1
|
Chi Z, Zhang M, Fu B, Wang X, Yang H, Fang X, Li Z, Teng T, Shi B. Branched Short-Chain Fatty Acid-Rich Fermented Protein Food Improves the Growth and Intestinal Health by Regulating Gut Microbiota and Metabolites in Young Pigs. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:21594-21609. [PMID: 39303156 DOI: 10.1021/acs.jafc.4c04526] [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: 09/22/2024]
Abstract
The diet in early life is essential for the growth and intestinal health later in life. However, beneficial effects of a diet enriched in branched short-chain fatty acids (BSCFAs) for infants are ambiguous. This study aimed to develop a novel fermented protein food, enriched with BSCFAs and assess the effects of dry and wet ferment products on young pig development, nutrient absorption, intestinal barrier function, and gut microbiota and metabolites. A total of 18 young pigs were randomly assigned to three groups. The dry corn gluten-wheat bran mixture (DFCGW) and wet corn gluten-wheat bran mixture (WFCGW) were utilized as replacements for 10% soybean meal in the basal diet. Our results exhibited that the WFCGW diet significantly increased the growth performance of young pigs, enhanced the expression of tight junction proteins, and regulated associated cytokines expression in the colonic mucosa. Simultaneously, the WFCGW diet led to elevated levels of colonic isobutyric and isovaleric acid, as well as the activation of GPR41 and GPR109A. Furthermore, more potential probiotics including Lactobacillus, Megasphaera, and Lachnospiraceae_ND3007_group were enriched in the WFCGW group and positively associated with the beneficial metabolites such as 5-hydroxyindole-3-acetic acid. Differential metabolite KEGG pathway analysis suggested that WFCGW might exert gut health benefits by modulating tryptophan metabolism. In addition, the WFCGW diet significantly increased ghrelin concentrations in serum and hypothalamus and promoted the appetite of young pigs by activating hypothalamic NPY/AGRP neurons. This study extends the knowledge of BSCFAs and provides a reference for the fermented food application in the infant diet.
Collapse
Affiliation(s)
- Zihan Chi
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Mengqi Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Botao Fu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoxu Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Hao Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xiuyu Fang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Zhongyu Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Teng Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Baoming Shi
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
2
|
Zeng X, Wang M, Chen L, Zheng B. Impact of using whole chestnut flour as a substitute for cake flour on digestion, functional and storage properties of chiffon cake: A potential application study. Food Chem 2024; 432:137016. [PMID: 37647706 DOI: 10.1016/j.foodchem.2023.137016] [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: 03/27/2023] [Revised: 07/08/2023] [Accepted: 07/24/2023] [Indexed: 09/01/2023]
Abstract
Developing fresh cake product with rich nutrition and high quality has become a hot spot in food industry. In this study, whole chestnut flour as a high-quality dietary source was successfully substituted for cake flour in the production of chestnut chiffon cake with 40-55% substitution rate, and its application prospects were further evaluated through studying nutritional and storage properties. The results showed that chestnut chiffon cake with 45% and 50% substitution rate could significantly increase the resistant component, scavenging activity and ferric reducing antioxidant power, surprisingly decrease predicted glycemic index to 54.05-57.28, and reduce the acetate/propionate ratio and Firmicutes/Bacteroidetes value for human gut microbiota as well. Comparatively, chestnut chiffon cake with 45% substitution rate had more application potential due to its higher free water retention at day 7 and higher resilience throughout the storage time. Overall, this study could provide valuable information for the development of modern nutritional cake industry.
Collapse
Affiliation(s)
- Xixi Zeng
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China
| | - Menghui Wang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China
| | - Ling Chen
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China.
| | - Bo Zheng
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
3
|
Koziol A, Odriozola I, Leonard A, Eisenhofer R, San José C, Aizpurua O, Alberdi A. Mammals show distinct functional gut microbiome dynamics to identical series of environmental stressors. mBio 2023; 14:e0160623. [PMID: 37650630 PMCID: PMC10653949 DOI: 10.1128/mbio.01606-23] [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: 06/24/2023] [Accepted: 07/24/2023] [Indexed: 09/01/2023] Open
Abstract
IMPORTANCE In our manuscript, we report the first interspecific comparative study about the plasticity of the gut microbiota. We conducted a captivity experiment that exposed wild-captured mammals to a series of environmental challenges over 45 days. We characterized their gut microbial communities using genome-resolved metagenomics and modeled how the taxonomic, phylogenetic, and functional microbial dynamics varied across a series of disturbances in both species. Our results indicate that the intrinsic properties (e.g., diversity and functional redundancy) of microbial communities coupled with physiological attributes (e.g., thermal plasticity) of hosts shape the taxonomic, phylogenetic, and functional response of gut microbiomes to environmental stressors, which might influence their contribution to the acclimation and adaptation capacity of animal hosts.
Collapse
Affiliation(s)
- Adam Koziol
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| | - Iñaki Odriozola
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| | - Aoife Leonard
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| | - Raphael Eisenhofer
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| | - Carlos San José
- Biodonostia Health Research Institute, Donostia-San Sebastian, Spain
| | - Ostaizka Aizpurua
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| | - Antton Alberdi
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Chen CY, Ho HC. Roles of gut microbes in metabolic-associated fatty liver disease. Tzu Chi Med J 2023; 35:279-289. [PMID: 38035063 PMCID: PMC10683521 DOI: 10.4103/tcmj.tcmj_86_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 12/02/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is the most common chronic liver disease. Gut dysbiosis is considered a significant contributing factor in disease development. Increased intestinal permeability can be induced by gut dysbiosis, followed by the entry of lipopolysaccharide into circulation to reach peripheral tissue and result in chronic inflammation. We reviewed how microbial metabolites push host physiology toward MAFLD, including short-chain fatty acids (SCFAs), bile acids, and tryptophan metabolites. The effects of SCFAs are generally reported as anti-inflammatory and can improve intestinal barrier function and restore gut microbiota. Gut microbes can influence intestinal barrier function through SCFAs produced by fermentative bacteria, especially butyrate and propionate producers. This is achieved through the activation of free fatty acid sensing receptors. Bile is directly involved in lipid absorption. Gut microbes can alter bile acid composition by bile salt hydrolase-producing bacteria and bacterial hydroxysteroid dehydrogenase-producing bacteria. These bile acids can affect host physiology by activating farnesoid X receptor Takeda G protein-coupled receptor 5. Gut microbes can also induce MAFLD-associated symptoms by producing tryptophan metabolites kynurenine, serotonin, and indole-3-propionate. A summary of bacterial genera involved in SCFAs production, bile acid transformation, and tryptophan metabolism is provided. Many bacteria have demonstrated efficacy in alleviating MAFLD in animal models and are potential therapeutic candidates for MAFLD.
