1
|
Wang L, Zhang Y, Zang X, Yang Y, Wang W, Zhang J, Que Y, Liang F, Wang T, Zhang J, Ma H, Guan L. Physicochemical properties and fermentation characteristics of a novel polysaccharide degraded from Flammulina velutipes residues polysaccharide. Food Chem X 2024; 24:102049. [PMID: 39717408 PMCID: PMC11665304 DOI: 10.1016/j.fochx.2024.102049] [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: 09/30/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/25/2024] Open
Abstract
Flammulina velutipes (F. velutipes) residues polysaccharide (FVRP) is a high molecular weight polysaccharide with diverse bioactivities extracted from F. velutipes residues (FVR). However, high molecular weight polysaccharides have been shown to face significant challenges in crossing the cell membrane barrier, thereby limiting their absorption and application in the body. Therefore, an ultrasonic-assisted H2O2-Fe3+ method was employed for the first time to degrade FVRP, resulting in the production of a new polysaccharide, FVRPF. Compared with FVRP, there was no significant difference in the main chemical structure of FVRPF, but the monosaccharide composition ratio varied. and FVRPF had lower molecular weight and stronger antioxidant capacity. Moreover, FVRPF could be degraded by human microbiota, modulate gut microbiota composition, and increase the production of total short-chain fatty acids (SCFAs). These findings suggest that FVRPF holds potential as a promising prebiotic for applications in the food and pharmaceutical industries.
Collapse
Affiliation(s)
- Liping Wang
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, Jilin, PR China
| | - Yao Zhang
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, Jilin, PR China
| | - Xinyuan Zang
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, Jilin, PR China
| | - Yiting Yang
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, Jilin, PR China
| | - Wanting Wang
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, Jilin, PR China
| | - Jingbo Zhang
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, Jilin, PR China
| | - Yunxiang Que
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, Jilin, PR China
| | - Fengxiang Liang
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, Jilin, PR China
| | - Tiezhu Wang
- Changchun Gaorong Biotechnological Co., Ltd., Changchun 130102, Jilin, PR China
| | - Jian Zhang
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, Jilin, PR China
- Institute for Safflower Industry Research of Shihezi University, Pharmacy College of Shihezi University, Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi 832003, Xinjiang, PR China
| | - Hongxia Ma
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, Jilin, PR China
| | - Lili Guan
- College of Life Sciences, Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, Jilin, PR China
| |
Collapse
|
2
|
Cheng H, Xu L, Zhu H, Bu T, Li Z, Zhao S, Yang K, Sun P, Cai M. Structural characterization of oligosaccharide from Dendrobium officinale and its properties in vitro digestion and fecal fermentation. Food Chem 2024; 460:140511. [PMID: 39047478 DOI: 10.1016/j.foodchem.2024.140511] [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: 03/15/2024] [Revised: 05/16/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Oligosaccharides from Dendrobium officinale (DOO) is a kind of new potential prebiotic for health. In this study, structural characteristics, digestion properties and regulatory function on intestinal flora of DOO were investigated. An oligosaccharide, DOO 1-1, was purified by DEAE-Sepharose Fast Flow and Sephadex G-25, and its physicochemical properties were characterized as a glucomannan oligosaccharide with a molecular weight of 1560 Da (DP = 9). In vitro simulated digestion, it proved that the structure of DOO 1-1 was degraded hardly in the simulated gastric and small intestinal fluid. By evaluating the gas, short-chain fatty acids and intestinal microbiota in vitro fermentation, DOO has an excellent regulatory effect on intestinal microbiota, especially promoting the proliferation of Bacteroidetes and Actinobacteria. Therefore, DOO can be used as a potential prebiotic in functional foods.
