1
|
Gao Z, Gao Y, Wang X, Wang T, Zhou H, Liu C, Mai K, He G. Administration of EPA improves lipopolysaccharide-induced intestinal dysfunction in turbot (Scophthalmus maximus L.) through modulation of TORC1 signaling. FISH & SHELLFISH IMMUNOLOGY 2025; 163:110391. [PMID: 40345275 DOI: 10.1016/j.fsi.2025.110391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/20/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
Intestinal health is crucial for digestive and absorptive functions, which are associated with fish growth performance. Searching for nutraceuticals and bioactive ingredients to improve intestinal health has become urgent for the aquaculture industry. In the present study, the effects of eicosapentaenoic acid (EPA) on intestinal function were investigated in turbot with induced intestinal damage caused by lipopolysaccharide (LPS). Juvenile turbot (initial weight 100 ± 5 g) were subjected to a 10-day enforced feeding regimen. Each fish was fed twice daily with either a 100 mg amino acid mixture (CON), an additional 1 mg of LPS (LPS), or an additional combination of 1 mg LPS and 1.7 mg EPA (LE). The results indicated that EPA supplementation restored intestinal villi integrity and the mucosal barrier. The number of goblet cells and the gene expression of MUC-2 were increased by EPA. Additionally, EPA suppressed the expression of inflammatory factors (MyD88 and NF-κB p65) and pro-inflammatory cytokines (TNFα, IL-1β, and IFN-γ). Meanwhile, post-feeding plasma free amino acid levels as well as intestinal protein synthesis were improved by EPA supplementation. The target of rapamycin (TOR) signaling pathway was significantly activated by EPA. Furthermore, EPA supplementation positively influenced intestinal microbiota, reducing the abundance of prevalent pathogens while enhancing the abundance of probiotics. Collectively, the administration of EPA effectively mitigates LPS-induced damage to the intestinal barrier, inflammatory response, and dysbiosis of microbiota through modulation of the TOR signaling pathway.
Collapse
Affiliation(s)
- Zongyu Gao
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Ya Gao
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Xuan Wang
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Tingting Wang
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Huihui Zhou
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Chengdong Liu
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Kangsen Mai
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Gen He
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
2
|
Wang F, Wang X, Odle J, Maltecca C, Lin X. Maternal Supplementation of Dietary Choline and DHA During Gestational Nutrition Restriction Alters Hepatic mRNA and miRNA Expression Patterns in Full-Term Fetal Pigs. J Nutr 2025; 155:804-816. [PMID: 39805404 DOI: 10.1016/j.tjnut.2025.01.012] [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: 08/26/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Supplementing choline and DHA to pregnant gilts modified fetal pig hepatic global DNA methylation induced by gestational malnutrition, suggesting that gene expression and regulation and its associated metabolic pathways are affected in the liver of offspring during growth and development. OBJECTIVES This study aimed to investigate the effect of maternal supplementation of choline, DHA, and their interaction on hepatic mRNA expression, miRNA regulation, and metabolic pathways in the fetal pigs born to malnourished mothers. METHODS The abundance of mRNA and miRNA was profiled in fetal liver from sows with undernutrition supplemented with choline and DHA in a 2 × 2 factorial design. The effects of choline, DHA, and their interaction on mRNA and miRNA expression were evaluated. Identification of the Biological Processes from the Gene Ontology database and miRNA Target Prediction Analysis were performed using the DAVID Functional Annotation Tool and Ingenuity Pathway Analysis. The identified miRNA-mRNA pairings were validated using RT-qPCR. RESULTS In total, 144 mRNA and 1 miRNA were altered by supplementation of choline, and the alterations were associated with the inhibitions of cardiac hypertrophy signaling, IL-6 signaling, IL-3 signaling, the Th1 pathway, and the acute phase response signaling pathway. Further, 151 mRNAs and 6 miRNAs were altered by maternal supplementation DHA and were associated with inhibition of 5 inositol-related pathways, 5 immune-related pathways, and 7 other pathways and the stimulation of peroxisome proliferator-activated receptor signaling and RhoGDI signaling pathways. In addition, 383 mRNAs and 25 miRNAs displayed choline × DHA interactions including synergistic effects on acute phase response signaling, and antagonistic effects on tRNA splicing, peroxisome proliferator-activated receptor α/retinoid X receptor α activation, and sirtuin signaling, NAD signaling, and RNA polymerase I transcription pathways. Ten of the identified 20 miRNA-mRNA pairings were validated using RT-qPCR. CONCLUSIONS The supplementation of choline, DHA, or choline plus DHA to pregnant gilts modifies liver mRNA, miRNA, and pathways in fetal pigs during gestational undernutrition.
