1
|
Zhou P, Wu Y, Shen J, Duan T, Che L, Zhang Y, Zhao Y, Yan H. Gestational Inulin Supplementation in Low-/High-Fat Sow Diets: Effects on Growth Performance, Lipid Metabolism, and Meat Quality of Offspring Pigs. Foods 2025; 14:1314. [PMID: 40282717 PMCID: PMC12027208 DOI: 10.3390/foods14081314] [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: 02/22/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
This study investigated whether the supplementation of prebiotic inulin to gestating sows programmatically affects offspring growth performance and meat quality while exploring its epigenetic effects through histone acetylation modulation. After mating, sixty multiparous sows (Landrace × Yorkshire; parity 2-3) were assigned to a 2 × 2 factorial arrangement with inulin (0% vs. 1.5%) and fat (0% or 5%) supplementation until farrowing. Post-weaning, five litters (10 piglets per litter) per treatment were selected and maintained in their original litter for fattening under standardized feeding. The results demonstrated that maternal inulin supplementation during gestation accomplished the following: (1) Increased offspring liver index by 13.4% at weaning and 6.8% at finishing (p < 0.05) while reducing the finishing-phase backfat thickness by 11.6% (p < 0.01), with a significant inulin × fat interaction attenuating fat-induced abdominal lipid accumulation at weaning (p = 0.05). (2) Decreased longissimus dorsi muscle lightness (L*) by 4.5% in finishing pigs (p = 0.02) without altering the other meat quality parameters. (3) Suppressed offspring liver lipid deposition at birth and finishing (p < 0.05), concomitant with upregulated hepatic PGC-1α and CPT1A expression (p < 0.05). (4) Elevated neonatal serum butyrate by 15.6% (p = 0.06) while inhibiting hepatic histone deacetylase (HDAC) activity and enhancing histone H3/H4 acetylation (p < 0.01). These findings suggest that maternal inulin supplementation during gestation mitigates offspring hepatic lipid deposition through butyrate-mediated epigenetic regulation, where microbial-derived butyrate from inulin fermentation inhibits HDAC activity, enhances histone acetylation levels, and upregulates fatty acid β-oxidation gene expression. This study provides novel mechanistic insights into how maternal dietary fiber nutrition programs offspring development through epigenetic reprogramming.
Collapse
Affiliation(s)
- Pan Zhou
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| | - Yachao Wu
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| | - Jianbo Shen
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| | - Tao Duan
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| | - Long Che
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, No. 6 North Longzihu Road, Zhengdong New District, Zhengzhou 450046, China;
| | - Yong Zhang
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| | - Yang Zhao
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Honglin Yan
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| |
Collapse
|
2
|
Wei W, Gao J, Qin F, Zhao X, Jiang X, Che L, Lin Y, Zhuo Y, Feng B, Hua L, Liu G, Sun M, Wu D, Xu S. Effects of unconventional diets and unconventional low-protein diets on reproductive performance, placental nutrient transport, and fecal microorganisms of multiparous sows during gestation. J Anim Sci 2025; 103:skaf095. [PMID: 40208004 PMCID: PMC12080709 DOI: 10.1093/jas/skaf095] [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: 01/02/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
The objective of this experiment was to investigate the effects of unconventional ingredients (wheat, broken rice, rapeseed meal, rice bran meal, and fermented distiller grains) in sow diets on sow reproductive performance, placental nutrient transport function, and fecal microbiota. Thirty multiparous sows with similar parity and backfat thickness were randomly assigned to 3 groups: corn-soybean meal diet (CG), unconventional diet (DY), and unconventional low-protein diet (DYL). The DYL group had 2% lower crude protein than CG and DY. Sows were fed experimental diets during gestation and a corn-soybean meal diet during lactation. Results showed that unconventional ingredients did not negatively affect sow reproductive performance. Compared to the CG treatment, the DY treatment showed a trend of increasing the weaned litter weight (P = 0.061). Compared to the DY treatment, the DYL treatment showed a trend of reducing the number of mummified fetuses (P = 0.066) and increasing the number of weaned piglets (P = 0.096). Additionally, unconventional ingredients enhanced placental nutrient transport gene expression (P < 0.05) and fecal butyric acid content (P < 0.05). Compared to the CG treatment, the DY treatment reduced the apparent digestibility of organic matter, energy, crude protein, and crude fiber but increased the digestibility of crude fat (P < 0.05). In terms of inflammatory factors, the DYL treatment significantly decreased the serum IL-6 content in sows at 90 and 110 d of gestation (P < 0.05). In terms of fecal microbiota, the DY treatment significantly increased the Observed_features and Chao1 indices (P < 0.05), indicating an improvement in fecal microbiota diversity, compared to the CG treatment. At the phylum level, the DYL treatment increased the relative abundance of Proteobacteria. At the genus level, compared to the CG treatment, the DY treatment significantly increased the relative abundance of Anaerovibrio and Ruminococcus, while reducing the relative abundance of Treponema. Additionally, compared to the DY treatment, the DYL treatment significantly increased the relative abundance of Alloprevotella, Prevotella, and Parabacteroides. In summary, replacing corn and soybean meal with unconventional ingredients and reducing protein levels during gestation did not adversely affect sow reproductive performance. During periods of significant price fluctuations in corn and soybean meal, incorporating unconventional ingredients into feed formulations can serve as an alternative solution.
Collapse
Affiliation(s)
- Wenyan Wei
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Junjie Gao
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Feng Qin
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xilun Zhao
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xuemei Jiang
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lianqiang Che
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Lin
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yong Zhuo
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Bin Feng
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lun Hua
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Guangmang Liu
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Mengmeng Sun
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- College of Science, Sichuan Agricultural University, Yucheng, 625014, P.R. China
| | - De Wu
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shengyu Xu
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
3
|
Correnti S, Preianò M, Fregola A, Gamboni F, Stephenson D, Savino R, D'Alessandro A, Terracciano R. Seminal plasma untargeted metabolomic and lipidomic profiling for the identification of a novel panel of biomarkers and therapeutic targets related to male infertility. Front Pharmacol 2023; 14:1275832. [PMID: 37829298 PMCID: PMC10565040 DOI: 10.3389/fphar.2023.1275832] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/15/2023] [Indexed: 10/14/2023] Open
Abstract
Male infertility occurs approximately in about 50% of all infertility cases and represents a serious concern worldwide. Traditional semen analysis alone is insufficient to diagnose male infertility. Over the past two decades, advances in omics technologies have led to the widespread application of metabolomics profiling as a valuable diagnostic tool for various diseases and disorders. Seminal plasma represents a rich and easily accessible source of metabolites surrounding spermatozoa, a milieu that provides several indispensable nutrients to sustain sperm motility and fertilization. Changes of metabolic profiles in seminal plasma reflect male reproductive tract disorders. Here, we performed seminal plasma metabolomics and lipidomics profiling to identify a new pattern of biomarkers of male infertility. Seminal plasma samples from unfertile subjects (n = 31) and fertile controls (n = 19) were analyzed using an untargeted metabolomics/lipidomics integrated approach, based on Ultra-High-Pressure Liquid Chromatography-tandem Mass Spectrometry. Partial Least Squares-Discriminant Analysis showed a distinct separation between healthy fertile men and infertile subjects. Among the 15 selected candidate biomarkers based on Variable Importance in Projection scores, phosphatidylethanolamine (PE) (18:1; 18:1) resulted with the highest score. In total, 40 molecular species showed statistically significant variations between fertile and infertile men. Heat-map and volcano plot analysis indicated that acylcarnitines, phosphatidylserine (PS) (40:2) and lactate were decreased, while PE (18:1; 18:1), Phosphatidic acid (PA) (O-19:2; 18:1), Lysophosphatidylethanolamine (LPE) (O-16:1) and Phosphatidylcholine (PC) (O-16:2; 18:1)-CH3 were increased in the infertile group. The present study is the first one to analyze the metabolomics/lipidomics dysregulation in seminal plasma between fertile and infertile individuals regardless of sub-infertility condition. Association of several metabolites/lipids dysregulation with male infertility reinforced data of previous studies performed with different approaches. In particular, we confirmed significantly decreased levels of PS and carnitines in infertile patients as well as the positive correlation with sperm motility and morphology. If validated on a larger prospective cohort, the metabolite biomarkers of infertility in seminal plasma we identified in the present study might inform novel strategies for diagnosis and interventions to overcome male infertility.