Collapse
Affiliation(s)
- Chun-Yao Chen
- Department of Biomedical Sciences and Engineering, Tzu Chi University, Hualien, Taiwan
| | - Han-Chen Ho
- Department of Anatomy, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
5
|
Xie Z, Yao M, Castro-Mejía JL, Ma M, Zhu Y, Fu X, Huang Q, Zhang B. Propionylated high-amylose maize starch alleviates obesity by modulating gut microbiota in high-fat diet-fed mice. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
|
6
|
Liu Y, Yang J, Liu X, Liu R, Wang Y, Huang X, Li Y, Liu R, Yang X. Dietary folic acid addition reduces abdominal fat deposition mediated by alterations in gut microbiota and SCFA production in broilers. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 12:54-62. [PMID: 36439290 PMCID: PMC9684696 DOI: 10.1016/j.aninu.2022.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/15/2022] [Accepted: 08/09/2022] [Indexed: 06/09/2023]
Abstract
Intensive selective breeding for high growth rate and body weight cause excess abdominal fat in broilers. Gut microbiota and folic acid were reported to regulate lipid metabolism. A total of 210 one-day-old broilers were divided into the control (folic acid at 1.3 mg/kg) and folic acid groups (folic acid at 13 mg/kg) to illustrate the effects of folic acid on growth performance, abdominal fat deposition, and gut microbiota, and the experiment lasted 28 d. Results revealed that dietary folic acid addition decreased abdominal fat percentage (P < 0.05) and down-regulated genes expression related to cell proliferation and differentiation in abdominal fat including IGF1, EGF, C/EBPα, PPARγ, PLIN1, FABP4 and PCNA (P < 0.05). Folic acid addition decreased caecal Firmicutes-to-Bacteroidetes ratio (P < 0.01) and increased the proportions of Alistipes, Oscillospira, Ruminococcus, Clostridium, Dehalobacterium and Parabacteroides (P < 0.05). Caecal acetic acid, and propionic acid contents were found to be higher under folic acid treatment (P < 0.05), which were negatively related to genes expression associated with adipocyte proliferation and differentiation (P < 0.05). Ruminococcus was positively correlated with caecal acetic acid content, and the same phenomenon was detected between propionic acid and Oscillospira and Ruminococcus (P < 0.05). Acetic acid and Oscillospira were identified to be negatively associated with abdominal fat percentage (P < 0.05). In conclusion, our data demonstrated that dietary supplementation of folic acid reduced fat deposition in broilers by inhibiting abdominal adipocyte proliferation and differentiation, which might be mediated by changes in gut microbiota and short chain fatty acid production.
Collapse
Affiliation(s)
- Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Jiantao Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaoying Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Rui Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yibin Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xinhuo Huang
- Nano Vitamin Engineering Research Center of Shaanxi Province, Xi'an 710000, China
| | - Yingge Li
- Shaanxi Province Animal Husbandry Technology Extension Station, Xi'an 710016, China
| | - Ruifang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
7
|
Xu M, Lan R, Qiao L, Lin X, Hu D, Zhang S, Yang J, Zhou J, Ren Z, Li X, Liu G, Liu L, Xu J. Bacteroides vulgatus Ameliorates Lipid Metabolic Disorders and Modulates Gut Microbial Composition in Hyperlipidemic Rats. Microbiol Spectr 2023; 11:e0251722. [PMID: 36625637 PMCID: PMC9927244 DOI: 10.1128/spectrum.02517-22] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Hyperlipidemia is a risk factor and key indicator for cardiovascular diseases, and the gut microbiota is highly associated with hyperlipidemia. Bacteroides vulgatus is a prevalent mutualist across human populations and confers multiple health benefits such as immunoregulation, antiobesity, and coronary artery disease intervention. However, its role in antihyperlipidemia has not been systematically characterized. This study sought to identify the effect of B. vulgatus Bv46 on hyperlipidemia. Hyperlipidemic rats were modeled by feeding them a high-fat diet for 6 weeks. The effect of B. vulgatus Bv46 supplementation was evaluated by measuring anthropometric parameters, lipid and inflammation markers, and the liver pathology. Multi-omics was used to explore the underlying mechanisms. The ability of B. vulgatus Bv46 to produce bile salt hydrolase was confirmed by gene annotation and in vitro experiments. Oral administration of B. vulgatus Bv46 in hyperlipidemic rats significantly reduced the body weight gain, food efficiency, and liver index, improved the serum lipid profile, lowered the levels of serum inflammatory cytokines, promoted the loss of fecal bile acids (BAs), and extended the fecal pool of short-chain fatty acids (SCFAs), especially propionate and butyrate. B. vulgatus Bv46 induced compositional shifts of the gut microbial community of hyperlipidemic rats, characterized by a lower ratio of Firmicutes to Bacteroidetes with an increase of genera Bacteroides and Parabacteroides. After intervention, serum metabolite profiling exhibited an adaptation in amino acids and glycerophospholipid metabolism. Transcriptomics further detected altered biological processes, including primary bile acid biosynthesis and fatty acid metabolic process. Taken together, the findings suggest that B. vulgatus Bv46 could be a promising candidate for interventions against hyperlipidemia. IMPORTANCE As a core microbe of the human gut ecosystem, Bacteroides vulgatus has been linked to multiple aspects of metabolic disorders in a collection of associative studies, which, while indicative, warrants more direct experimental evidence to verify. In this study, we experimentally demonstrated that oral administration of B. vulgatus Bv46 ameliorated the serum lipid profile and systemic inflammation of high-fat diet-induced hyperlipidemic rats in a microbiome-regulated manner, which appears to be associated with changes of bile acid metabolism, short-chain fatty acid biosynthesis, and serum metabolomic profile. This finding supports the causal contribution of B. vulgatus in host metabolism and helps to form the basis of novel therapies for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Mingchao Xu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruiting Lan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Lei Qiao
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaoying Lin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dalong Hu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Suping Zhang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jing Yang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
| | - Juan Zhou
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhihong Ren
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xianping Li
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guoxing Liu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liyun Liu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
| | - Jianguo Xu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Public Health, Nankai University, Tianjin, China
| |
Collapse
|
8
|
Sex Differences of Radiation Damage in High-Fat-Diet-Fed Mice and the Regulatory Effect of Melatonin. Nutrients 2022; 15:nu15010064. [PMID: 36615722 PMCID: PMC9823527 DOI: 10.3390/nu15010064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
The consumption of a high-fat diet (HFD) and exposure to ionizing radiation (IR) are closely associated with many diseases. To evaluate the interaction between HFDs and IR-induced injury, we gave mice whole abdominal irradiation (WAI) to examine the extent of intestinal injury under different dietary conditions. Melatonin (MLT) is a free radical scavenger that effectively prevents hematopoietic, immune, and gastrointestinal damage induced by IR. However, its effects on WAI-induced intestinal injury in HFD-fed mice remain unclear. We demonstrated that MLT can promote intestinal structural repair following WAI and enhance the regeneration capacity of Lgr5+ intestinal stem cells. In addition, we investigated the effects of radiation damage on sexual dimorphism in HFD-fed mice. The results showed that the degree of IR-induced intestinal injury was more severe in the HFD-fed female mice. MLT preserved the intestinal microbiota composition of HFD-fed mice and increased the abundance of Bacteroides and Proteobacteria in male and female mice, respectively. In conclusion, MLT may reduce the negative effects of HFD and IR, thereby providing assistance in preserving the structure and function of the intestine.
Collapse
|
9
|
Weng G, Huang J, Ma X, Song M, Yin Y, Deng D, Deng J. Brevibacillus laterosporus BL1, a promising probiotic, prevents obesity and modulates gut microbiota in mice fed a high-fat diet. Front Nutr 2022; 9:1050025. [PMID: 36505236 PMCID: PMC9729748 DOI: 10.3389/fnut.2022.1050025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
Scope Probiotics are a potential preventive strategy for obesity. However, with discrete efficacy and limited species of probiotics, there is a demand for novel strains with excellent anti-obesity properties. This study aimed to investigate the effects of Brevibacillus laterosporus BL1 on preventing obesity in high-fat diet (HFD)-fed mice. Methods and results C57BL/6 male mice were randomly assigned to four groups (n = 10) and fed a control diet, HFD, HFD plus B. laterosporus BL1, and HFD plus supernatant of B. laterosporus BL1, respectively for 8 weeks. The results showed that prophylactic B. laterosporus BL1 treatment reduced body weight gain by 41.26% in comparison to the HFD group, and this difference was accompanied by a reduction in body fat mass and the weight of inguinal white adipose tissues and epididymal white adipose tissue (-33.39%, -39.07%, and -43.75%, respectively). Moreover, the B. laterosporus BL1-mediated improvements in lipid profile, insulin resistance, and chronic inflammation were associated with the regulation of gene expression related to lipid metabolism and enhancement of brown adipose tissue thermogenesis. Particularly, B. laterosporus BL1 intervention significantly improved HFD-induced gut flora dysbiosis, as evidenced by a reverse in the relative abundance of Bacillota and Bacteroidota, as well as an increase in the relative abundance of bacteria that produce short-chain fatty acids (SCFAs), which in turn increased SCFAs levels. Conclusion Our findings found for the first time that B. laterosporus BL1 may be a promising probiotic for prevention of obesity associated with the regulation of gut microbiota.