Collapse
Affiliation(s)
- Feng Wang
- Department of Animal Sciences, North Carolina State University, Raleigh, NC, United States
| | - Xiaoqiu Wang
- Department of Animal Sciences, North Carolina State University, Raleigh, NC, United States
| | - Jack Odle
- Department of Animal Sciences, North Carolina State University, Raleigh, NC, United States
| | - Christian Maltecca
- Department of Animal Sciences, North Carolina State University, Raleigh, NC, United States
| | - Xi Lin
- Department of Animal Sciences, North Carolina State University, Raleigh, NC, United States.
| |
Collapse
|
3
|
Yi K, An L, Qi Y, Yang T, Duan Y, Zhao X, Zhang P, Huang X, Su X, Tang Z, Sun D. Docosahexaenoic acid (DHA) promotes recovery from postoperative ileus and the repair of the injured intestinal barrier through mast cell-nerve crosstalk. Int Immunopharmacol 2024; 136:112316. [PMID: 38823183 DOI: 10.1016/j.intimp.2024.112316] [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: 02/11/2024] [Revised: 03/31/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
The objective of this study was to investigate the neuroimmune mechanisms implicated in the enhancement of gastrointestinal function through the administration of oral DHA. Mast cell-deficient mice (KitW-sh) and C57BL/6 mice were used to establish postoperative ileus (POI) models. To further validate our findings, we conducted noncontact coculture experiments involving dorsal root ganglion (DRG) cells, bone marrow-derived mast cells (BMMCs) and T84 cells. Furthermore, the results obtained from investigations conducted on animals and cells were subsequently validated through clinical trials. The administration of oral DHA had ameliorative effects on intestinal barrier injury and postoperative ileus. In a mechanistic manner, the anti-inflammatory effect of DHA was achieved through the activation of transient receptor potential ankyrin 1 (TRPA1) on DRG cells, resulting in the stabilization of mast cells and increasing interleukin 10 (IL-10) secretion in mast cells. Furthermore, the activation of the pro-repair WNT1-inducible signaling protein 1 (WISP-1) signaling pathways by mast cell-derived IL-10 resulted in an enhancement of the intestinal barrier integrity. The current study demonstrated that the neuroimmune interaction between mast cells and nerves played a crucial role in the process of oral DHA improving the intestinal barrier integrity of POI, which further triggered the activation of CREB/WISP-1 signaling in intestinal mucosal cells.
Collapse
Affiliation(s)
- Keqian Yi
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Liya An
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Yuxing Qi
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Ting Yang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Yongqing Duan
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Xiaohu Zhao
- Department of Medicine, Monash University, Clayton, Victoria 3168, Australia
| | - Pengcheng Zhang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Xingzong Huang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Xianming Su
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Zhiyi Tang
- Department of Gastroenterology, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China.
| | - Dali Sun
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China.
| |
Collapse
|
4
|
Li X, Zhang H, Mao X. Liposomes delivery systems of functional substances for precision nutrition. ADVANCES IN FOOD AND NUTRITION RESEARCH 2024; 112:257-300. [PMID: 39218504 DOI: 10.1016/bs.afnr.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Natural bioactive compounds with antioxidant, antimicrobial, anticancer, and other biological activities are vital for maintaining the body's physiological functions and enhancing immunity. These compounds have great potential as nutritional therapeutic agents, but they can be limited due to their poor flavor, color, unstable nature, and poor water solubility, and degradation by gastrointestinal enzymes. Liposomes, as ideal carriers, can encapsulate both water-soluble and fat-soluble nutrients, enhance the bioavailability of functional substances, promote the biological activity of functional substances, and control the release of nutrients. Despite their potential, liposomes still face obstacles in nutrient delivery. Therefore, the design of liposomes for special needs, optimization of the liposome preparation process, enhancement of liposome encapsulation efficiency, and industrial production are key issues that must be addressed in order to develop food-grade liposomes. Moreover, the research on surface-targeted modification and surface functionalization of liposomes is valuable for expanding the scope of application of liposomes and achieving the release of functional substances from liposomes at the appropriate time and site. The establishment of in vivo and in vitro digestion models of nutrient-loaded liposomes, in-depth study of gastrointestinal digestive behavior after liposome ingestion, targeted nutrient release, and deciphering the nutritional intervention of human diseases and positive health promotion are promising fields with broad development prospects.