Collapse
Affiliation(s)
- Serena Correnti
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | | | | | - Fabia Gamboni
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rocco Savino
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rosa Terracciano
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| |
Collapse
|
4
|
Ye Q, Li H, Xu B, He Z, Yan X. Butyrate Improves Porcine Endometrial Epithelial Cell Receptivity via Enhancing Acetylation of Histone H3K9. Mol Nutr Food Res 2023; 67:e2200703. [PMID: 37417211 DOI: 10.1002/mnfr.202200703] [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: 10/17/2022] [Revised: 04/26/2023] [Indexed: 07/08/2023]
Abstract
SCOPE Uterine receptivity is a major restriction of embryo implantation and survival, and the endometrial luminal epithelium serves as the transient gateway for uterine receptivity and embryo implantation. Butyrate is reported to promote the success of embryo implantation, but the effects and mechanism of butyrate on uterine receptivity are still unknown. METHODS AND RESULTS Porcine endometrial epithelial cells (PEECs) are used as a model, and the cellular receptivity changes, metabolism, and gene expression profiles influenced by butyrate are analyzed. The study finds that butyrate improves receptive changes in PEECs, including inhibiting proliferation, exhibiting more pinocytosis on the cell surface, and increasing adhesiveness to porcine trophoblast cells. In addition, butyrate increases prostaglandin synthesis and markedly impacts purine metabolism, pyrimidine metabolism, and the FoxO signaling pathway. siRNA to inhibit the expression of FoxO1 and chromatin immunoprecipitation-sequencing (ChIP-seq) of H3K9ac are used to demonstrate that the H3K9ac/FoxO1/PCNA pathway can contribute to the effects of cell proliferation inhibition and uterine receptivity improvement induced by butyrate. CONCLUSION The findings reveal that butyrate improves endometrial epithelial cell receptivity by enhancing the acetylation of histone H3K9, which shows nutritional regulation and therapeutic potential for poor uterine receptivity and difficulty in embryo implantation.
Collapse
Affiliation(s)
- Qianhong Ye
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal, Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, Hubei, 430070, China
| | - Haoyu Li
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal, Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, Hubei, 430070, China
| | - Baoyang Xu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal, Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, Hubei, 430070, China
| | - Ziyi He
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal, Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, Hubei, 430070, China
| | - Xianghua Yan
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal, Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, Hubei, 430070, China
| |
Collapse
|
5
|
Zeng T, Sun H, Huang M, Guo R, Gu T, Cao Y, Li C, Tian Y, Chen L, Li G, Lu L. Dietary supplementation of coated sodium butyrate improves growth performance of laying ducks by regulating intestinal health and immunological performance. Front Immunol 2023; 14:1142915. [PMID: 36969242 PMCID: PMC10034168 DOI: 10.3389/fimmu.2023.1142915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
IntroductionThis study was conducted to assess the effects of dietary supplementation of coated sodium butyrate (CSB) on the growth performance, serum antioxidant, immune performance, and intestinal microbiota of laying ducks.MethodsA total of 120 48-week-old laying ducks were randomly divided into 2 treatment groups: the control group (group C fed a basal diet) and the CSB-treated group (group CSB fed the basal diet + 250 g/t of CSB). Each treatment consisted of 6 replicates, with 10 ducks per replicate, and the trial was conducted for 60 days.ResultsCompared with the group C, the group CSB showed a significant increase in the laying rate (p<0.05) of the 53-56 week-old ducks. Additionally, the serum total antioxidant capacity, superoxide dismutase activity and immunoglobulin G level were significantly higher (p<0.05), while the serum malondialdehyde content and tumor necrosis factor (TNF)-a level were significantly lower (p<0.05) in the serum of the group CSB compared to the group C. Moreover, the expression of IL-1b and TNF-a in the spleen of the group CSB was significantly lower (p<0.05) compared to that of the group C. In addition, compared with the group C, the expression of Occludin in the ileum and the villus height in the jejunum were significantly higher in the group CSB (p<0.05). Furthermore, Chao1, Shannon, and Pielou-e indices were higher in the group CSB compared to the group C (p<0.05). The abundance of Bacteroidetes in the group CSB was lower than that in the group C (p<0.05), while the abundances of Firmicutes and Actinobacteria were higher in the group CSB compared to the group C (p<0.05).ConclusionsOur results suggest that the dietary supplementation of CSB can alleviate egg-laying stress in laying ducks by enhancing immunity and maintaining the intestinal health of the ducks.
Collapse
Affiliation(s)
- Tao Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Hanxue Sun
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Manman Huang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Rongbing Guo
- College of Animal Science, Zhejiang A&F University, Hangzhou, China
| | - Tiantian Gu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yongqing Cao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Chengfeng Li
- Hubei Shendan Health Food Co., Ltd., Xiaogan, China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Li Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Guoqin Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Lizhi Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- *Correspondence: Lizhi Lu,
| |
Collapse
|
6
|
Zeng X, Li S, Ye Q, Cai S, Quan S, Liu L, Zhang S, Chen F, Cai C, Wang F, Qiao S, Zeng X. The Combined Use of Medium- and Short-Chain Fatty Acids Improves the Pregnancy Outcomes of Sows by Enhancing Ovarian Steroidogenesis and Endometrial Receptivity. Nutrients 2022; 14:nu14204405. [PMID: 36297089 PMCID: PMC9607977 DOI: 10.3390/nu14204405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Fatty acids play important roles in maintaining ovarian steroidogenesis and endometrial receptivity. Porcine primary ovarian granulosa cells (PGCs) and endometrial epithelial cells (PEECs) were treated with or without medium- and short-chain fatty acids (MSFAs) for 24 h. The mRNA abundance of genes was detected by fluorescence quantitative PCR. The hormone levels in the PGCs supernatant and the rate of adhesion of porcine trophoblast cells (pTrs) to PEECs were measured. Sows were fed diets with or without MSFAs supplementation during early gestation. The fecal and vaginal microbiomes were identified using 16S sequencing. Reproductive performance was recorded at parturition. MSFAs increased the mRNA abundance of genes involved in steroidogenesis, luteinization in PGCs and endometrial receptivity in PEECs (p < 0.05). The estrogen level in the PGC supernatant and the rate of adhesion increased (p < 0.05). Dietary supplementation with MSFAs increased serum estrogen levels and the total number of live piglets per litter (p < 0.01). Moreover, MSFAs reduced the fecal Trueperella abundance and vaginal Escherichia-Shigella and Clostridium_sensu_stricto_1 abundance. These data revealed that MSFAs improved pregnancy outcomes in sows by enhancing ovarian steroidogenesis and endometrial receptivity while limiting the abundance of several intestinal and vaginal pathogens at early stages of pregnancy.