Collapse
Affiliation(s)
- Hao Cheng
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, People's Republic of China; Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Hangzhou, Zhejiang 310014, People's Republic of China
| | - Lei Xu
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, People's Republic of China; Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Hangzhou, Zhejiang 310014, People's Republic of China
| | - Hua Zhu
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, People's Republic of China; Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Hangzhou, Zhejiang 310014, People's Republic of China
| | - Tingting Bu
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, People's Republic of China; Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Hangzhou, Zhejiang 310014, People's Republic of China
| | - Zhenhao Li
- Longevity Valley Botanical Co., Ltd., Zhejiang 321200, People's Republic of China
| | - Shuna Zhao
- School of Food and Health, Beijing Technology and Business University, Beijing, 100048, People's Republic of China
| | - Kai Yang
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, People's Republic of China; Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Hangzhou, Zhejiang 310014, People's Republic of China
| | - Peilong Sun
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, People's Republic of China; Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Hangzhou, Zhejiang 310014, People's Republic of China
| | - Ming Cai
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, People's Republic of China; Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Hangzhou, Zhejiang 310014, People's Republic of China.
| |
Collapse
|
3
|
Liu H, Lu H, Wang Y, Yu C, He Z, Dong H. Unlocking the power of short-chain fatty acids in ameliorating intestinal mucosal immunity: a new porcine nutritional approach. Front Cell Infect Microbiol 2024; 14:1449030. [PMID: 39286812 PMCID: PMC11402818 DOI: 10.3389/fcimb.2024.1449030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Short-chain fatty acids (SCFAs), a subset of organic fatty acids with carbon chains ranging from one to six atoms in length, encompass acetate, propionate, and butyrate. These compounds are the endproducts of dietary fiber fermentation, primarily catalyzed by the glycolysis and pentose phosphate pathways within the gut microbiota. SCFAs act as pivotal energy substrates and signaling molecules in the realm of animal nutrition, exerting a profound influence on the intestinal, immune system, and intestinal barrier functions. Specifically, they contibute to 60-70% of the total energy requirements in ruminants and 10-25% in monogastric animals. SCFAs have demonstrated the capability to effectively modulate intestinal pH, optimize the absorption of mineral elements, and impede pathogen invasion. Moreover, they enhance the expression of proteins associated with intestinal tight junctions and stimulate mucus production, thereby refining intestinal tissue morphology and preserving the integrity of the intestinal structure. Notably, SCFAs also exert anti-inflammatory properties, mitigating inflammation within the intestinal epithelium and strengthening the intestinal barrier's defensive capabilities. The present review endeavors to synthesize recent findings regarding the role of SCFAs as crucial signaling intermediaries between the metabolic activities of gut microbiota and the status of porcine cells. It also provides a comprehensive overview of the current literature on SCFAs' impact on immune responses within the porcine intestinal mucosa.
Collapse
Affiliation(s)
- Haoyang Liu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
- Beijing Engineering Research Center of Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Hongde Lu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
- Beijing Engineering Research Center of Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Yuxuan Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
- Beijing Engineering Research Center of Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Chenyun Yu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
- Beijing Engineering Research Center of Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Zhiyuan He
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Hong Dong
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
- Beijing Engineering Research Center of Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| |
Collapse
|
4
|
Zhang XL, An ZY, Lu GJ, Zhang T, Liu CW, Liu MQ, Wei QX, Quan LH, Kang JD. MCT1-mediated transport of valeric acid promotes porcine preimplantation embryo development by improving mitochondrial function and inhibiting the autophagic AMPK-ULK1 pathway. Theriogenology 2024; 225:152-161. [PMID: 38805997 DOI: 10.1016/j.theriogenology.2024.05.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
Oocytes and embryos are highly sensitive to environmental stress in vivo and in vitro. During in vitro culture, many stressful conditions can affect embryo quality and viability, leading to adverse clinical outcomes such as abortion and congenital abnormalities. In this study, we found that valeric acid (VA) increased the mitochondrial membrane potential and ATP content, decreased the level of reactive oxygen species that the mitochondria generate, and thus improved mitochondrial function during early embryonic development in pigs. VA decreased expression of the autophagy-related factors LC3B and BECLIN1. Interestingly, VA inhibited expression of autophagy-associated phosphorylation-adenosine monophosphate-activated protein kinase (p-AMPK), phosphorylation-UNC-51-like autophagy-activated kinase 1 (p-ULK1, Ser555), and ATG13, which reduced apoptosis. Short-chain fatty acids (SCFAs) can signal through G-protein-coupled receptors on the cell membrane or enter the cell directly through transporters. We further show that the monocarboxylate transporter 1 (MCT1) was necessary for the effects of VA on embryo quality, which provides a new molecular perspective of the pathway by which SCFAs affect embryos. Importantly, VA significantly inhibited the AMPK-ULK1 autophagic signaling pathway through MCT1, decreased apoptosis, increased expression of embryonic pluripotency genes, and improved embryo quality.