Collapse
Affiliation(s)
- Guangying Weng
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, Guangdong, China,State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China
| | - Jian Huang
- Institute for Quality & Safety and Standards of Agricultural Products Research, Jiangxi Academy of Agricultural Sciences, Nanchang, Jiangxi, China
| | - Xianyong Ma
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China
| | - Min Song
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China
| | - Yulong Yin
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, Guangdong, China
| | - Dun Deng
- State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China,*Correspondence: Dun Deng,
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, Guangdong, China,Jinping Deng,
| |
Collapse
|
10
|
Sun C, Wang Z, Hu L, Zhang X, Chen J, Yu Z, Liu L, Wu M. Targets of statins intervention in LDL-C metabolism: Gut microbiota. Front Cardiovasc Med 2022; 9:972603. [PMID: 36158845 PMCID: PMC9492915 DOI: 10.3389/fcvm.2022.972603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Increasing researches have considered gut microbiota as a new “metabolic organ,” which mediates the occurrence and development of metabolic diseases. In addition, the liver is an important organ of lipid metabolism, and abnormal lipid metabolism can cause the elevation of blood lipids. Among them, elevated low-density lipoprotein cholesterol (LDL-C) is related with ectopic lipid deposition and metabolic diseases, and statins are widely used to lower LDL-C. In recent years, the gut microbiota has been shown to mediate statins efficacy, both in animals and humans. The effect of statins on microbiota abundance has been deeply explored, and the pathways through which statins reduce the LDL-C levels by affecting the abundance of microbiota have gradually been explored. In this review, we discussed the interaction between gut microbiota and cholesterol metabolism, especially the cholesterol-lowering effect of statins mediated by gut microbiota, via AMPK-PPARγ-SREBP1C/2, FXR and PXR-related, and LPS-TLR4-Myd88 pathways, which may help to explain the individual differences in statins efficacy.
Collapse
Affiliation(s)
- ChangXin Sun
- Beijing University of Chinese Medicine, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - ZePing Wang
- Beijing University of Chinese Medicine, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - LanQing Hu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - XiaoNan Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - JiYe Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - ZongLiang Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - LongTao Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: LongTao Liu
| | - Min Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Min Wu
| |
Collapse
|
11
|
Lv XC, Wu Q, Yuan YJ, Li L, Guo WL, Lin XB, Huang ZR, Rao PF, Ai LZ, Ni L. Organic chromium derived from the chelation of Ganoderma lucidum polysaccharide and chromium (III) alleviates metabolic syndromes and intestinal microbiota dysbiosis induced by high-fat and high-fructose diet. Int J Biol Macromol 2022; 219:964-979. [DOI: 10.1016/j.ijbiomac.2022.07.211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/10/2022] [Accepted: 07/26/2022] [Indexed: 11/05/2022]
|
12
|
Feng Y, Liu D, Liu Y, Yang X, Zhang M, Wei F, Li D, Hu Y, Guo Y. Host-genotype-dependent cecal microbes are linked to breast muscle metabolites in Chinese chickens. iScience 2022; 25:104469. [PMID: 35707722 PMCID: PMC9189123 DOI: 10.1016/j.isci.2022.104469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 04/08/2022] [Accepted: 05/20/2022] [Indexed: 11/18/2022] Open
Abstract
In chickens, the effect of host genetics on the gut microbiota is not fully understood, and the extent to which the heritable gut microbes affect chicken metabolism and physiology is still an open question. Here, we explored the interactions among chicken genetics, the cecal microbiota and metabolites in breast muscle from ten chicken breeds in China. We found that different chicken breeds displayed distinct cecal microbial community structures and functions, and 15 amplicon sequence variants (ASVs) were significantly associated with host genetics through different genetic loci, such as those related to the intestinal barrier function. We identified five heritable ASVs significantly associated with 53 chicken muscle metabolites, among which the Megamonas probably affected lipid metabolism through the production of propionate. Our study revealed that the chicken genetically associated cecal microbes may have the potential to affect the bird’s physiology and metabolism. The cecal microbiota are different among ten chicken breeds The chicken genetics influences the cecal microbiota structures and functions The chicken heritable cecal microbes are associated with muscle metabolites Megamonas may affect lipid metabolism by the production of propionate
Collapse
Affiliation(s)
- Yuqing Feng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Dan Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Yan Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xinyue Yang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Meihong Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Fuxiao Wei
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Depeng Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Yongfei Hu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
- Corresponding author
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
- Corresponding author
| |
Collapse
|
13
|
Hitch TCA, Hall LJ, Walsh SK, Leventhal GE, Slack E, de Wouters T, Walter J, Clavel T. Microbiome-based interventions to modulate gut ecology and the immune system. Mucosal Immunol 2022; 15:1095-1113. [PMID: 36180583 PMCID: PMC9705255 DOI: 10.1038/s41385-022-00564-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/12/2022] [Accepted: 08/22/2022] [Indexed: 02/04/2023]
Abstract
The gut microbiome lies at the intersection between the environment and the host, with the ability to modify host responses to disease-relevant exposures and stimuli. This is evident in how enteric microbes interact with the immune system, e.g., supporting immune maturation in early life, affecting drug efficacy via modulation of immune responses, or influencing development of immune cell populations and their mediators. Many factors modulate gut ecosystem dynamics during daily life and we are just beginning to realise the therapeutic and prophylactic potential of microbiome-based interventions. These approaches vary in application, goal, and mechanisms of action. Some modify the entire community, such as nutritional approaches or faecal microbiota transplantation, while others, such as phage therapy, probiotics, and prebiotics, target specific taxa or strains. In this review, we assessed the experimental evidence for microbiome-based interventions, with a particular focus on their clinical relevance, ecological effects, and modulation of the immune system.
Collapse
Affiliation(s)
- Thomas C A Hitch
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Lindsay J Hall
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, UK
- Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Sarah Kate Walsh
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
- APC Microbiome Ireland, School of Microbiology and Department of Medicine, University College Cork, Cork, Ireland
| | | | - Emma Slack
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | | | - Jens Walter
- APC Microbiome Ireland, School of Microbiology and Department of Medicine, University College Cork, Cork, Ireland
| | - Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany.
| |
Collapse
|
14
|
Weng G, Duan Y, Zhong Y, Song B, Zheng J, Zhang S, Yin Y, Deng J. Plant Extracts in Obesity: A Role of Gut Microbiota. Front Nutr 2021; 8:727951. [PMID: 34631766 PMCID: PMC8495072 DOI: 10.3389/fnut.2021.727951] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity has become one of the most serious chronic diseases threatening human health. Its occurrence and development are closely associated with gut microbiota since the disorders of gut microbiota can promote endotoxin production and induce inflammatory response. Recently, numerous plant extracts have been proven to mitigate lipid dysmetabolism and obesity syndrome by regulating the abundance and composition of gut microbiota. In this review, we summarize the potential roles of different plant extracts including mulberry leaf extract, policosanol, cortex moutan, green tea, honokiol, and capsaicin in regulating obesity via gut microbiota. Based on the current findings, plant extracts may be promising agents for the prevention and treatment of obesity and its related metabolic diseases, and the mechanisms might be associated with gut microbiota.