Collapse
Affiliation(s)
- Xiangzhou Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, China
- Beijing Key Laboratory of Biofeed Additives, Beijing 100193, China
| | - Siyu Li
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, China
- Beijing Key Laboratory of Biofeed Additives, Beijing 100193, China
| | - Qianhong Ye
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Shuang Cai
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, China
- Beijing Key Laboratory of Biofeed Additives, Beijing 100193, China
| | - Shuang Quan
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, China
- Beijing Key Laboratory of Biofeed Additives, Beijing 100193, China
| | - Lu Liu
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, China
- Beijing Key Laboratory of Biofeed Additives, Beijing 100193, China
| | - Shihai Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Fang Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Chuanjiang Cai
- College of Animal Science and Technology, Northwest A&F University, Xi’an 712100, China
| | - Fenglai Wang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, China
- Beijing Key Laboratory of Biofeed Additives, Beijing 100193, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, China
- Beijing Key Laboratory of Biofeed Additives, Beijing 100193, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, China
- Beijing Key Laboratory of Biofeed Additives, Beijing 100193, China
- Correspondence:
| |
Collapse
|
7
|
Lin Y, Wang K, Che L, Fang Z, Xu S, Feng B, Zhuo Y, Li J, Wu C, Zhang J, Xiong H, Yu C, Wu D. The Improvement of Semen Quality by Dietary Fiber Intake Is Positively Related With Gut Microbiota and SCFA in a Boar Model. Front Microbiol 2022; 13:863315. [PMID: 35633720 PMCID: PMC9130837 DOI: 10.3389/fmicb.2022.863315] [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: 01/27/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Although fiber-rich diets have been positively associated with sperm quality, there have not been any studies that have examined the effects of dietary fiber and its metabolites on sperm quality in young or pre-pubescent animals. In this study, we aimed to explore the effect of dietary fiber supplementation on semen quality and the underlying mechanisms in a boar model. Sixty purebred Yorkshire weaning boars were randomly divided into the four groups (T1–T4). Groups T1, T2, and T3 boars were fed diets with different levels of fiber until reaching 160 days of age and were then fed the same diet, while group T4 boars were fed a basal diet supplemented with butyrate and probiotics. Compared with T1 boars, sperm motility and effective sperm number were significantly higher among T3 boars. Meanwhile, at 240 days of age, the acetic acid and total short-chain fatty acid (SCFA) contents in the sera of T3 and T4 boars were significantly higher than those in T1 boars. The abundance of microbiota in T2 and T3 boars was significantly higher than that in T1 boars (P < 0.01). Moreover, dietary fiber supplementation increased “beneficial gut microbes” such as UCG-005, Rumenococcus, Rikenellaceae_RC9_gut_group and Lactobacillus and decreased the relative abundance of “harmful microbes” such as Clostridium_sensu_stricto_1, Romboutsia and Turicibacter. Collectively, the findings of this study indicate that dietary fiber supplementation improves gut microbiota and promotes SCFA production, thereby enhancing spermatogenesis and semen quality. Moreover, the effects of dietary fiber are superior to those of derived metabolites.
Collapse
Affiliation(s)
- Yan Lin
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Ke Wang
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Lianqiang Che
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Zhengfeng Fang
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Shengyu Xu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Bin Feng
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Yong Zhuo
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Jian Li
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Caimei Wu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Junjie Zhang
- College of Life Science, Sichuan Agricultural University, Ya'an, China
| | - Haoyu Xiong
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Chenglong Yu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - De Wu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| |
Collapse
|
8
|
Mandala A, Dobrinskikh E, Janssen RC, Fiehn O, D’Alessandro A, Friedman JE, Jonscher KR. Maternal Pyrroloquinoline Quinone Supplementation Improves Offspring Liver Bioactive Lipid Profiles throughout the Lifespan and Protects against the Development of Adult NAFLD. Int J Mol Sci 2022; 23:6043. [PMID: 35682720 PMCID: PMC9181499 DOI: 10.3390/ijms23116043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 02/06/2023] Open
Abstract
Maternal obesity and consumption of a high-fat diet significantly elevate risk for pediatric nonalcoholic fatty liver disease (NAFLD), affecting 10% of children in the US. Almost half of these children are diagnosed with nonalcoholic steatohepatitis (NASH), a leading etiology for liver transplant. Animal models show that signs of liver injury and perturbed lipid metabolism associated with NAFLD begin in utero; however, safe dietary therapeutics to blunt developmental programming of NAFLD are unavailable. Using a mouse model of maternal Western-style diet (WD), we previously showed that pyrroloquinoline quinone (PQQ), a potent dietary antioxidant, protected offspring of WD-fed dams from development of NAFLD and NASH. Here, we used untargeted mass spectrometry-based lipidomics to delineate lipotoxic effects of WD on offspring liver and identify lipid targets of PQQ. PQQ exposure during pregnancy altered hepatic lipid profiles of WD-exposed offspring, upregulating peroxisome proliferator-activated receptor (PPAR) α signaling and mitochondrial fatty acid oxidation to markedly attenuate triglyceride accumulation beginning in utero. Surprisingly, the abundance of very long-chain ceramides, important in promoting gut barrier and hepatic function, was significantly elevated in PQQ-treated offspring. PQQ exposure reduced the hepatic phosphatidylcholine/phosphatidylethanolamine (PC/PE) ratio in WD-fed offspring and improved glucose tolerance. Notably, levels of protective n - 3 polyunsaturated fatty acids (PUFAs) were elevated in offspring exposed to PQQ, beginning in utero, and the increase in n - 3 PUFAs persisted into adulthood. Our findings suggest that PQQ supplementation during gestation and lactation augments pathways involved in the biosynthesis of long-chain fatty acids and plays a unique role in modifying specific bioactive lipid species critical for protection against NAFLD risk in later life.
Collapse
Affiliation(s)
- Ashok Mandala
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (J.E.F.)
| | - Evgenia Dobrinskikh
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (J.E.F.)
| | - Oliver Fiehn
- Genome Center-Metabolomics, University of California Davis, Davis, CA 95616, USA;
| | - Angelo D’Alessandro
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (J.E.F.)
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Karen R. Jonscher
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (J.E.F.)