Collapse
Affiliation(s)
- Xuehan Li
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, P.R. China; Qingdao Key Laboratory of Food Biotechnology, Qingdao, P.R. China; Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao, P.R. China
| | - Haiyang Zhang
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, P.R. China; Qingdao Key Laboratory of Food Biotechnology, Qingdao, P.R. China; Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao, P.R. China
| | - Xiangzhao Mao
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, P.R. China; Qingdao Key Laboratory of Food Biotechnology, Qingdao, P.R. China; Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao, P.R. China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, P.R. China.
| |
Collapse
|
5
|
Tian A, Zheng Y, Li H, Zhang Z, Du L, Huang X, Sun L, Wu H. Eicosapentaenoic acid activates the P62/KEAP1/NRF2 pathway for the prevention of diabetes-associated cognitive dysfunction. Food Funct 2024; 15:5251-5271. [PMID: 38680120 DOI: 10.1039/d4fo00774c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Diabetes-associated cognitive dysfunction (DCD) is a severe complication of diabetes mellitus (DM), threatening the life quality of the diabetic population. However, there is still a lack of effective approaches for its intervention. Eicosapentaenoic acid (EPA) is an omega-3 polyunsaturated fatty acid that was not previously investigated for its effect on DCD. In this study, EPA was found to improve DCD in a mouse model of type 2 DM (T2DM) induced by streptozotocin and a high-fat diet, exhibiting profound protective effects on cognitive dysfunction, neuronal loss, and cerebral oxidative stress and inflammation. While EPA did not attenuate advanced glycation end product-induced neuron injury, we hypothesized that EPA might protect neurons by regulating microglia polarization, the effect of which was confirmed by the co-culture of neurons and lipopolysaccharide-stimulated microglia. RNA sequencing identified nuclear factor-erythroid-2-related factor 2 (NRF2) antioxidant signaling as a major target of EPA in microglia. Mechanistically, EPA increased sequestosome-1 (SQSTM1 or P62) levels that might structurally inhibit Kelch-like ECH associated protein 1 (KEAP1), leading to nuclear translocation of NRF2. P62 and NRF2 predominantly mediated EPA's effect since the knockdown of P62 or NRF2 abolished EPA's protective effect on microglial oxidative stress and inflammation and sequential neuron injuries. Moreover, the regulation of P62/KEPA1/NRF2 axes by EPA was confirmed in the hippocampi of diabetic mice. The present work presents EPA as an effective nutritional approach and microglial P62/KEAP1/NRF2 as molecular targets for the intervention of DCD.
Collapse
Affiliation(s)
- Ao Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| | - Yan Zheng
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Hui Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Zhiyue Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Lei Du
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| | - Xiaoli Huang
- Department of Nutrition, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Rd., Jinan, Shandong 250012, China.
| | - Lei Sun
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Rd., Jinan, Shandong 250012, China.
| | - Hao Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| |
Collapse
|
6
|
Liu Y, Robinson AM, Su XQ, Nurgali K. Krill Oil and Its Bioactive Components as a Potential Therapy for Inflammatory Bowel Disease: Insights from In Vivo and In Vitro Studies. Biomolecules 2024; 14:447. [PMID: 38672464 PMCID: PMC11048140 DOI: 10.3390/biom14040447] [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: 12/26/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Krill oil is extracted from krill, a small crustacean in the Antarctic Ocean. It has received growing attention because of krill oil's unique properties and diverse health benefits. Recent experimental and clinical studies suggest that it has potential therapeutic benefits in preventing the development of a range of chronic conditions, including inflammatory bowel disease (IBD). Krill oil is enriched with long-chain n-3 polyunsaturated fatty acids, especially eicosapentaenoic and docosahexaenoic acids, and the potent antioxidant astaxanthin, contributing to its therapeutic properties. The possible underlying mechanisms of krill oil's health benefits include anti-inflammatory and antioxidant actions, maintaining intestinal barrier functions, and modulating gut microbiota. This review aims to provide an overview of the beneficial effects of krill oil and its bioactive components on intestinal inflammation and to discuss the findings on the molecular mechanisms associated with the role of krill oil in IBD prevention and treatment.