Collapse
Affiliation(s)
- Guangying Weng
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, China.,CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yehui Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yinzhao Zhong
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Bo Song
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jie Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shiyu Zhang
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yulong Yin
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, China.,CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, China
| |
Collapse
|
15
|
Luo Z, Zhao Y, Zeng L, Yin J, Zeng Q, Li X, He J, Wang J, Tan B. Effects of Fermented Radix puerariae Residue on Nutrient Digestibility and Reproductive Performance of Sows. Front Nutr 2021; 8:715713. [PMID: 34527689 PMCID: PMC8435608 DOI: 10.3389/fnut.2021.715713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/05/2021] [Indexed: 11/19/2022] Open
Abstract
This study was conducted to investigate the effect of fermented Radix puerariae residue (FRPR) on reproductive performance, apparent total tract digestibility (ATTD) of nutrients, and fecal short-chain fatty acid (SCFA) contents of sows. A total of 36 landrace × large white multiparous sows were randomly arranged into three treatments, representing supplementation with 0, 2, and 4% FRPR to a corn-soybean meal and wheat bran-based diet during the whole gestation period. The results showed that dietary FRPR had no effects on litter size and the number of total alive piglets (P > 0.05), and that the number of weaned piglets and weaning weight of litter were increased in sows with 4% FRPR treatment compared with control treatment (P < 0.05). Dietary 4% FRPR significantly decreased constipation rate, improved the ATTD of dry matter and organics, and fecal contents of acetate, propionate, and total SCFAs (P < 0.05). In the offspring piglets, serum concentrations of total protein, alkaline phosphatase, IgG, IL-10, and TGF-β were increased, but blood urea nitrogen content was decreased with 4% FRPR treatment (P < 0.05). There were no significant differences in all determined indexes except for fecal acetic acid and total SCFAs between control and 2% FRPR treatment (P > 0.05). These findings indicated that FRPR used in the diets of sows showed positive effects on fecal characteristics, utilization of nutrients, and reproductive performance. Maternal supplementation with 4% FRPR is recommended for improving immune responses, weaning litter size, and litter weight of offspring piglets, which provide useful information for the application of residues of R. puerariae.
Collapse
Affiliation(s)
- Zhenfu Luo
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yuanyuan Zhao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Liming Zeng
- College of Animal Science, Jiangxi Agricultural University, Nanchang, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Qinghua Zeng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xilong Li
- Key Laboratory of Feed Biotechnology, The Ministry of Agriculture of the People's Republic of China, Beijing, China
| | - Jianhua He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Jing Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Bi'e Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| |
Collapse
|
16
|
Wang Y, Zhou Y, Fu J. Advances in antiobesity mechanisms of capsaicin. Curr Opin Pharmacol 2021; 61:1-5. [PMID: 34537583 DOI: 10.1016/j.coph.2021.08.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 02/07/2023]
Abstract
Obesity is a global epidemic that affects the lives and health of millions of people. The prevention and treatment of obesity have become a significant public health challenge worldwide. Numerous studies showed that the gut microbiota is associated with the development of obesity, and the regulatory mechanisms mediating the relationship between gut microbiota and obesity have become an intense research area. Capsaicin is a vanilla amide alkaloid that is an active ingredient in pepper. Much research demonstrated the antiobesity activity of capsaicin. This article reviews recent research on the antiobesity mechanisms of capsaicin involving alterations of the gut microbial composition, reduction of intestinal permeability, and regulation of the microbiome-gut-brain axis. This summary will establish a basis for further developing capsaicin as an ingredient in medications and health products.
Collapse
Affiliation(s)
- Yuanwei Wang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China.
| | - Yahan Zhou
- School of Light Industry, Beijing Technology and Business University, Beijing 100048, PR China
| | - Jia Fu
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, PR China.
| |
Collapse
|
17
|
Zheng J, Zheng C, Song B, Guo Q, Zhong Y, Zhang S, Zhang L, Duan G, Li F, Duan Y. HMB Improves Lipid Metabolism of Bama Xiang Mini-Pigs via Modulating the Bacteroidetes-Acetic Acid-AMPKα Axis. Front Microbiol 2021; 12:736997. [PMID: 34484171 PMCID: PMC8415715 DOI: 10.3389/fmicb.2021.736997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 07/28/2021] [Indexed: 12/29/2022] Open
Abstract
Here, we used Bama Xiang mini-pigs to explore the effects of different dietary β-hydroxy-β-methylbutyrate (HMB) levels (0, 0.13, 0.64 or 1.28%) on lipid metabolism of adipose tissue. Results showed that HMB decreased the fat percentage of pigs (linearly, P < 0.05), and the lowest value was observed in the 0.13% HMB group. Moreover, the colonic acetic acid concentration and the relative Bacteroidetes abundance were increased in response to HMB supplementation (P < 0.05). Correlation analysis identified a positive correlation between the relative Bacteroidetes abundance and acetic acid production, and a negative correlation between fat percentage and the relative Bacteroidetes abundance or acetic acid production. HMB also upregulated the phosphorylation (p) of AMPKα, Sirt1, and FoxO1, and downregulated the p-mTOR expression. Collectively, these findings indicate that reduced fat percentage in Bama Xiang mini-pigs could be induced by HMB supplementation and the mechanism might be associated with the Bacteroidetes-acetic acid-AMPKα axis.
Collapse
Affiliation(s)
- Jie Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Changbing Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Bo Song
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qiuping Guo
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yinzhao Zhong
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Shiyu Zhang
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Lingyu Zhang
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Geyan Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Fengna Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yehui Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
18
|
Tobin D, Vige R, Calder PC. Review: The Nutritional Management of Multiple Sclerosis With Propionate. Front Immunol 2021; 12:676016. [PMID: 34394076 PMCID: PMC8355737 DOI: 10.3389/fimmu.2021.676016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/14/2021] [Indexed: 12/13/2022] Open
Abstract
Over the last 15 years there has been an accumulation of data supporting the concept of a gut-brain axis whereby dysbiosis of the gut microbiota can impact neurological function. Such dysbiosis has been suggested as a possible environmental exposure triggering multiple sclerosis (MS). Dysbiosis has been consistently shown to result in a reduction in short-chain fatty acid (SCFA) producing bacteria and a reduction in stool and plasma levels of propionate has been shown for MS patients independent of disease stage and in different geographies. A wealth of evidence supports the action of propionate on T-cell activity, resulting in decreased T-helper cell 1 (Th1) and T-helper cell 17 (Th17) numbers/activity and increased regulatory T cell (Treg cell) numbers/activity and an overall anti-inflammatory profile. These different T-cell populations play various roles in the pathophysiology of MS. A recent clinical study in MS patients demonstrated that supplementation of propionate reduces the annual relapse rate and slows disease progression. This review discusses this data and the relevant mechanistic background and discusses whether taming of the overactive immune system in MS is likely to allow easier bacterial and viral infection.