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
9
|
Lin Y, Wu D, Che L, Fang Z, Xu S, Feng B, Zhuo Y, Li J, Wu C, Zhang J, Li L. Dietary Fibre Supplementation Improves Semen Production by Increasing Leydig Cells and Testosterone Synthesis in a Growing Boar Model. Front Vet Sci 2022; 9:850685. [PMID: 35359689 PMCID: PMC8963373 DOI: 10.3389/fvets.2022.850685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/14/2022] [Indexed: 02/01/2023] Open
Abstract
Testicular development is imperative to spermatogenesis, and pre-puberty is the key period for testis development. This study, therefore, investigated the effects of fibre supplementation on testis development and its possible mechanism in a growing boar model. Thirty Yorkshire boars were randomly divided into a control group (Control) and a fibre group (Fibre) from day 0 to 90 after weaning, with three pigs per pen and five pens per treatment. Blood and testes were collected for analysis. Dietary fibre supplementation had no significant effect on growth performance, testicular volume, or libido but increased the semen production of boars. Boars fed with fibre had lower serum cholesterol (CHO) and low-density lipoprotein (LDL) levels compared to those on the Control diet; however, testicular CHO, triglyceride (TG), and LDL concentration in the Fibre group were significantly higher than the Control group (P < 0.01). Testicular histological analysis showed that seminiferous tubules and testicular germ cells of 120-day-old boars were densely arranged in the Fibre group, and the number of Leydig cells was significantly higher than that of the Control group (P < 0.001). Furthermore, the diet supplemented with fibre significantly decreased leptin, leptin receptor (Leptor), and luteinising hormone (LH) concentrations in boar serum (P < 0.05), whereas follicle-stimulating hormone (FSH) and testosterone concentrations were significantly increased (P < 0.05). Meanwhile, the expression of AMH, AMHR2, and SYCP3 genes related to proliferation and differentiation, and hormone-related genes STAR and SOCS3, were significantly up-regulated (P < 0.05). OCCLUDIN expression was up-regulated, whereas CDH2 expression was down-regulated. In conclusion, increased fibre intake during the pre-puberty period in growing boar is crucial for Leydig cell proliferation, up-regulating the expression of genes related to hormone synthesis and thereby promoting the secretion of testosterone and semen production.
Collapse
Affiliation(s)
- Yan Lin
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Ministry of Agriculture, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- *Correspondence: Yan Lin
| | - De Wu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Ministry of Agriculture, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Lianqiang Che
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Ministry of Agriculture, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Zhengfeng Fang
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Ministry of Agriculture, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Shengyu Xu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Ministry of Agriculture, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Bin Feng
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Ministry of Agriculture, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Yong Zhuo
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Ministry of Agriculture, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Jian Li
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Ministry of Agriculture, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Caimei Wu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Ministry of Agriculture, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| | - Junjie Zhang
- College of Life Science, Sichuan Agricultural University, Ya'an, China
| | - Lujie Li
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Ministry of Agriculture, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
| |
Collapse
|
10
|
Alterations in Intestinal Antioxidant and Immune Function and Cecal Microbiota of Laying Hens Fed on Coated Sodium Butyrate Supplemented Diets. Animals (Basel) 2022; 12:ani12050545. [PMID: 35268114 PMCID: PMC8908843 DOI: 10.3390/ani12050545] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/15/2022] [Accepted: 02/19/2022] [Indexed: 02/04/2023] Open
Abstract
This study was designed to evaluate the effects of dietary coated sodium butyrate (CSB) on the intestinal antioxidant, immune function, and cecal microbiota of laying hens. A total of 720 52-week-old Huafeng laying hens were randomly allocated into five groups and fed a basal diet supplemented with CSB at levels of 0 (control), 250 (S250), 500 (S500), 750 (S750), and 1000 (S1000) mg/kg for eight weeks. The results revealed that CSB supplementation quadratically decreased the malondialdehyde content and increased the superoxide dismutase activity of the jejunum as well as the total antioxidative capacity activity of the ileum (p < 0.05). Dietary CSB supplementation linearly decreased the diamine oxidase and D-lactic acid content of the serum (p < 0.05). Compared with the control group, the addition of CSB resulted in linear and/or quadratic effects on the mRNA expression of inflammatory cytokines TNF-α, IL-6, and IL-10 in the jejunum and ileum (p < 0.05). The short-chain fatty acid concentrations increased quadratically as supplemental CSB improved (p < 0.05). Additionally, dietary CSB levels had no effect on microbial richness estimators, but ameliorated cecal microbiota by raising the abundance of probiotics and lowering pathogenic bacteria enrichment. In conclusion, our results suggest that dietary supplementation with CSB could improve the intestinal health of laying hens via positively influencing the antioxidant capacity, inflammatory cytokines, short-chain fatty acids, and gut microbiota. In this study, 500 mg/kg CSB is the optimal supplement concentration in the hens’ diet.
Collapse
|
11
|
Wang J, Zhang H, Bai S, Zeng Q, Su Z, Zhuo Y, Mao X, Yin H, Feng B, Liu J, Zhang K, Ding X. Dietary tributyrin improves reproductive performance, antioxidant capacity, and ovary function of broiler breeders. Poult Sci 2021; 100:101429. [PMID: 34555757 PMCID: PMC8458981 DOI: 10.1016/j.psj.2021.101429] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/02/2021] [Accepted: 07/20/2021] [Indexed: 11/25/2022] Open
Abstract
The objective of this experiment was to investigate the influence of dietary tributyrin on reproduction performance and ovary function of broiler breeders with different egg laying rate. Two hundred fifty-six AA broiler breeders (48-wk-old) were allocated to 4 treatment in a 2 × 2 factorial arrangement with the main effects of tributyrin supplementation (0 and 1,000 mg/kg tributyrin [TRI]) and 2 egg laying rate levels (average [AR, 81.01 ± 0.79%] and low [LR, 70.98 ± 0.95%]). The results shown that the LR breeders presented higher egg weight, but lower egg laying rate, qualified egg rate and feed efficiency than the AR breeders (P(laying) < 0.05). Also, the superoxidase dismutase (SOD) activity in magnum was lower while malondialdehyde (MDA) was higher in ovary and magnum of LR breeders than that in the AR breeders (P(laying) < 0.05). Dietary supplementation with tributyrin significantly enhanced egg weight (P(TRI) < 0.05), increased albumen height as well as Haugh unit (HU) in AR breeders (P(interaction) < 0.05), and also had higher total antioxidant capacity (T-AOC) and lower MDA in ovary (P(TRI) < 0.05). The cell apoptosis rate and proapoptosis related gene expression (caspase 8, 9 and Bax) in the ovary of LR breeders was higher, while anti-apoptosis related gene (Bcl-2) expression were lower in LR breeders when compared with the AR breeders (P(laying) < 0.05). Dietary supplementation with tributyrin decreased the cell apoptosis rate and downregulated caspase 9 expression in LR breeders (P(Interaction) < 0.05), up-regulated the Bcl-2 expression in both 2 breeders (P(TRI) < 0.05). These findings suggest that the breeders with lower egg laying rate also characterized by deteriorate ovary function indicated by lower antioxidant capacity and higher cell apoptosis rate. Dietary supplementation with tributyrin increased egg albumen quality, decreased ovarian proapoptosis related gene expression to improve reproductive tract function; and the positive effect on egg albumen quality is more pronounced in average reproductive breeders.