Collapse
Affiliation(s)
- Yingying Liu
- Institute for Health & Sport, Victoria University, Melbourne, VIC 3021, Australia; (Y.L.); (A.M.R.)
| | - Ainsley M. Robinson
- Institute for Health & Sport, Victoria University, Melbourne, VIC 3021, Australia; (Y.L.); (A.M.R.)
- School of Rural Health, La Trobe University, Melbourne, VIC 3010, Australia
- Department of Medicine Western Health, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Xiao Qun Su
- Institute for Health & Sport, Victoria University, Melbourne, VIC 3021, Australia; (Y.L.); (A.M.R.)
| | - Kulmira Nurgali
- Institute for Health & Sport, Victoria University, Melbourne, VIC 3021, Australia; (Y.L.); (A.M.R.)
- Department of Medicine Western Health, The University of Melbourne, Melbourne, VIC 3010, Australia
- Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| |
Collapse
|
7
|
Ge J, Ye L, Cheng M, Xu W, Chen Z, Guan F. Preparation of microgels loaded with lycopene/NMN and their protective mechanism against acute liver injury. Food Funct 2024; 15:809-822. [PMID: 38131354 DOI: 10.1039/d3fo03293k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
This study aimed to enhance the stability and bioavailability of lycopene (LYC) and nicotinamide mononucleotide (NMN) by incorporating them into porous microgels after loading LYC into liposomes. The particle size, zeta potential, encapsulation rate (%), scanning electron microscopy images, and stability and release kinetics characteristics in simulating digestion confirmed that the microgels had high LYC and NMN encapsulation rates (99.11% ± 0.12% and 68.98% ± 0.26%, respectively) and good stability and release characteristics. The protective effect and potential mechanism of microgels loaded with LYC and NMN on lipopolysaccharide (LPS)-induced acute liver injury in C57BL/6 mice were investigated by intragastric administration for 28 days prior to LPS exposure. The results showed that the microgels loaded with LYC and NMN significantly ameliorated LPS-induced liver injury and reduced the inflammatory response and oxidative stress. In addition, LYC and NMN can not only act on the Toll-like receptor 4 (TLR4)/MD2 complex but also regulate TLR4-related miRNAs (miR-145a-5p and miR-217-5p) in serum extracellular vesicles, thereby synergistically inhibiting the TLR4/NF-κB signaling pathway. In addition, the microgels loaded with LYC and NMN were able to enrich beneficial bacteria that produced short-chain fatty acids and reduce harmful bacteria. In conclusion, LYC and NMN protected against LPS-induced acute liver injury via inhibition of oxidative stress and inflammation, as well as regulating the gut microbiota.
Collapse
Affiliation(s)
- Jian Ge
- College of Life Sciences, China Jiliang University, 258 XueYuan Street, XiaSha Higher Education Zone, Hangzhou, 310018, Zhejiang Province, People's Republic of China.
| | - Luting Ye
- College of Life Sciences, China Jiliang University, 258 XueYuan Street, XiaSha Higher Education Zone, Hangzhou, 310018, Zhejiang Province, People's Republic of China.
| | - Min Cheng
- College of Life Sciences, China Jiliang University, 258 XueYuan Street, XiaSha Higher Education Zone, Hangzhou, 310018, Zhejiang Province, People's Republic of China.
| | - Weijia Xu
- College of Life Sciences, China Jiliang University, 258 XueYuan Street, XiaSha Higher Education Zone, Hangzhou, 310018, Zhejiang Province, People's Republic of China.
| | - Zhaowen Chen
- College of Life Sciences, China Jiliang University, 258 XueYuan Street, XiaSha Higher Education Zone, Hangzhou, 310018, Zhejiang Province, People's Republic of China.
| | - Feng Guan
- College of Life Sciences, China Jiliang University, 258 XueYuan Street, XiaSha Higher Education Zone, Hangzhou, 310018, Zhejiang Province, People's Republic of China.