Collapse
Affiliation(s)
- Xiu-Li Zhang
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133002, China.
| | - Zhi-Yong An
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133002, China.
| | - Gao-Jie Lu
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133002, China.
| | - Tuo Zhang
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133002, China.
| | - Cheng-Wei Liu
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133002, China.
| | - Meng-Qi Liu
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133002, China.
| | - Qing-Xin Wei
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133002, China.
| | - Lin-Hu Quan
- College of Pharmacy, Yanbian University, Yanji, 133002, China.
| | - Jin-Dan Kang
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133002, China; Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanji, 133002, China.
| |
Collapse
|
5
|
Xi L, Weibing X, Shuyong F, Sheng-Hua L, Xiong F, Chin-Ping T, Ping-Ping W, Zu-Man D, Chun C. The effect of the molecular weight of blackberry polysaccharides on gut microbiota modulation and hypoglycemic effect in vivo. Food Funct 2024; 15:8586-8603. [PMID: 39078268 DOI: 10.1039/d4fo01989j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Blackberry polysaccharides with certain molecular weight distribution have good bioactivity. In this research, type 2 diabetes mice were used to investigate the hypoglycemic effect of blackberry polysaccharides with three different molecular weights, BBP (603.59 kDa), BBP-8 (408.13 kDa) and BBP-24 (247.62 kDa), through gut microbiota modulation. Blackberry polysaccharides exhibited stronger hypoglycemic activity after degradation, and the FBG of BBP, BBP-8 and BBP-24 was reduced to 20.21 ± 4.17 mmol L-1, 20.6 ± 7.23 mmol L-1 and 17.32 ± 6.59 mmol L-1 and OGTT-AUC was reduced by 14.76%, 19.80% and 25.04%, respectively, after 8-week intervention. Furthermore, 16S rRNA gene sequencing analysis indicated that BBP, BBP-8 and BBP-24 could reshape the diversity and composition of the gut microbiota. From 0 to 4 weeks, the F/B of BBP, BBP-8 and BBP-24 reduced by 56.44%, 47.19% and 62.04%, reaching 3.39, 6.54, and 3.11 in the 8th week, respectively, which suggested the faster utilization of BBP-24. Moreover, the intervention the three blackberry polysaccharides increased the relative abundance of the targeted beneficial bacteria Oscillospira and Bacteroidaceae Bacteroides and decreased the relative abundance of the pathogenic bacterium Allobaculum. In general, the result demonstrated that blackberry polysaccharides with a lower molecular weight are more easily fermented, making the theoretical basis for the development of blackberry polysaccharides as a probiotic food to rapidly regulate intestinal flora for type 2 diabetes.
Collapse
Affiliation(s)
- Lai Xi
- SCUT-Zhuhai Institute of Modern Industrial Innovation, School of Food Science and Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China.
| | - Xu Weibing
- Guangzhou Restaurant Group Likofu Food Company Ltd, Guangzhou 510640, China
| | - Fu Shuyong
- Guangzhou Restaurant Group Likofu Food Company Ltd, Guangzhou 510640, China
| | - Li Sheng-Hua
- Guangzhou Restaurant Group Likofu Food Company Ltd, Guangzhou 510640, China
| | - Fu Xiong
- SCUT-Zhuhai Institute of Modern Industrial Innovation, School of Food Science and Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China.
- 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
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou 510640, China
| | - Tan Chin-Ping
- Univ Putra Malaysia, Fac Food Sci & Technol, Dept Food Technol, Serdang 43400, Selangor, Malaysia
| | - Wang Ping-Ping
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Dou Zu-Man
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Chen Chun
- SCUT-Zhuhai Institute of Modern Industrial Innovation, School of Food Science and Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China.