Collapse
Affiliation(s)
- Jianping Wang
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, 611130, China
| | - Hongye Zhang
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shiping Bai
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qiufeng Zeng
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhuowei Su
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yong Zhuo
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiangbing Mao
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, 611130, China
| | - Huadong Yin
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, 611130, China
| | - Bin Feng
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, China
| | - Keying Zhang
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xuemei Ding
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
12
|
Ye Q, Zeng X, Wang S, Zeng X, Yang G, Ye C, Cai S, Chen M, Li S, Qiao S. Butyrate drives the acetylation of histone H3K9 to activate steroidogenesis through PPARγ and PGC1α pathways in ovarian granulosa cells. FASEB J 2021; 35:e21316. [PMID: 33433947 DOI: 10.1096/fj.202000444r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 11/26/2020] [Accepted: 12/14/2020] [Indexed: 12/21/2022]
Abstract
Maintaining ovarian steroidogenesis is of critical importance, considering that steroid hormones are required for successful establishment and maintenance of pregnancy and proper development of embryos and fetuses. Investigating the mechanism that butyrate modulates the ovarian steroidogenesis is beneficial for understanding the impact of lipid nutrition on steroidogenesis. Herein, we identified that butyrate improved estradiol and progesterone synthesis in rat primary ovarian granulosa cells and human granulosa KGN cells and discovered the related mechanism. Our data indicated that butyrate was sensed by GPR41 and GPR43 in ovarian granulosa cells. Butyrate primarily upregulated the acetylation of histone H3K9 (H3K9ac). Chromatin immune-precipitation and sequencing (ChIP-seq) data of H3K9ac revealed the influenced pathways involving in the mitochondrial function (including cellular metabolism and steroidogenesis) and cellular antioxidant capacity. Additionally, increasing H3K9ac by butyrate further stimulated the PPARγ/CD36/StAR pathways to increase ovarian steroidogenesis and activated PGC1α to enhance mitochondrial dynamics and alleviate oxidative damage. The improvement in antioxidant capacity and mitochondrial dynamics by butyrate enhanced ovarian steroidogenesis. Collectively, butyrate triggers histone H3K9ac to activate steroidogenesis through PPARγ and PGC1α pathways in ovarian granulosa cells.
Collapse
Affiliation(s)
- Qianhong Ye
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, P.R. China.,Beijing Key Laboratory of Biofeed Additives, Beijing, P.R. China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, P.R. China.,Beijing Key Laboratory of Biofeed Additives, Beijing, P.R. China
| | - Shuai Wang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, P.R. China.,Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Xiangzhou Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, P.R. China.,Beijing Key Laboratory of Biofeed Additives, Beijing, P.R. China
| | - Guangxin Yang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, P.R. China.,Beijing Key Laboratory of Biofeed Additives, Beijing, P.R. China
| | - Changchuan Ye
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, P.R. China.,Beijing Key Laboratory of Biofeed Additives, Beijing, P.R. China
| | - Shuang Cai
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, P.R. China.,Beijing Key Laboratory of Biofeed Additives, Beijing, P.R. China
| | - Meixia Chen
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, P.R. China.,Beijing Key Laboratory of Biofeed Additives, Beijing, P.R. China
| | - Siyu Li
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, P.R. China.,Beijing Key Laboratory of Biofeed Additives, Beijing, P.R. China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, P.R. China.,Beijing Key Laboratory of Biofeed Additives, Beijing, P.R. China
| |
Collapse
|
13
|
Li X, Su C, Jiang Z, Yang Y, Zhang Y, Yang M, Zhang X, Du Y, Zhang J, Wang L, Jiang J, Hong B. Berberine attenuates choline-induced atherosclerosis by inhibiting trimethylamine and trimethylamine-N-oxide production via manipulating the gut microbiome. NPJ Biofilms Microbiomes 2021; 7:36. [PMID: 33863898 PMCID: PMC8052457 DOI: 10.1038/s41522-021-00205-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/16/2021] [Indexed: 12/11/2022] Open
Abstract
Trimethylamine-N-oxide (TMAO), a derivative from the gut microbiota metabolite trimethylamine (TMA), has been identified to be an independent risk factor for promoting atherosclerosis. Evidences suggest that berberine (BBR) could be used to treat obesity, diabetes and atherosclerosis, however, its mechanism is not clear mainly because of its poor oral bioavailability. Here, we show that BBR attenuated TMA/TMAO production in the C57BL/6J and ApoE KO mice fed with choline-supplemented chow diet, and mitigated atherosclerotic lesion areas in ApoE KO mice. Inhibition of TMA/TMAO production by BBR-modulated gut microbiota was proved by a single-dose administration of d9-choline in vivo. Metagenomic analysis of cecal contents demonstrated that BBR altered gut microbiota composition, microbiome functionality, and cutC/cntA gene abundance. Furthermore, BBR was shown to inhibit choline-to-TMA conversion in TMA-producing bacteria in vitro and in gut microbial consortium from fecal samples of choline-fed mice and human volunteers, and the result was confirmed by transplantation of TMA-producing bacteria in mice. These results offer new insights into the mechanisms responsible for the anti-atherosclerosis effects of BBR, which inhibits commensal microbial TMA production via gut microbiota remodeling.
Collapse
Affiliation(s)
- Xingxing Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chunyan Su
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhibo Jiang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuxin Yang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yue Zhang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mengxia Yang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiumin Zhang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yu Du
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jin Zhang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Li Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Jiandong Jiang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Bin Hong
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
14
|
Zhou Y, Ji X, Chen J, Fu Y, Huang J, Guo R, Zhou J, Cen J, Zhang Q, Chu A, Huang Y, Xu C, Wang F. Short-chain fatty acid butyrate: A novel shield against chronic gastric ulcer. Exp Ther Med 2021; 21:329. [PMID: 33732302 PMCID: PMC7903393 DOI: 10.3892/etm.2021.9760] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 12/14/2020] [Indexed: 01/16/2023] Open
Abstract
Butyrate is one of the most abundant short-chain fatty acids produced by intestinal bacteria. In the present study, the action of butyrate on chronic gastric mucosa lesions was investigated, as well as its underlying mechanism in mice. Male mice from the Institute of Cancer Research were randomly divided into three groups: Sham, model and butyrate groups. Butyrate was administered intragastrically for 7 days to butyrate group mice following the establishment of a gastric ulcer model. Hematoxylin and eosin staining, immunohistochemical analysis, enzyme-linked immunosorbent assay and quantitative polymerase chain reaction were used to determine the therapeutic effects and molecular mechanism of butyrate treatment. The findings demonstrated that butyrate induced a marked shift in superoxide dismutase and catalase activities, along with a decrease in malondialdehyde levels, thereby attenuating oxidative stress. Furthermore, butyrate decreased the levels of pro-inflammatory cytokines interleukin-1β, tumour necrosis factor-α and leukotriene B4, which helped combat inflammatory responses. Moreover, butyrate treatment exerted remarkable positive influences that mediate an increase in 6-keto-PGF-1α (a degradation product of prostacyclin), trefoil factor 2, MUC5AC and fibroblast growth factor-7 levels to promote gastric mucosal repair. The expression of specific receptor GPR109A for butyrate was upregulated, with no significant difference noted in the expression of GPR43 or GPR41. Overall, the present findings revealed that butyrate exerted therapeutic effects by upregulating mucosal repair factors and stimulating protective responses against oxidation and inflammation. GPR109A may be the key receptor for butyrate therapy.