| |
Collapse
|
8
|
Seethaler B, Lehnert K, Yahiaoui-Doktor M, Basrai M, Vetter W, Kiechle M, Bischoff SC. Omega-3 polyunsaturated fatty acids improve intestinal barrier integrity-albeit to a lesser degree than short-chain fatty acids: an exploratory analysis of the randomized controlled LIBRE trial. Eur J Nutr 2023; 62:2779-2791. [PMID: 37318580 PMCID: PMC10468946 DOI: 10.1007/s00394-023-03172-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/04/2023] [Indexed: 06/16/2023]
Abstract
PURPOSE Adherence to the Mediterranean diet is associated with beneficial health effects, including gastrointestinal disorders. Preclinical studies suggest that omega-3 polyunsaturated fatty acids (n-3 PUFAs), found in Mediterranean foods like nuts and fish, improve intestinal barrier integrity. Here, we assessed possible effects of n-3 PUFAs on barrier integrity in a randomized controlled trial. METHODS We studied 68 women from the open-label LIBRE trial (clinicaltrials.gov: NCT02087592) who followed either a Mediterranean diet (intervention group, IG) or a standard diet (control group, CG). Study visits comprised baseline, month 3, and month 12. Barrier integrity was assessed by plasma lipopolysaccharide binding protein (LBP) and fecal zonulin; fatty acids by gas chromatography with mass spectrometry. Median and interquartile ranges are shown. RESULTS Adherence to the Mediterranean diet increased the proportion of the n-3 docosahexaenoic acid (DHA) (IG + 1.5% [0.9;2.5, p < 0.001]/ + 0.3% [- 0.1;0.9, p < 0.050] after 3/12 months; CG + 0.9% [0.5;1.6, p < 0.001]/ ± 0%) and decreased plasma LBP (IG - 0.3 µg/ml [- 0.6;0.1, p < 0.010]/ - 0.3 µg/ml [- 1.1; - 0.1, p < 0.001]; CG - 0.2 µg/ml [- 0.8; - 0.1, p < 0.001]/ ± 0 µg/ml) and fecal zonulin levels (IG - 76 ng/mg [- 164; - 12, p < 0.010]/ - 74 ng/mg [- 197;15, p < 0.001]; CG - 59 ng/mg [- 186;15, p < 0.050]/ + 10 ng/mg [- 117;24, p > 0.050]). Plasma DHA and LBP (R2: 0.14-0.42; all p < 0.070), as well as plasma DHA and fecal zonulin (R2: 0.18-0.48; all p < 0.050) were found to be inversely associated in bi- and multivariate analyses. Further multivariate analyses showed that the effect of DHA on barrier integrity was less pronounced than the effect of fecal short-chain fatty acids on barrier integrity. CONCLUSIONS Our data show that n-3 PUFAs can improve intestinal barrier integrity. TRIAL REGISTRATION NUMBER The trial was registered prospectively at ClinicalTrials.gov (reference: NCT02087592).
Collapse
Affiliation(s)
- Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Fruwirthstr. 12, 70593, Stuttgart, Germany
| | - Katja Lehnert
- Institute of Food Chemistry, University of Hohenheim, Stuttgart, Germany
| | - Maryam Yahiaoui-Doktor
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
| | - Maryam Basrai
- Institute of Nutritional Medicine, University of Hohenheim, Fruwirthstr. 12, 70593, Stuttgart, Germany
| | - Walter Vetter
- Institute of Food Chemistry, University of Hohenheim, Stuttgart, Germany
| | - Marion Kiechle
- Department of Gynecology, Center for Hereditary Breast and Ovarian Cancer, Klinikum Rechts der Isar, Technical University Munich and Comprehensive Cancer Center Munich, Munich, Germany
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Fruwirthstr. 12, 70593, Stuttgart, Germany.