Collapse
Affiliation(s)
| | | | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, United Kingdom
| |
Collapse
|
19
|
Xie Z, Ding L, Huang Q, Fu X, Liu F, Dhital S, Zhang B. In vitro colonic fermentation profiles and microbial responses of propionylated high-amylose maize starch by individual Bacteroides-dominated enterotype inocula. Food Res Int 2021; 144:110317. [PMID: 34053522 DOI: 10.1016/j.foodres.2021.110317] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/13/2021] [Accepted: 03/14/2021] [Indexed: 12/28/2022]
Abstract
The concept of "enterotype" has been proposed to differentiate the gut microbiota between individual humans, and different dominant bacteria utilize fiber substrates with different fermentation properties and microbial changes. In this study, we made propionylated high-amylose maize starch and investigated both in vitro fecal fermentation properties and microbial responses by individual Bacteroides-dominated enterotype inocula. Propionyl group substitution of HAMS did not significantly change gas production profiles, suggesting that the gas production during fermentation is independent of propionylation. The final concentration of released propionate significantly increased (10.26-12.60 mM) as a function of propionylation degree, suggesting that the introduced propionyl groups can increase the concentration of short-chain fatty acids (SCFA) during colonic fermentation. At the genus level, Bacteroides was obviously promoted for all donors with the final abundance in the range of 0.1-0.24, indicating that propionylated high-amylose maize starch changed the structure and abundance of microbiota compared to unmodified starch. Besides, the non-metric dimensional scoring (NMDS) plots showed that those changes were related to the initial microbiota composition. The results may offer useful information for the design of personalized food products and relevant therapies at least within Bacteroides-dominated enterotype.
Collapse
Affiliation(s)
- Zhuqing Xie
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Li Ding
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Qiang Huang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China; SCUT-Zhuhai Institute of Modern Industrial Innovation, Zhuhai 519175, China; Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou 510640, China
| | - Xiong Fu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China; SCUT-Zhuhai Institute of Modern Industrial Innovation, Zhuhai 519175, China; Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou 510640, China
| | - Feitong Liu
- H&H Group Global Research and Technology Center, Guangzhou 510700, China
| | - Sushil Dhital
- Department of Chemical Engineering, Monash University, Clayton Campus, VIC 3800, Australia
| | - Bin Zhang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China; SCUT-Zhuhai Institute of Modern Industrial Innovation, Zhuhai 519175, China; Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou 510640, China; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.
| |
Collapse
|
20
|
Chen D, Gao ZQ, Wang YY, Wan BB, Liu G, Chen JL, Wu YX, Zhou Q, Jiang SY, Yu RQ, Pang QF. Sodium Propionate Enhances Nrf2-Mediated Protective Defense Against Oxidative Stress and Inflammation in Lipopolysaccharide-Induced Neonatal Mice. J Inflamm Res 2021; 14:803-816. [PMID: 33732006 PMCID: PMC7957230 DOI: 10.2147/jir.s303105] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 02/23/2021] [Indexed: 12/27/2022] Open
Abstract
Background Alveolar arrest and the impaired angiogenesis caused by chronic inflammation and oxidative stress are two main factors in bronchopulmonary dysplasia (BPD). Short-chain fatty acids (SCFAs), especially propionate, possess anti-oxidant and anti-inflammatory effects. The present study was designed to examine the roles of sodium propionate (SP) on lipopolysaccharide (LPS)-challenged BPD and its potential mechanisms. Methods WT, Nrf2-/- mice and pulmonary microvascular endothelial cells (HPMECs) were used in this study. LPS was performed to mimic BPD model both in vivo and vitro. Lung histopathology, inflammation and oxidative stress-related mRNA expressions in lungs involved in BPD pathogenesis were investigated. In addition, cell viability and angiogenesis were also tested. Results The increased nuclear factor erythroid 2-related factor (Nrf2) and decreased Kelch-like ECH-associated protein-1 (Keap-1) expressions were observed after SP treatment in the LPS-induced neonatal mouse model of BPD. In LPS-induced wild-type but not Nrf2-/- neonatal mice, SP reduced pulmonary inflammation and oxidative stress and exhibited obvious pathological alterations of the alveoli. Moreover, in LPS-evoked HPMECs, SP accelerated Nrf2 nuclear translocation presented and exhibited cytoprotective and pro-angiogenesis effects. In addition, SP diminished the LPS-induced inflammatory response by blocking the activation of nuclear factor-kappa B pathway. Moreover, pretreatment with ML385, an Nrf2 specific inhibitor, offsets the beneficial effects of SP on inflammation, oxidative stress and angiogenesis in LPS-evoked HPMECs. Conclusion SP protects against LPS-induced lung alveolar simplification and abnormal angiogenesis in neonatal mice and HPMECs in an Nrf2-dependent manner.
Collapse
Affiliation(s)
- Dan Chen
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Zhi-Qi Gao
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Ying-Ying Wang
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Bin-Bin Wan
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Gang Liu
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Jun-Liang Chen
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Ya-Xian Wu
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Qin Zhou
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, Jiangsu Province, People's Republic of China
| | - Shan-Yu Jiang
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, Jiangsu Province, People's Republic of China
| | - Ren-Qiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, Jiangsu Province, People's Republic of China
| | - Qing-Feng Pang
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| |
Collapse
|
21
|
Modulation of Short-Chain Fatty Acids as Potential Therapy Method for Type 2 Diabetes Mellitus. ACTA ACUST UNITED AC 2021; 2021:6632266. [PMID: 33488888 PMCID: PMC7801078 DOI: 10.1155/2021/6632266] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/07/2020] [Accepted: 12/18/2020] [Indexed: 12/25/2022]
Abstract
In recent years, the relationship between intestinal microbiota (IM) and the pathogenesis of type 2 diabetes mellitus (T2DM) has attracted much attention. The beneficial effects of IM on the metabolic phenotype of the host are often considered to be mediated by short-chain fatty acids (SCFAs), mainly acetate, butyrate, and propionate, the small-molecule metabolites derived from microbial fermentation of indigestible carbohydrates. SCFAs not only have an essential role in intestinal health but might also enter the systemic circulation as signaling molecules affecting the host's metabolism. In this review, we summarize the effects of SCFAs on glucose homeostasis and energy homeostasis and the mechanism through which SCFAs regulate the function of metabolically active organs (brain, liver, adipose tissue, skeletal muscle, and pancreas) and discuss the potential role of modulation of SCFAs as a therapeutic method for T2DM.
Collapse
|
22
|
Ma J, Zheng Y, Tang W, Yan W, Nie H, Fang J, Liu G. Dietary polyphenols in lipid metabolism: A role of gut microbiome. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2020; 6:404-409. [PMID: 33364456 PMCID: PMC7750795 DOI: 10.1016/j.aninu.2020.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/19/2020] [Accepted: 08/23/2020] [Indexed: 12/19/2022]
Abstract
Polyphenols are a class of non-essential phytonutrients, which are abundant in fruits and vegetables. Dietary polyphenols or foods rich in polyphenols are widely recommended for metabolic health. Indeed, polyphenols (i.e., catechins, resveratrol, and curcumin) are increasingly recognized as a regulator of lipid metabolism in host. The mechanisms, at least in part, may be highly associated with gut microbiome. This review mainly discussed the beneficial effects of dietary polyphenols on lipid metabolism. The potential mechanisms of gut microbiome are focused on the effect of dietary polyphenols on gut microbiota compositions and how gut microbiota affect polyphenol metabolism. Together, dietary polyphenols may be a useful nutritional strategy for manipulation of lipid metabolism or obesity care.