Collapse
Affiliation(s)
- Yan Zhou
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiawei Ji
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jiajing Chen
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yaoyang Fu
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Juewei Huang
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Rui Guo
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jinhui Zhou
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jianke Cen
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Qihao Zhang
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Anne Chu
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yingpeng Huang
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Changlong Xu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Fangyan Wang
- Department of Pathophysiology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
15
|
Heinecke F, Mazzucco MB, Fornes D, Roberti S, Jawerbaum A, White V. The offspring from rats fed a fatty diet display impairments in the activation of liver peroxisome proliferator activated receptor alpha and features of fatty liver disease. Mol Cell Endocrinol 2020; 511:110818. [PMID: 32298755 DOI: 10.1016/j.mce.2020.110818] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023]
Abstract
Maternal obesity programs liver derangements similar to those of NAFLD. Our main goal was to evaluate whether these liver anomalies were related to aberrant PPARα function. Obesity was induced in female Albino-Wistar rats by a fatty diet (FD rats). Several parameters related to NAFLD were evaluated in both plasma and livers from fetuses of 21 days of gestation and 140-day-old offspring. FD fetuses and offspring developed increased levels of AST and ALT, signs of inflammation and oxidative and nitrative stress-related damage. FD offspring showed dysregulation of Plin2, CD36, Cyp4A, Aco, Cpt-1, Hadha and Acaa2 mRNA levels, genes involved in lipid metabolism and no catabolic effect of the PPARα agonist clofibrate. These results suggest that the FD offspring is prone to develop fatty liver, a susceptibility that can be linked to PPARα dysfunction, and that this could in turn be related to the liver impairments programmed by maternal obesity.
Collapse
Affiliation(s)
- Florencia Heinecke
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina
| | - María Belén Mazzucco
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina
| | - Daiana Fornes
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina
| | - Sabrina Roberti
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina
| | - Alicia Jawerbaum
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina
| | - Verónica White
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina.
| |
Collapse
|
16
|
Yang LL, Millischer V, Rodin S, MacFabe DF, Villaescusa JC, Lavebratt C. Enteric short-chain fatty acids promote proliferation of human neural progenitor cells. J Neurochem 2019; 154:635-646. [PMID: 31784978 DOI: 10.1111/jnc.14928] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/02/2019] [Accepted: 11/21/2019] [Indexed: 12/22/2022]
Abstract
Short-chain fatty acids (SCFAs) are a group of fatty acids predominantly produced during the fermentation of dietary fibers by the gut anaerobic microbiota. SCFAs affect many host processes in health and disease. SCFAs play an important role in the 'gut-brain axis', regulating central nervous system processes, for example, cell-cell interaction, neurotransmitter synthesis and release, microglia activation, mitochondrial function, and gene expression. SCFAs also promote the growth of neurospheres from human neural stem cells and the differentiation of embryonic stem cells into neural cells. It is plausible that maternally derived SCFAs may pass the placenta and expose the fetus at key developmental periods. However, it is unclear how SCFA exposure at physiological levels influence the early-stage neural cells. In this study, we explored the effect of SCFAs on the growth rate of human neural progenitor cells (hNPCs), generated from human embryonic stem cell line (HS980), with IncuCyte live-cell analysis system and immunofluorescence. We found that physiologically relevant levels (µM) of SCFAs (acetate, propionate, butyrate) increased the growth rate of hNPCs significantly and induced more cells to undergo mitosis, while high levels (mM) of SCFAs had toxic effects on hNPCs. Moreover, no effect on apoptosis was observed in physiological-dose SCFA treatments. In support, data from q-RT PCR showed that SCFA treatments influenced the expression of the neurogenesis, proliferation, and apoptosis-related genes ATR, BCL2, BID, CASP8, CDK2, E2F1, FAS, NDN, and VEGFA. To conclude, our results propose that SCFAs regulates early neural system development. This might be relevant for a putative 'maternal gut-fetal brain-axis'. Cover Image for this issue: doi: 10.1111/jnc.14761.
Collapse
Affiliation(s)
- Liu L Yang
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Vincent Millischer
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Sergey Rodin
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Derrick F MacFabe
- The Kilee Patchell-Evans Autism Research Group, London, Canada.,Center for Healthy Eating and Food Innovation, Faculty of Medicine, Maastricht University, Maastricht, Netherlands
| | - Juan C Villaescusa
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
17
|
Ye Q, Cai S, Wang S, Zeng X, Ye C, Chen M, Zeng X, Qiao S. Maternal short and medium chain fatty acids supply during early pregnancy improves embryo survival through enhancing progesterone synthesis in rats. J Nutr Biochem 2019; 69:98-107. [PMID: 31063920 DOI: 10.1016/j.jnutbio.2019.03.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/23/2019] [Accepted: 03/14/2019] [Indexed: 12/22/2022]
Abstract
Exploring strategies to prevent miscarriage in women or early pregnancy loss in mammals is of great importance. Manipulating maternal lipid metabolism to maintain sufficient progesterone level is an effective way. To investigated the embryo loss and progesterone synthesis impacts of short and medium chain fatty acids on the lipid metabolism, pregnancy outcome and embryo implantation were investigated in rats fed the pregnancy diets supplemented without or with 0.1% sodium butyrate (SB), 0.1% sodium hexanoate (SH), or 0.1% sodium caprylate (SC) during the entire pregnancy and early pregnancy, respectively, followed with evaluation of potential mechanisms. Maternal SB, SH, or SC supply significantly improved live litter size and embryo implantation in rats. Serum progesterone, arachidonic acid, and phospholipid metabolites levels were significantly increased in response to maternal SB, SH, and SC supply. The expression of key genes involved in ovarian steroidogenesis and granulosa cell luteinization were elevated in ovaries and primary cultured granulosa cells, including cluster of differentiation 36 (CD36), steroidogenic acute regulatory protein (StAR), and cholesterol side-chain cleavage enzyme (CYP11A1). Additionally, the expression of lysophosphatidic acid receptor 3 (LPA3) and cyclooxygenase-2 (COX2) related with phospholipid metabolism were enhanced in uterus in vivo and in in vitro cultured uterine tissue. In conclusion, maternal SB, SH and SC supply reduced early pregnancy loss through modulating maternal phospholipid metabolism and ovarian progesterone synthesis in rats. Our results have important implications that short or medium chain fatty acids have the potential to prevent miscarriage in women or early pregnancy loss in mammals.
Collapse
Affiliation(s)
- Qianhong Ye
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, PR, China; Beijing Key Laboratory of bio-feed additives, Beijing 100193, PR, China.
| | - Shuang Cai
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, PR, China; Beijing Key Laboratory of bio-feed additives, Beijing 100193, PR, China.
| | - Shuai Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, PR. China.
| | - Xiangzhou Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, PR, China; Beijing Key Laboratory of bio-feed additives, Beijing 100193, PR, China.
| | - Changchuan Ye
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, PR, China; Beijing Key Laboratory of bio-feed additives, Beijing 100193, PR, China.
| | - Meixia Chen
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, PR, China; Beijing Key Laboratory of bio-feed additives, Beijing 100193, PR, China.
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, PR, China; Beijing Key Laboratory of bio-feed additives, Beijing 100193, PR, China.
