0.01% CDCA, and HDCA groups (p < 0.05). Albumen height in the 0.02% HDCA group was higher than the HFD group (p < 0.05). Eggshell weight in the HFD group was less than the CON group (p < 0.05). Haugh unit (HU) in the HDCA group was larger than the HFD group (p < 0.05). Albumen weight in the 0.02% HDCA group was greater than the CON and HFD groups (p < 0.05). In the HFD group, the levels of triglyceride (TG), total cholesterol (TC), and low-density lipo-protein cholesterol (LDL-C) were surpassing the other groups (p < 0.05). The levels of catalase (CAT) and total superoxide dismutase (T-SOD) in the HFD group was smaller than the other groups (p < 0.05). The level of malondialdehyde (MDA) in the HFD group was higher than in the other groups (p < 0.05). Tumor necrosis factor-α (TNF-α) levels were larger in the HFD group than in the other groups (p < 0.05). The 16S rRNA sequencing analysis indicated significant variations in the relative abundance of specific bacterial populations among the different treatment groups. The treatment and CON groups exhibited a higher presence of bacteria that inhibit host energy absorption or promote intestinal health such as Firmicutes, Bacteroidetes, and Ruminococcus, whereas the HFD group showed an increased prevalence of potentially pathogenic or deleterious bacteria, such as Desulfovibrio spp. In conclusion, the supplementation of BAs in poultry feed has been demonstrated to effectively mitigate the detrimental effects of FLHS in laying hens. This intervention regulates lipid metabolism, bolsters antioxidant defenses, reduces inflammation, and modulates the gut microbiota, offering a novel perspective on the application of BAs in the poultry industry.
Collapse
Affiliation(s)
- Wen Li
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (W.L.); (Y.Z.); (J.Y.); (H.X.); (R.Y.); (J.W.); (M.C.); (C.Z.); (B.Y.)
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang 233100, China
| | - Yu Zhang
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (W.L.); (Y.Z.); (J.Y.); (H.X.); (R.Y.); (J.W.); (M.C.); (C.Z.); (B.Y.)
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang 233100, China
| | - Jingyi Yang
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (W.L.); (Y.Z.); (J.Y.); (H.X.); (R.Y.); (J.W.); (M.C.); (C.Z.); (B.Y.)
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang 233100, China
| | - Hao Xu
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (W.L.); (Y.Z.); (J.Y.); (H.X.); (R.Y.); (J.W.); (M.C.); (C.Z.); (B.Y.)
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang 233100, China
| | - Ruiqi Ye
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (W.L.); (Y.Z.); (J.Y.); (H.X.); (R.Y.); (J.W.); (M.C.); (C.Z.); (B.Y.)
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang 233100, China
| | - Jiale Wu
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (W.L.); (Y.Z.); (J.Y.); (H.X.); (R.Y.); (J.W.); (M.C.); (C.Z.); (B.Y.)
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang 233100, China
| | - Mixia Cao
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (W.L.); (Y.Z.); (J.Y.); (H.X.); (R.Y.); (J.W.); (M.C.); (C.Z.); (B.Y.)
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang 233100, China
- Key Laboratory of Quality & Safety Control for Pork, Ministry of Agriculture and Rural, Fengyang 233100, China
| | - Chunfang Zhao
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (W.L.); (Y.Z.); (J.Y.); (H.X.); (R.Y.); (J.W.); (M.C.); (C.Z.); (B.Y.)
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang 233100, China
- Key Laboratory of Quality & Safety Control for Pork, Ministry of Agriculture and Rural, Fengyang 233100, China
| | - Bing Yang
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (W.L.); (Y.Z.); (J.Y.); (H.X.); (R.Y.); (J.W.); (M.C.); (C.Z.); (B.Y.)
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang 233100, China
- Key Laboratory of Quality & Safety Control for Pork, Ministry of Agriculture and Rural, Fengyang 233100, China
| | - Chang Liu
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (W.L.); (Y.Z.); (J.Y.); (H.X.); (R.Y.); (J.W.); (M.C.); (C.Z.); (B.Y.)