Collapse
Affiliation(s)
- Jie Ma
- College of Bioscience and Biotechnology, College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
| | - Yongmin Zheng
- College of Bioscience and Biotechnology, College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
| | - Wenjie Tang
- College of Bioscience and Biotechnology, College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
- Sichuan Academy of Animal Sciences, Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Wenxin Yan
- College of Bioscience and Biotechnology, College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
| | - Houfu Nie
- Agricultural and Rural Office of Chunkou Town, Liuyang, Hunan, China
| | - Jun Fang
- College of Bioscience and Biotechnology, College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
| | - Gang Liu
- College of Bioscience and Biotechnology, College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
| |
Collapse
|
23
|
Li H, Zhao L, Liu S, Zhang Z, Wang X, Lin H. Propionate inhibits fat deposition via affecting feed intake and modulating gut microbiota in broilers. Poult Sci 2020; 100:235-245. [PMID: 33357686 PMCID: PMC7772713 DOI: 10.1016/j.psj.2020.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/03/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
As one of the 3 main short-chain fatty acids, the role of propionate in chicken fat metabolism is largely unknown. In this study, we demonstrated that dietary supplementation of coated sodium propionate (SP) moderately inhibits fat deposition in broiler chickens, as evidenced by the decreased adipocyte mean area (P < 0.01), the lowered triglyceride content in abdominal fat tissue (P < 0.01), and the reduced transcription of several lipogenic genes in liver and abdominal fat tissues (P < 0.05). Surprisingly, the propionate content was not significantly elevated either in serum or in the cecal chyme by SP administration (P > 0.05). However, SP application significantly decreased the average daily feed intake of broilers (P < 0.05). In addition, the composition of the cecal microbial communities was altered, with the ratio of Firmicutes to Bacteroidetes decreasing in particular (P < 0.05). At the genus level, SP application increased the richness of Alistipes, Lactobacillus, and Bifidobacterium, while reduced the abundance of Lachnospiraceae and Helicobacter significantly (P < 0.05). Moreover, in vitro experiments indicated that, although physiological concentrations of propionate (0.01 to 0.1 mmol) upregulated or downregulated the transcription of some fat synthesis-associated genes (P < 0.05), they did not significantly affect the triglyceride accumulation in hepatocytes and adipocytes (P > 0.05). These results suggest that feed supplementation with SP inhibits fat deposition in broilers by reducing feed and caloric intake, but not via direct regulation on hepatic fat synthesis or adipocytic fat deposition. Alteration in the relative populations of the gut microflora suggests that SP may have gut health implications.
Collapse
Affiliation(s)
- Haifang Li
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Liqin Zhao
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Shuang Liu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, China
| | - Zhihao Zhang
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Xiaojuan Wang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, China
| | - Hai Lin
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, China.
| |
Collapse
|
24
|
He YJ, You CG. The Potential Role of Gut Microbiota in the Prevention and Treatment of Lipid Metabolism Disorders. Int J Endocrinol 2020; 2020:8601796. [PMID: 33005189 PMCID: PMC7509545 DOI: 10.1155/2020/8601796] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/24/2020] [Accepted: 09/03/2020] [Indexed: 12/17/2022] Open
Abstract
Due to changes in lifestyle, diet structure, and aging worldwide, the incidence of metabolic syndromes such as hyperlipidemia, hypertension, diabetes, and obesity is increasing. Metabolic syndrome is considered to be closely related to cardiovascular disease and severely affects human health. In recent years, researchers have revealed that the gut microbiota, through its own or interacting metabolites, has a positive role in regulating metabolic syndrome. Therefore, the gut microbiota has been a new "organ" for the treatment of metabolic syndrome. The role has not been clarified, and more research is necessary to prove the specific role of specific strains. Probiotics are also believed to regulate metabolic syndromes by regulating the gut microbiota and are expected to become a new preparation for treating metabolic syndromes. This review focuses on the regulation of lipid metabolism disorders by the gut microbiota through the effects of bile acids (BA), short-chain fatty acids (SCFAs), bile salt hydrolase (BSH), and genes such as ABCG5 and ABCG8, FXR, NPC1L, and LDL-R.
Collapse
Affiliation(s)
- Yan-Jun He
- Laboratory Medicine Center, Lanzhou University Second Hospital, No. 82 Cuiyingmen Lanzhou, Lanzhou 730030, Gansu, China
| | - Chong-Ge You
- Laboratory Medicine Center, Lanzhou University Second Hospital, No. 82 Cuiyingmen Lanzhou, Lanzhou 730030, Gansu, China
| |
Collapse
|
25
|
Trefely S, Lovell CD, Snyder NW, Wellen KE. Compartmentalised acyl-CoA metabolism and roles in chromatin regulation. Mol Metab 2020; 38:100941. [PMID: 32199817 PMCID: PMC7300382 DOI: 10.1016/j.molmet.2020.01.005] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/03/2020] [Accepted: 01/07/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Many metabolites serve as important signalling molecules to adjust cellular activities and functions based on nutrient availability. Links between acetyl-CoA metabolism, histone lysine acetylation, and gene expression have been documented and studied over the past decade. In recent years, several additional acyl modifications to histone lysine residues have been identified, which depend on acyl-coenzyme A thioesters (acyl-CoAs) as acyl donors. Acyl-CoAs are intermediates of multiple distinct metabolic pathways, and substantial evidence has emerged that histone acylation is metabolically sensitive. Nevertheless, the metabolic sources of acyl-CoAs used for chromatin modification in most cases remain poorly understood. Elucidating how these diverse chemical modifications are coupled to and regulated by cellular metabolism is important in deciphering their functional significance. SCOPE OF REVIEW In this article, we review the metabolic pathways that produce acyl-CoAs, as well as emerging evidence for functional roles of diverse acyl-CoAs in chromatin regulation. Because acetyl-CoA has been extensively reviewed elsewhere, we will focus on four other acyl-CoA metabolites integral to major metabolic pathways that are also known to modify histones: succinyl-CoA, propionyl-CoA, crotonoyl-CoA, and butyryl-CoA. We also briefly mention several other acyl-CoA species, which present opportunities for further research; malonyl-CoA, glutaryl-CoA, 3-hydroxybutyryl-CoA, 2-hydroxyisobutyryl-CoA, and lactyl-CoA. Each acyl-CoA species has distinct roles in metabolism, indicating the potential to report shifts in the metabolic status of the cell. For each metabolite, we consider the metabolic pathways in which it participates and the nutrient sources from which it is derived, the compartmentalisation of its metabolism, and the factors reported to influence its abundance and potential nuclear availability. We also highlight reported biological functions of these metabolically-linked acylation marks. Finally, we aim to illuminate key questions in acyl-CoA metabolism as they relate to the control of chromatin modification. MAJOR CONCLUSIONS A majority of acyl-CoA species are annotated to mitochondrial metabolic processes. Since acyl-CoAs are not known to be directly transported across mitochondrial membranes, they must be synthesized outside of mitochondria and potentially within the nucleus to participate in chromatin regulation. Thus, subcellular metabolic compartmentalisation likely plays a key role in the regulation of histone acylation. Metabolite tracing in combination with targeting of relevant enzymes and transporters will help to map the metabolic pathways that connect acyl-CoA metabolism to chromatin modification. The specific function of each acyl-CoA may be determined in part by biochemical properties that affect its propensity for enzymatic versus non-enzymatic protein modification, as well as the various enzymes that can add, remove and bind each modification. Further, competitive and inhibitory effects of different acyl-CoA species on these enzymes make determining the relative abundance of acyl-CoA species in specific contexts important to understand the regulation of chromatin acylation. An improved and more nuanced understanding of metabolic regulation of chromatin and its roles in physiological and disease-related processes will emerge as these questions are answered.