| |
Collapse
|
9
|
Ma R, Wang S, Xue M, Zhang H, He Z, Jueraitetibaike K, Ge X, Chen L, Yao B. Effects of n-3 PUFA supplementation on oocyte in vitro maturation in mice with polycystic ovary syndrome. J Ovarian Res 2023; 16:87. [PMID: 37120599 PMCID: PMC10148539 DOI: 10.1186/s13048-023-01162-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/16/2023] [Indexed: 05/01/2023] Open
Abstract
n-3 PUFAs are classic antioxidant that can be used to treat follicular dysplasia and hyperinsulinemia caused by excessive oxidative stress in PCOS women. To investigate the effect of n-3 PUFA supplementation on the oocyte quality of polycystic ovary syndrome (PCOS) mice during in vitro maturation, a PCOS mouse model was established by dehydroepiandrosterone (DHEA). The GV oocytes of the control and PCOS groups were collected and cultured in vitro with or without n-3 PUFAs. After 14 h, the oocytes were collected. Our data demonstrated that the oocyte maturation rate of PCOS mice significantly increased after the addition of 50 µM n-3 PUFAs. The results of immunofluorescence showed that the abnormal rates of spindles and chromosomes in the PCOS + n-3 PUFA group were lower than those in the PCOS group. The mRNA expression of an antioxidant-related gene (Sirt1) and DNA damage repair genes (Brca1/Msh2) was found to be significantly rescued after n-3 treatment. Additionally, the results of living cell staining showed that the addition of n-3 PUFAs could reduce the levels of reactive oxygen species and mitochondrial superoxide in PCOS oocytes. In conclusion, the addition of 50 µM n-3 PUFAs during the in vitro maturation of PCOS mouse oocytes can improve the maturation rate by reducing the level of oxidative stress and the rate of spindle/chromosome abnormalities, providing valuable support during the IVM process.
Collapse
Affiliation(s)
- Rujun Ma
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China
- Center of Reproductive Medicine, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Shuxian Wang
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China
| | - Mengqi Xue
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China
- Center of Reproductive Medicine, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Hong Zhang
- Center of Reproductive Medicine, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Zhaowanyue He
- Center of Reproductive Medicine, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Kadiliya Jueraitetibaike
- Center of Reproductive Medicine, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Xie Ge
- Center of Reproductive Medicine, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Li Chen
- Center of Reproductive Medicine, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Bing Yao
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China.
- Center of Reproductive Medicine, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
10
|
Gao N, Yang Y, Liu S, Fang C, Dou X, Zhang L, Shan A. Gut-Derived Metabolites from Dietary Tryptophan Supplementation Quench Intestinal Inflammation through the AMPK-SIRT1-Autophagy Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:16080-16095. [PMID: 36521060 DOI: 10.1021/acs.jafc.2c05381] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Tryptophan has drawn wide attention due to its involvement in improving intestinal immune defense directly and indirectly by regulating metabolic pathways. The study aims to elucidate the potential modulating roles of tryptophan to protect against intestinal inflammation and elucidate the underlying molecular mechanisms. The protective effects of tryptophan against intestinal inflammation are examined in the lipopolysaccharide (LPS)-induced inflammatory model. We first found that tryptophan markedly (p < 0.01) inhibited proinflammatory cytokines production and nuclear factor κB (NF-κB) pathway activation upon LPS challenge. Next, we demonstrated that tryptophan (p < 0.05) attenuated LPS-caused intestinal mucosal barrier damage by increasing the number of goblet cells, mucins, and antimicrobial peptides (AMPs) in the ileum of mice. In addition, tryptophan (p < 0.05) inhibited LPS-induced autophagic flux through the AMP-activated protein kinase (AMPK)-sirtuin 1 (SIRT1) pathway in the intestinal systems to maintain autophagy homeostasis. Meanwhile, tryptophan also reshaped the gut microbiota composition in LPS-challenge mice by increasing the abundance of short-chain fatty acid (SCFA)-producing bacteria such as Acetivibrio (0.053 ± 0.017 to 0.21 ± 0.0041%). Notably, dietary tryptophan resulted in the activation of metabolic pathways during the inflammatory response. Furthermore, exogenous treatment of tryptophan metabolites kynurenine (Kyn) and 5-HT in porcine intestinal epithelial cells (IPEC-J2 cells) reproduced similar protective effects as tryptophan to attenuate LPS-induced intestinal inflammation through regulating the AMPK-SIRT1-autophagy. Taken together, the present study indicates that tryptophan exhibits intestinal protective and immunoregulatory effects resulting from the activation of metabolic pathways, maintenance of gut mucosal barrier integrity, microbiota composition, and AMPK-SIRT1-autophagy level.