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing 100193, PR, China; Beijing Key Laboratory of bio-feed additives, Beijing 100193, PR, China.
| |
Collapse
|
18
|
Wang C, Cao S, Zhang Q, Shen Z, Feng J, Hong Q, Lu J, Xie F, Peng Y, Hu C. Dietary Tributyrin Attenuates Intestinal Inflammation, Enhances Mitochondrial Function, and Induces Mitophagy in Piglets Challenged with Diquat. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:1409-1417. [PMID: 30599507 DOI: 10.1021/acs.jafc.8b06208] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The study evaluated the effects of butyric acid, in the form of tributyrin on the oxidative stress, inflammation, and mitochondrial function in diquat-challenged pigs. Twenty-four weaned pigs were allocated to four treatments in a 2 × 2 factorial arrangement with the main effects of tributyrin supplementation and diquat challenge. The results showed that supplemental tributyrin increased ( P < 0.05) average daily gain and average daily feed intake of diquat-challenged pigs. Tributyrin elevated ( P < 0.05) the activities of total antioxidant capacity and superoxide dismutase, reduced ( P < 0.05) malondialdehyde content, and increased ( P < 0.05) mRNA levels of copper and zinc superoxide dismutase and manganese-containing superoxide dismutase of diquat-challenged pigs. Tributyrin relieved ( P < 0.05) intestinal inflammation reflected by decreased mRNA abundances of tumor necrosis factor-α, interferon-γ, and interleukin-6 in the intestine. Tributyrin reduced ( P < 0.05) serum diamine oxidase activity and d-lactate content, increased ( P < 0.05) transepithelial electrical resistance, decreased paracellular flux of dextran (4 kDa), and prevented the diquat-induced decrease ( P < 0.05) in the expressions of claudin-1, occludin, and zonula occludens-1. Tributyrin alleviated ( P < 0.05) diquat-induced mitochondrial dysfunction shown by lowered reactive oxygen species, increased mitochondrial membrane potential, and increased adenosine triphosphate content. Furthermore, tributyrin increased ( P < 0.05) expressions of mitophagy proteins (PTEN-induced putative kinase 1 and Parkin), and ratio of light chain 3-II to light chain 3-I in intestine. Collectively, tributyrin attenuated oxidative stress and intestinal inflammation, improved mitochondrial function, and induced mitophagy in diquat-challenged pigs.
Collapse
Affiliation(s)
- Chunchun Wang
- Animal Science College , Zhejiang University , Hangzhou 310058 , P.R. China
| | - Shuting Cao
- Animal Science College , Zhejiang University , Hangzhou 310058 , P.R. China
| | - Qianhui Zhang
- Animal Science College , Zhejiang University , Hangzhou 310058 , P.R. China
| | - Zhuojun Shen
- Animal Science College , Zhejiang University , Hangzhou 310058 , P.R. China
| | - Jie Feng
- Animal Science College , Zhejiang University , Hangzhou 310058 , P.R. China
| | - Qihua Hong
- Animal Science College , Zhejiang University , Hangzhou 310058 , P.R. China
| | - Jianjun Lu
- Animal Science College , Zhejiang University , Hangzhou 310058 , P.R. China
| | - Fei Xie
- Shanghai Menon Animal Nutrition Technology Co. Ltd. , Shanghai 201807 , P.R. China
| | - Yan Peng
- Shanghai Menon Animal Nutrition Technology Co. Ltd. , Shanghai 201807 , P.R. China
| | - Caihong Hu
- Animal Science College , Zhejiang University , Hangzhou 310058 , P.R. China
| |
Collapse
|
19
|
Maes M, Anderson G, Betancort Medina SR, Seo M, Ojala JO. Integrating Autism Spectrum Disorder Pathophysiology: Mitochondria, Vitamin A, CD38, Oxytocin, Serotonin and Melatonergic Alterations in the Placenta and Gut. Curr Pharm Des 2019; 25:4405-4420. [PMID: 31682209 DOI: 10.2174/1381612825666191102165459] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND A diverse array of data has been associated with autism spectrum disorder (ASD), reflecting the complexity of its pathophysiology as well as its heterogeneity. Two important hubs have emerged, the placenta/prenatal period and the postnatal gut, with alterations in mitochondria functioning crucial in both. METHODS Factors acting to regulate mitochondria functioning in ASD across development are reviewed in this article. RESULTS Decreased vitamin A, and its retinoic acid metabolites, lead to a decrease in CD38 and associated changes that underpin a wide array of data on the biological underpinnings of ASD, including decreased oxytocin, with relevance both prenatally and in the gut. Decreased sirtuins, poly-ADP ribose polymerase-driven decreases in nicotinamide adenine dinucleotide (NAD+), hyperserotonemia, decreased monoamine oxidase, alterations in 14-3-3 proteins, microRNA alterations, dysregulated aryl hydrocarbon receptor activity, suboptimal mitochondria functioning, and decreases in the melatonergic pathways are intimately linked to this. Many of the above processes may be modulating, or mediated by, alterations in mitochondria functioning. Other bodies of data associated with ASD may also be incorporated within these basic processes, including how ASD risk factors such as maternal obesity and preeclampsia, as well as more general prenatal stressors, modulate the likelihood of offspring ASD. CONCLUSION Such a mitochondria-focussed integrated model of the pathophysiology of ASD has important preventative and treatment implications.
Collapse
Affiliation(s)
- Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - George Anderson
- CRC Scotland & London, Eccleston Square, London, United Kingdom
| | | | - Moonsang Seo
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Johanna O Ojala
- Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
20
|
Supplementing the maternal diet of rats with butyrate enhances mitochondrial biogenesis in the skeletal muscles of weaned offspring. Br J Nutr 2017; 117:12-20. [DOI: 10.1017/s0007114516004402] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AbstractThe present study aimed to investigate the effects of maternal dietary butyrate supplementation on energy metabolism and mitochondrial biogenesis in offspring skeletal muscle and the possible mediating mechanisms. Virgin female rats were randomly assigned to either control or butyrate diets (1 % butyrate sodium) throughout gestation and lactation. At the end of lactation (21 d), the offspring were killed by exsanguination from the abdominal aorta under anaesthesia. The results showed that maternal butyrate supplementation throughout gestation and lactation did not affect offspring body weight. However, the protein expressions of G-protein-coupled receptors (GPR) 43 and 41 were significantly enhanced in offspring skeletal muscle of the maternal butyrate-supplemented group. The ATP content, most of mitochondrial DNA-encoded gene expressions, the cytochrome c oxidase subunit 1 and 4 protein contents and the mitochondrial DNA copy number were significantly higher in the butyrate group than in the control group. Meanwhile, the protein expressions of type 1 myosin heavy chain, mitochondrial transcription factor A, PPAR-coactivator-1α (PGC-1α) and uncoupling protein 3 were significantly increased in the gastrocnemius muscle of the treatment group compared with the control group. These results indicate for the first time that maternal butyrate supplementation during the gestation and lactation periods influenced energy metabolism and mitochondrial biogenesis through the GPR and PGC-1α pathways in offspring skeletal muscle at weaning.