Collapse
Affiliation(s)
- Sophie Trefely
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Metabolic Disease Research, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Claudia D Lovell
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nathaniel W Snyder
- Center for Metabolic Disease Research, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
26
|
Su CW, Chen CY, Jiao L, Long SR, Mao T, Ji Q, O'Donnell S, Stanton C, Zheng S, Walker WA, Cherayil BJ, Shi HN. Helminth-Induced and Th2-Dependent Alterations of the Gut Microbiota Attenuate Obesity Caused by High-Fat Diet. Cell Mol Gastroenterol Hepatol 2020; 10:763-778. [PMID: 32629118 PMCID: PMC7498948 DOI: 10.1016/j.jcmgh.2020.06.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Epidemiological and animal studies have indicated an inverse correlation between the rising prevalence of obesity and metabolic syndrome and exposure to helminths. Whether helminth-induced immune response contributes to microbiota remodeling in obesity remains unknown. The aim of this study is to explore the immune-regulatory role of helminth in the prevention of HFD-induced obesity through remodeling gut microbiome. METHODS C57BL/6J WT and STAT6-/- mice were infected with Heligmosomoides polygyrus and followed by high fat diet (HFD) feeding for 6 weeks. The host immune response, body weight, and fecal microbiota composition were analyzed. We used adoptive transfer of M2 macrophages and microbiota transplantation approaches to determine the impact of these factors on HFD-obesity. We also examined stool microbiota composition and short chain fatty acids (SCFAs) concentration and determined the expression of SCFA-relevant receptors in the recipient mice. RESULTS Helminth infection of STAT6-/- (Th2-deficient) mice and adoptive transfer of helminth-induced alternatively activated (M2) macrophages demonstrated that the helminth-associated Th2 immune response plays an important role in the protection against obesity and induces changes in microbiota composition. Microbiota transplantation showed that helminth-induced, Th2-dependent alterations of the gut microbiota are sufficient to confer protection against obesity. Collectively, these results indicate that helminth infection protects against HFD-induced obesity by Th2-dependent, M2 macrophage-mediated alterations of the intestinal microbiota. CONCLUSION Our findings provide new mechanistic insights into the complex interplay between helminth infection, the immune system and the gut microbiota in a HFD-induced obesity model and holds promise for gut microbiome-targeted immunotherapy in obesity prevention.
Collapse
Affiliation(s)
- Chien Wen Su
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Chih-Yu Chen
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Lefei Jiao
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Shao Rong Long
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Tangyou Mao
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Qiaorong Ji
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Shane O'Donnell
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Shasha Zheng
- Department of Nutrition, California Baptist University, Riverside, California
| | - W Allan Walker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Bobby J Cherayil
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Hai Ning Shi
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts.
| |
Collapse
|
27
|
Chen D, Qiu YB, Gao ZQ, Wu YX, Wan BB, Liu G, Chen JL, Zhou Q, Yu RQ, Pang QF. Sodium Propionate Attenuates the Lipopolysaccharide-Induced Epithelial-Mesenchymal Transition via the PI3K/Akt/mTOR Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:6554-6563. [PMID: 32452677 DOI: 10.1021/acs.jafc.0c01302] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Short-chain fatty acids (SCFAs), especially propionate, originate from the fermentation of dietary fiber in the gut and play a key role in inhibiting pulmonary inflammation. Chronic inflammation may induce an epithelial-mesenchymal transition (EMT) in alveolar epithelial cells and result in fibrotic disorders. This study was designed to investigate the beneficial effect of sodium propionate (SP) on lipopolysaccharide (LPS)-induced EMT. In cultured BEAS-2B cells, the protein expression levels of E-cadherin, α-smooth muscle actin (SMA), and vimentin were 0.66 ± 0.20, 1.44 ± 0.23, and 1.32 ± 0.21 in the LPS group vs 1.11 ± 0.36 (P < 0.05), 1.04 ± 0.30 (P < 0.05), and 0.96 ± 0.13 (P < 0.01) in the LPS + SP group (mean ± standard deviation), respectively. Meanwhile, LPS-triggered inflammatory cytokines and extracellular proteins were also reduced by SP administration in BEAS-2B cells. Moreover, SP treatment attenuated inflammation, EMT, extracellular matrix (ECM) deposition, and even fibrosis in a mouse EMT model. In terms of mechanism, LPS-treated BEAS-2B cells exhibited a higher level of phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) phosphorylation, which was interrupted by SP treatment. It is worth noting that the blockade of the PI3K/Akt/mTOR signaling cascade reduced the LPS-evoked EMT process in BEAS-2B cells. These results suggest that SP can block LPS-induced EMT via inhibition of the PI3K/Akt/mTOR signaling cascade, which provides a basis for possible clinical use of SP in airway and lung diseases.
Collapse
Affiliation(s)
- Dan Chen
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Yu-Bao Qiu
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Zhi-Qi Gao
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Ya-Xian Wu
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Bin-Bin Wan
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Gang Liu
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Jun-Liang Chen
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Qin Zhou
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Ren-Qiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Qing-Feng Pang
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| |
Collapse
|
28
|
Liu Y, Yang Y, Lei Y, Yang L, Zhang X, Yuan J, Lei Z. Effects of dihydroartemisinin on the gut microbiome of mice. Mol Med Rep 2020; 22:707-714. [PMID: 32468008 PMCID: PMC7339414 DOI: 10.3892/mmr.2020.11165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
Dihydroartemisinin (DHA) is a semisynthetic derivative of artemisinin, which has been found to exhibit a broad range of biological activities, excluding antimalarial effects; however its effects on the gut microbiota remain poorly understood. The present study aimed to investigate the effects of DHA on the gut microbiome in mice and to determine its potential biological and pharmaceutical activities through its alteration of the gut microbiota. Serum glucose, triglyceride (TG), total cholesterol, lipopolysaccharide, high density lipoprotein‑cholesterol, low density lipoprotein‑cholesterol, alanine aminotransferase and aspartate aminotransferase levels in mice treated with DHA were analyzed using the corresponding detection kits. In addition, hematoxylin and eosin staining was performed to determine the pathological effects of DHA on the liver, kidney and intestinal tissues of mice, and the effects of DHA on the gut microbiome were analyzed using 16S ribosomal (r)DNA gene analysis. The results demonstrated that the TG serum levels of mice treated with DHA were significantly decreased compared with the control group. Furthermore, 16S rDNA gene analysis demonstrated that the bacterial diversity of mice treated with DHA was enriched compared with the control group. The DHA group exhibited increased numbers of Firmicutes and Saccharibacteria, and decreased Deferribacteres and Actinobacteria compared with the control group at the phylum level. Kyoto Encyclopedia of Genes and Genomes signaling pathway enrichment analysis also revealed that the signaling pathways associated with 'Energy metabolism' and 'Nucleotide metabolism' were upregulated, whereas the signaling pathways associated with 'Infectious diseases and 'Neurodegenerative diseases' were downregulated in the DHA group compared with the control group. In conclusion, the findings of the present study indicated that DHA may significantly decrease the serum TG levels and alter the gut microbiota, which suggested its potential to be used for the treatment of hyperlipidemia, inflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Yanyan Liu
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Yanhong Yang
- The First Affiliated Hospital (School of Clinical Medicine), Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, P.R. China
| | - Yuting Lei
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Lanxiang Yang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Xueying Zhang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Jian Yuan
- Department of Pathology and Guangdong Key Laboratory for Bioactive Drugs Research, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Zili Lei
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
29
|