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang 233100, China
- Key Laboratory of Quality & Safety Control for Pork, Ministry of Agriculture and Rural, Fengyang 233100, China
| | - Lei Li
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (W.L.); (Y.Z.); (J.Y.); (H.X.); (R.Y.); (J.W.); (M.C.); (C.Z.); (B.Y.)
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Fengyang 233100, China
- Key Laboratory of Quality & Safety Control for Pork, Ministry of Agriculture and Rural, Fengyang 233100, China
| |
Collapse
|
7
|
Zhu FL, Huang T, Lv ZL, Liang G, Yao Z, Lan LC, Qadir A, Chen XQ, Shan QW. Taurine Regulates the Expression of Interleukin -17/10 and Intestinal Flora and Protects the Liver and Intestinal Mucosa in a Nonalcoholic Fatty Liver Disease Rat Model. Diabetes Metab Syndr Obes 2024; 17:675-689. [PMID: 38352234 PMCID: PMC10863462 DOI: 10.2147/dmso.s440978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
Purpose To investigate the intestinal inflammatory response and the abundance of intestinal bacteria in rats with high-fat diet (HFD)-induced nonalcoholic fatty liver disease (NAFLD) and assess the intervention effects of taurine (TAU). Methods Forty male Sprague-Dawley rats were randomly divided into five groups: group I, normal diet and normal saline gavage; group II, normal diet and TAU gavage; group III, HFD and normal saline gavage; group IV, HFD and TAU gavage (from the 1st week); group V, HFD and TAU gavage (from the 10th week). At the end of the 16th week, all the animals were sacrificed. Body weight, liver weight, liver function, and serum lipid levels were measured. The histopathologies of the liver and ileum were observed. The mRNA and protein expression levels of interleukin 17 (IL-17) and IL-10 in the ileum were detected by reverse transcription quantitative polymerase chain reaction (qPCR) and immunohistochemistry. Three types of bacteria were detected in intestinal feces using the 16S rDNA qPCR method. Results The ileal IL-17 level in group III was significantly higher than those in the other four groups (P < 0.01). The ileal IL-10 mRNA levels in group IV was significantly higher than those in groups III and V (P < 0.05), and IL-10 protein MOD levels in group III was significantly lower than those in the other four groups (P < 0.01). The numbers of Lactobacillus in group III were significantly lower than those in the other four groups (P < 0.01 or P < 0.05). The numbers of Bifidobacteria in groups IV and V were significantly increased compared with that in group III (P < 0.05). Conclusion TAU may down-regulate the expression of IL-17, up-regulate the expression of IL-10 and regulate the intestinal flora, and alleviate the liver and intestinal damage in rats with HFD-induced NAFLD.
Collapse
Affiliation(s)
- Fu-Li Zhu
- Department of Pediatrics, the First College of Clinical Medical Science, China Three Gorges University, Yichang Central People’s Hospital, Yichang, Hubei, 443000, People’s Republic of China
- Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Ting Huang
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Science, Nanning, Guangxi, 530021, People’s Republic of China
| | - Zi-Li Lv
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Gang Liang
- Department of Pathophysiology, School of Basic Medical Sciences of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Zhen Yao
- Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Lian-Cheng Lan
- Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Abdul Qadir
- Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xiu-Qi Chen
- Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Qing-Wen Shan
- Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| |
Collapse
|
8
|
Miao S, Mu T, Li R, Li Y, Zhao W, Li J, Dong X, Zou X. Coated sodium butyrate ameliorates high-energy and low-protein diet induced hepatic dysfunction via modulating mitochondrial dynamics, autophagy and apoptosis in laying hens. J Anim Sci Biotechnol 2024; 15:15. [PMID: 38302976 PMCID: PMC10835823 DOI: 10.1186/s40104-023-00980-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/17/2023] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Fatty liver hemorrhagic syndrome (FLHS), a fatty liver disease in laying hens, poses a grave threat to the layer industry, stemming from its ability to trigger an alarming plummet in egg production and usher in acute mortality among laying hens. Increasing evidence suggests that the onset and progression of fatty liver was closely related to mitochondria dysfunction. Sodium butyrate was demonstrated to modulate hepatic lipid metabolism, alleviate oxidative stress and improve mitochondrial dysfunction in vitro and mice models. Nevertheless, there is limited existing research on coated