Collapse
|
21
|
Gomez-Arango LF, Barrett HL, McIntyre HD, Callaway LK, Morrison M, Dekker Nitert M. Increased Systolic and Diastolic Blood Pressure Is Associated With Altered Gut Microbiota Composition and Butyrate Production in Early Pregnancy. Hypertension 2016; 68:974-81. [PMID: 27528065 DOI: 10.1161/hypertensionaha.116.07910] [Citation(s) in RCA: 277] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/13/2016] [Indexed: 12/11/2022]
Abstract
The risk of developing pregnancy-induced hypertension and preeclampsia is higher in obese pregnant women. In obesity, the composition of the gut microbiota is altered. Obesity is also associated with low-grade inflammation. Metabolites from the gut microbiota may contribute to both hypertension and inflammation. The aim of this study is to investigate whether the composition of the gut microbiota in overweight and obese pregnant women is associated with blood pressure and levels of plasminogen activator inhibitor-1. The composition of the gut microbiota was determined with 16S ribosomal RNA sequencing in 205 women at 16 weeks gestation from the SPRING study (the Study of Probiotics in Gestational Diabetes). Expression of butyrate-producing genes in the gut microbiota was assessed by real-time polymerase chain reaction. Plasminogen activator inhibitor-1 levels were measured in fasting serum of a subset of 70 women. Blood pressure was slightly but significantly higher in obese compared with overweight women. The abundance of the butyrate-producing genus Odoribacter was inversely correlated with systolic blood pressure. Butyrate production capacity was decreased, but plasminogen activator inhibitor-1 concentrations increased in obese pregnant women. Plasminogen activator inhibitor-1 levels were inversely correlated with expression of butyrate kinase and Odoribacter abundance. This study shows that in overweight and obese pregnant women at 16 weeks gestation, the abundance of butyrate-producing bacteria and butyrate production in the gut microbiota is significantly negatively associated with blood pressure and with plasminogen activator inhibitor-1 levels. Increasing butyrate-producing capacity may contribute to maintenance of normal blood pressure in obese pregnant women.
Collapse
Affiliation(s)
- Luisa F Gomez-Arango
- From the School of Medicine (L.F.G.-A., H.L.B., H.D.M., L.K.C., M.D.N.), UQ Centre for Clinical Research (L.F.G.-A., H.L.B., L.K.C., M.D.N.), Mater Research Institute (H.D.M.), and Diamantina Institute, Faculty of Medicine and Biomedical Sciences (M.M.), The University of Queensland, Brisbane, Australia; and Obstetric Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia (H.L.B., L.K.C.)
| | - Helen L Barrett
- From the School of Medicine (L.F.G.-A., H.L.B., H.D.M., L.K.C., M.D.N.), UQ Centre for Clinical Research (L.F.G.-A., H.L.B., L.K.C., M.D.N.), Mater Research Institute (H.D.M.), and Diamantina Institute, Faculty of Medicine and Biomedical Sciences (M.M.), The University of Queensland, Brisbane, Australia; and Obstetric Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia (H.L.B., L.K.C.)
| | - H David McIntyre
- From the School of Medicine (L.F.G.-A., H.L.B., H.D.M., L.K.C., M.D.N.), UQ Centre for Clinical Research (L.F.G.-A., H.L.B., L.K.C., M.D.N.), Mater Research Institute (H.D.M.), and Diamantina Institute, Faculty of Medicine and Biomedical Sciences (M.M.), The University of Queensland, Brisbane, Australia; and Obstetric Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia (H.L.B., L.K.C.)
| | - Leonie K Callaway
- From the School of Medicine (L.F.G.-A., H.L.B., H.D.M., L.K.C., M.D.N.), UQ Centre for Clinical Research (L.F.G.-A., H.L.B., L.K.C., M.D.N.), Mater Research Institute (H.D.M.), and Diamantina Institute, Faculty of Medicine and Biomedical Sciences (M.M.), The University of Queensland, Brisbane, Australia; and Obstetric Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia (H.L.B., L.K.C.)
| | - Mark Morrison
- From the School of Medicine (L.F.G.-A., H.L.B., H.D.M., L.K.C., M.D.N.), UQ Centre for Clinical Research (L.F.G.-A., H.L.B., L.K.C., M.D.N.), Mater Research Institute (H.D.M.), and Diamantina Institute, Faculty of Medicine and Biomedical Sciences (M.M.), The University of Queensland, Brisbane, Australia; and Obstetric Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia (H.L.B., L.K.C.)
| | - Marloes Dekker Nitert
- From the School of Medicine (L.F.G.-A., H.L.B., H.D.M., L.K.C., M.D.N.), UQ Centre for Clinical Research (L.F.G.-A., H.L.B., L.K.C., M.D.N.), Mater Research Institute (H.D.M.), and Diamantina Institute, Faculty of Medicine and Biomedical Sciences (M.M.), The University of Queensland, Brisbane, Australia; and Obstetric Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia (H.L.B., L.K.C.).
| | | |
Collapse
|
22
|
Zhou J, Gao S, Chen J, Zhao R, Yang X. Maternal sodium butyrate supplement elevates the lipolysis in adipose tissue and leads to lipid accumulation in offspring liver of weaning-age rats. Lipids Health Dis 2016; 15:119. [PMID: 27449927 PMCID: PMC4957328 DOI: 10.1186/s12944-016-0289-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 07/12/2016] [Indexed: 01/22/2023] Open
Abstract
Background Sodium butyrate (SB) is reported to regulate lipid metabolism in mammals, and the relationship between maternal nutrition and offspring growth has drawn much attention in the last several years. Methods To elucidate the effects of maternal dietary SB supplementation on hepatic lipid metabolism in weaning rats, we fed 16 primiparous purebred female SD rats either a chow-diet or a 1 % sodium butyrate diet throughout pregnancy and lactation. At weaning age, samples of the maternal subcutaneous adipose tissue and offspring liver were taken. The serum indexes and expressions of proteins related to lipid metabolism were detected in the mother and offspring, respectively. Results The results showed that the maternal SB supplement increased the concentration of non-esterified fatty acid (NEFA) in the maternal and offspring serum (P < 0.05). Total cholesterol (Tch) increased significantly in the weaning-rat serum (P < 0.05). Maternal adipose tissue from the SB-supplemented rats showed higher content of protein G-coupled protein (GPR43) and protein kinase A (PKA) (P < 0.05). The expression of protein adipose triglyceride lipase (ATGL), and of total and phosphorylated hormone sensitive lipase (HSL), in the maternal adipose tissue increased significantly (P < 0.05) compared to the control group. However the proteins related to lipogenesis showed no significant changes. Moreover, the concentration of triglyceride in the offspring liver increased significantly, and this likely resulted from an increase in the levels of fatty acids binding protein (FABP) and fatty acid translocase (CD36) protein (P < 0.05). SB exposure during pregnancy and lactation increased the hepatic total cholesterol (Tch) content (P < 0.01), which was related to a significantly up-regulated offspring hepatic expression of low density lipoprotein receptor (LDLR) protein (P < 0.05). Conclusion These results indicate that a maternal SB supplement during pregnancy and the lactation period promotes maternal fat mobilization, which may result in fatty acid uptake and lipid accumulation in the liver of the offspring.
Collapse
Affiliation(s)
- Jiabin Zhou
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Shixing Gao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Jinglong Chen
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.,Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing, 210095, People's Republic of China
| | - Xiaojing Yang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| |
Collapse
|
23
|
Effects of sodium butyrate supplementation on reproductive performance and colostrum composition in gilts. Animal 2016; 10:1722-7. [DOI: 10.1017/s1751731116000537] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|