Administration of Exogenous Melatonin Improves the Diurnal Rhythms of the Gut Microbiota in Mice Fed a High-Fat Diet. mSystems 2020; 5:5/3/e00002-20. [PMID: 32430404 PMCID: PMC7253360 DOI: 10.1128/msystems.00002-20] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota is strongly shaped by a high-fat diet, and obese humans and animals are characterized by low gut microbial diversity and impaired gut microbiota compositions. Comprehensive data on mammalian gut metagenomes shows gut microbiota exhibit circadian rhythms, which is disturbed by a high-fat diet. On the other hand, melatonin is a natural and ubiquitous molecule showing multiple mechanisms of regulating the circadian clock and lipid metabolism, while the role of melatonin in the regulation of the diurnal patterns of gut microbial structure and function in obese animals is not yet known. This study delineates an intricate picture of melatonin-gut microbiota circadian rhythms and may provide insight for obesity intervention. Melatonin, a circadian hormone, has been reported to improve host lipid metabolism by reprogramming the gut microbiota, which also exhibits rhythmicity in a light/dark cycle. However, the effect of the administration of exogenous melatonin on the diurnal variation in the gut microbiota in mice fed a high-fat diet (HFD) is unclear. Here, we further confirmed the antiobesogenic effect of melatonin on mice fed an HFD for 2 weeks. Samples were collected every 4 h within a 24-h period, and diurnal rhythms of clock gene expression (Clock, Cry1, Cry2, Per1, and Per2) and serum lipid indexes varied with diurnal time. Notably, Clock and triglycerides (TG) showed a marked rhythm in the control in melatonin-treated mice but not in the HFD-fed mice. The rhythmicity of these parameters was similar between the control and melatonin-treated HFD-fed mice compared with that in the HFD group, indicating an improvement caused by melatonin in the diurnal clock of host metabolism in HFD-fed mice. Moreover, 16S rRNA gene sequencing showed that most microbes exhibited daily rhythmicity, and the trends were different for different groups and at different time points. We also identified several specific microbes that correlated with the circadian clock genes and serum lipid indexes, which might indicate the potential mechanism of action of melatonin in HFD-fed mice. In addition, effects of melatonin exposure during daytime or nighttime were compared, but a nonsignificant difference was noticed in response to HFD-induced lipid dysmetabolism. Interestingly, the responses of microbiota-transplanted mice to HFD feeding also varied at different transplantation times (8:00 and 16:00) and with different microbiota donors. In summary, the daily oscillations in the expression of circadian clock genes, serum lipid indexes, and the gut microbiota appeared to be driven by short-term feeding of an HFD, while administration of exogenous melatonin improved the composition and diurnal rhythmicity of some specific gut microbiota in HFD-fed mice. IMPORTANCE The gut microbiota is strongly shaped by a high-fat diet, and obese humans and animals are characterized by low gut microbial diversity and impaired gut microbiota compositions. Comprehensive data on mammalian gut metagenomes shows gut microbiota exhibit circadian rhythms, which is disturbed by a high-fat diet. On the other hand, melatonin is a natural and ubiquitous molecule showing multiple mechanisms of regulating the circadian clock and lipid metabolism, while the role of melatonin in the regulation of the diurnal patterns of gut microbial structure and function in obese animals is not yet known. This study delineates an intricate picture of melatonin-gut microbiota circadian rhythms and may provide insight for obesity intervention.
Collapse
|
30
|
Adipokines and Adipose Tissue-Related Metabolites, Nuts and Cardiovascular Disease. Metabolites 2020; 10:metabo10010032. [PMID: 31940832 PMCID: PMC7022531 DOI: 10.3390/metabo10010032] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/06/2020] [Accepted: 01/10/2020] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue is a complex structure responsible for fat storage and releasing polypeptides (adipokines) and metabolites, with systemic actions including body weight balance, appetite regulation, glucose homeostasis, and blood pressure control. Signals sent from different tissues are generated and integrated in adipose tissue; thus, there is a close connection between this endocrine organ and different organs and systems such as the gut and the cardiovascular system. It is known that functional foods, especially different nuts, may be related to a net of molecular mechanisms contributing to cardiometabolic health. Despite being energy-dense foods, nut consumption has been associated with no weight gain, weight loss, and lower risk of becoming overweight or obese. Several studies have reported beneficial effects after nut consumption on glucose control, appetite suppression, metabolites related to adipose tissue and gut microbiota, and on adipokines due to their fatty acid profile, vegetable proteins, l-arginine, dietary fibers, vitamins, minerals, and phytosterols. The aim of this review is to briefly describe possible mechanisms implicated in weight homeostasis related to different nuts, as well as studies that have evaluated the effects of nut consumption on adipokines and metabolites related to adipose tissue and gut microbiota in animal models, healthy individuals, and primary and secondary cardiovascular prevention.
Collapse
|
31
|
Yu M, Li Z, Chen W, Wang G, Cui Y, Ma X. Dietary Supplementation With Citrus Extract Altered the Intestinal Microbiota and Microbial Metabolite Profiles and Enhanced the Mucosal Immune Homeostasis in Yellow-Feathered Broilers. Front Microbiol 2019; 10:2662. [PMID: 31849855 PMCID: PMC6887900 DOI: 10.3389/fmicb.2019.02662] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/01/2019] [Indexed: 01/10/2023] Open
Abstract
The present study aimed to investigate the effects of citrus extract (CE) on intestinal microbiota, microbial metabolite profiles, and the mucosal immune status in broilers. A total of 540 one-day-old yellow-feathered broilers were randomly allotted into three groups and fed a basal diet (control group), or a basal diet containing 10 mg/kg of zinc bacitracin (antibiotic group), or 10 mg/kg of CE (CE group). Each treatment consisted of six replicates, with 30 broilers per replicate. After 63-day feeding, two broilers per replicate were randomly selected and slaughtered, and their ileal and cecal digesta and ileal tissue were collected for microbial composition, microbial metabolites, and gene expression analysis. The results showed that CE significantly increased the abundance of Barnesiella and Blautia than did the antibiotic group (adjusted P < 0.05), whereas it decreased the abundance of Alistipes and Bacteroides (adjusted P < 0.05). Meanwhile, the CE group also increased the numbers of Bifidobacterium and Lactobacillus than did the control and antibiotic groups (P < 0.05), whereas it decreased the number of Escherichia coli (P < 0.05). For microbial metabolites, dietary supplementation with CE increased the concentrations of lactate, total short-chain fatty acids, acetate, and butyrate in the cecum than did the control and antibiotic groups (P < 0.05), whereas it decreased the concentrations of amino acid fermentation products (ammonia, amines, p-cresol, and indole) (P < 0.05). Additionally, supplementation with CE up-regulated (P < 0.05) the mRNA expression of intestinal barrier genes (ZO-1 and Claudin) in the ileum than did both the control and antibiotic groups. However, antibiotic treatment induced gut microbiota dysbiosis, altered the microbial metabolism, and disturbed the innate immune homeostasis. In summary, these results provide evidence that dietary supplementation with CE can improve the intestinal barrier function by changing microbial composition and metabolites, likely toward a host-friendly gut environment. This suggests that CE may possibly act as an efficient antibiotic alternative for yellow-feathered broiler production.
Collapse
Affiliation(s)
- Miao Yu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China.,Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou, China.,Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China.,Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China
| | - Zhenming Li
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China.,Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou, China.,Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China.,Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China
| | - Weidong Chen
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China.,Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou, China.,Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China.,Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China
| | - Gang Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China.,Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou, China.,Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China.,Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China
| | - Yiyan Cui
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China.,Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou, China.,Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China.,Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China
| | - Xianyong Ma
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China.,Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou, China.,Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China.,Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China
| |
Collapse
|