sodium butyrate (CSB) to prevent FLHS in laying hens, and whether and how CSB exerts the anti-FLHS effect still needs to be explored. In this experiment, the FLHS model was induced by administering a high-energy low-protein (HELP) diet in laying hens. The objective was to investigate the effects of CSB on alleviating FLHS with a focus on the role of CSB in modulating mitochondrial function. METHODS A total of 288 healthy 28-week-old Huafeng laying hens were arbitrarily allocated into 4 groups with 6 replicates each, namely, the CON group (normal diet), HELP group (HELP diet), CH500 group (500 mg/kg CSB added to HELP diet) and CH750 group (750 mg/kg CSB added to HELP diet). The duration of the trial encompassed a period of 10 weeks. RESULTS The result revealed that CSB ameliorated the HELP-induced FLHS by improving hepatic steatosis and pathological damage, reducing the gene levels of fatty acid synthesis, and promoting the mRNA levels of key enzymes of fatty acid catabolism. CSB reduced oxidative stress induced by the HELP diet, upregulated the activity of GSH-Px and SOD, and decreased the content of MDA and ROS. CSB also mitigated the HELP diet-induced inflammatory response by blocking TNF-α, IL-1β, and F4/80. In addition, dietary CSB supplementation attenuated HELP-induced activation of the mitochondrial unfolded protein response (UPRmt), mitochondrial damage, and decline of ATPase activity. HELP diet decreased the autophagosome formation, and downregulated LC3B but upregulated p62 protein expression, which CSB administration reversed. CSB reduced HELP-induced apoptosis, as indicated by decreases in the Bax/Bcl-2, Caspase-9, Caspase-3, and Cyt C expression levels. CONCLUSIONS Dietary CSB could ameliorate HELP diet-induced hepatic dysfunction via modulating mitochondrial dynamics, autophagy, and apoptosis in laying hens. Consequently, CSB, as a feed additive, exhibited the capacity to prevent FLHS by modulating autophagy and lipid metabolism.
Collapse
Affiliation(s)
- Sasa Miao
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Tianming Mu
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ru Li
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yan Li
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wenyan Zhao
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jiankui Li
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xinyang Dong
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoting Zou
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
9
|
Cheng X, Hu Y, Yu X, Chen J, Guo X, Cao H, Hu G, Zhuang Y. Sodium Butyrate Alleviates Free Fatty Acid-Induced Steatosis in Primary Chicken Hepatocytes via Regulating the ROS/GPX4/Ferroptosis Pathway. Antioxidants (Basel) 2024; 13:140. [PMID: 38397738 PMCID: PMC10886346 DOI: 10.3390/antiox13020140] [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: 12/21/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Fatty liver hemorrhagic syndrome (FLHS) in laying hens is a nutritional metabolic disease commonly observed in high-yielding laying hens. Sodium butyrate (NaB) and ferroptosis were reported to contribute to the pathogenesis of fatty liver-related diseases. However, the underlying mechanism of NaB in FLHS and whether it mediates ferroptosis remains unclear. A chicken primary hepatocyte induced by free fatty acids (FFAs, keeping the ratio of sodium oleate and sodium palmitate concentrations at 2:1) was established, which received treatments with NaB, the ferroptosis inducer RAS-selective lethal 3 (RSL3), and the inhibitor ferrostatin-1 (Fer-1). As a result, NaB increased biochemical and lipid metabolism indices, and the antioxidant level, while inhibiting intracellular ROS accumulation and the activation of the ferroptosis signaling pathway, as evidenced by a reduction in intracellular iron concentration, upregulated GPX4 and xCT expression, and inhibited NCOA4 and ACSL4 expression. Furthermore, treatment with Fer-1 reinforced the protective effects of NaB, while RSL3 reversed it by blocking the ROS/GPX4/ferroptosis pathway, leading to the accumulation of lipid droplets and oxidative stress. Collectively, our findings demonstrated that NaB protects hepatocytes by regulating the ROS/GPX4-mediated ferroptosis pathway, providing a new strategy and target for the treatment of FLHS.
Collapse
Affiliation(s)
- Xinyi Cheng
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Yang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Xiaoqing Yu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Jinyan Chen
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Huabin Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| | - Yu Zhuang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, China
| |
Collapse
|