Collapse
Affiliation(s)
- Nan Gao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yang Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Siqi Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Chunyang Fang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xiujing Dou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Licong Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
11
|
Wang CC, Kong JY, Li XY, Yang JY, Xue CH, Yanagita T, Wang YM. Antarctic krill oil exhibited synergistic effects with nobiletin and theanine in ameliorating memory and cognitive deficiency in SAMP8 mice: Applying the perspective of the sea–land combination to retard brain aging. Front Aging Neurosci 2022; 14:964077. [PMID: 36185487 PMCID: PMC9523088 DOI: 10.3389/fnagi.2022.964077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
The complex pathogenesis of Alzheimer's disease (AD) leads to a limited therapeutic effect; therefore, the combination of multiple bioactive ingredients may be more effective in improving AD due to synergistic effects. Based on the perspective of the sea–land combination, the effects of sea-derived Antarctic krill oil (AKO) combined with land-derived nobiletin (Nob) and L-theanine (The) on memory loss and cognitive deficiency were studied in senescence-accelerated prone 8 mice (SAMP8). The results demonstrated that AKO combined with The significantly increased the number of platform crossings in the Morris water maze test by 1.6-fold, and AKO combined with Nob significantly increased the preference index in a novel object recognition test. AKO exhibited synergistic effects with Nob and The in ameliorating recognition memory and spatial memory deficiency in SAMP8 mice, respectively. Further research of the mechanism indicated that AKO exhibited synergistic effects with Nob in suppressing β-amyloid (Aβ) aggregation, neurofibrillary tangles, and apoptosis and neuroinflammation, while the synergistic effects of AKO and The involved in synaptic plasticity and anti-neuroinflammation, which revealed that the combination was complex, not a mechanical addition. These findings revealed that the sea–land combination may be an effective strategy to treat and alleviate AD.
Collapse
Affiliation(s)
- Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Jing-Ya Kong
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Xiao-Yue Li
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Jin-Yue Yang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Teruyoshi Yanagita
- Laboratory of Nutrition Biochemistry, Department of Applied Biochemistry and Food Science, Saga University, Saga, Japan
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
- *Correspondence: Yu-Ming Wang
| |
Collapse
|
12
|
Wang Z, Li F, Liu J, Luo Y, Guo H, Yang Q, Xu C, Ma S, Chen H. Intestinal Microbiota - An Unmissable Bridge to Severe Acute Pancreatitis-Associated Acute Lung Injury. Front Immunol 2022; 13:913178. [PMID: 35774796 PMCID: PMC9237221 DOI: 10.3389/fimmu.2022.913178] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022] Open
Abstract
Severe acute pancreatitis (SAP), one of the most serious abdominal emergencies in general surgery, is characterized by acute and rapid onset as well as high mortality, which often leads to multiple organ failure (MOF). Acute lung injury (ALI), the earliest accompanied organ dysfunction, is the most common cause of death in patients following the SAP onset. The exact pathogenesis of ALI during SAP, however, remains unclear. In recent years, advances in the microbiota-gut-lung axis have led to a better understanding of SAP-associated lung injury (PALI). In addition, the bidirectional communications between intestinal microbes and the lung are becoming more apparent. This paper aims to review the mechanisms of an imbalanced intestinal microbiota contributing to the development of PALI, which is mediated by the disruption of physical, chemical, and immune barriers in the intestine, promotes bacterial translocation, and results in the activation of abnormal immune responses in severe pancreatitis. The pathogen-associated molecular patterns (PAMPs) mediated immunol mechanisms in the occurrence of PALI via binding with pattern recognition receptors (PRRs) through the microbiota-gut-lung axis are focused in this study. Moreover, the potential therapeutic strategies for alleviating PALI by regulating the composition or the function of the intestinal microbiota are discussed in this review. The aim of this study is to provide new ideas and therapeutic tools for PALI patients.
Collapse
Affiliation(s)
- Zhengjian Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fan Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jin Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yalan Luo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Haoya Guo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qi Yang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Caiming Xu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Shurong Ma
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- *Correspondence: Shurong Ma, ; Hailong Chen,
| | - Hailong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- *Correspondence: Shurong Ma, ; Hailong Chen,
| |
Collapse
|