- 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
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou 510640, China
| |
Collapse
|
6
|
Li M, Zhang X, Gao Z, Wu M, Ren T, Wu C, Wang J, Geng Y, Lv W, Zhou Q, Zhao W. Metabolomic insights into the profile, bioaccessibility, and transepithelial transport of polyphenols from germinated quinoa during in vitro gastrointestinal digestion/Caco-2 cell transport, and their prebiotic effects during colonic fermentation. Food Res Int 2024; 186:114339. [PMID: 38729694 DOI: 10.1016/j.foodres.2024.114339] [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: 11/24/2023] [Revised: 03/30/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024]
Abstract
The health-promoting activities of polyphenols and their metabolites originating from germinated quinoa (GQ) are closely related to their digestive behavior, absorption, and colonic fermentation; however, limited knowledge regarding these properties hinder further development. The aim of this study was to provide metabolomic insights into the profile, bioaccessibility, and transepithelial transport of polyphenols from germinated quinoa during in vitro gastrointestinal digestion and Caco-2 cell transport, whilst also investigating the changes in the major polyphenol metabolites and the effects of prebiotics during colonic fermentation. It was found that germination treatment increased the polyphenol content of quinoa by 21.91%. Compared with RQ group, 23 phenolic differential metabolites were upregulated and 47 phenolic differential metabolites were downregulated in GQ group. Compared with RQ group after simulated digestion, 7 kinds of phenolic differential metabolites were upregulated and 17 kinds of phenolic differential metabolites were downregulated in GQ group. Compared with RQ group after cell transport, 7 kinds of phenolic differential metabolites were upregulated and 9 kinds of phenolic differential metabolites were downregulated in GQ group. In addition, GQ improved the bioaccessibilities and transport rates of various polyphenol metabolites. During colonic fermentation, GQ group can also increase the content of SCFAs, reduce pH value, and adjust gut microbial populations by increasing the abundance of Actinobacteria, Bacteroidetes, Verrucomicrobiota, and Spirochaeota at the phylum level, as well as Bifidobacterium, Megamonas, Bifidobacterium, Brevundimonas, and Bacteroides at the genus level. Furthermore, the GQ have significantly inhibited the activity of α-amylase and α-glucosidase. Based on these results, it was possible to elucidate the underlying mechanisms of polyphenol metabolism in GQ and highlight its beneficial effects on the gut microbiota.
Collapse
Affiliation(s)
- Meijiao Li
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, PR China
| | - Xuan Zhang
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, PR China
| | - Zhe Gao
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, PR China
| | - Mengying Wu
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, PR China
| | - Ting Ren
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, PR China
| | - Chen Wu
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, PR China
| | - Jie Wang
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, PR China
| | - Yanlou Geng
- National Semi-arid Agricultural Engineering Technology Research Center, Shijiazhuang 050011, PR China
| | - Wei Lv
- National Semi-arid Agricultural Engineering Technology Research Center, Shijiazhuang 050011, PR China
| | - Qian Zhou
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, PR China.
| | - Wen Zhao
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, PR China.
| |
Collapse
|
7
|
Wei S, Wang L, Chen X, Wang Y, Tong L, Han Q, Ren B, Guo D. Anti-inflammatory activity of Boletus aereus polysaccharides: Involvement of digestion and gut microbiota fermentation. Food Chem X 2024; 21:101052. [PMID: 38187943 PMCID: PMC10770587 DOI: 10.1016/j.fochx.2023.101052] [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: 09/05/2023] [Revised: 11/24/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Boletus aereus, an edible mushroom, has gained popularity as a medicinal and functional food. This study aimed to investigate the digestive characteristics of B. aereus polysaccharide (BAP) and its effects on gut microbiota. In vitro digestion results indicated partial degradation of BAP. Furthermore, the digested BAP displayed significantly enhanced antioxidant ability. The 16S rRNA sequencing data revealed that BAP positively influenced the abundance of Phascolarctobacterium, Prevotella, and Bifidobacterium in the gut microbiota. Additionally, BAP promoted the production of short-chain fatty acids (SCFAs). Metabolites of BAP utilized by the gut microbiota effectively reduced the concentration of TNF-α, IL-1β, and NO in an LPS-stimulated RAW 264.7 cell inflammation model. Mantel tests demonstrated a strong correlation among fermentation indicators, gut microbiome composition, SCFAs, and inflammatory cytokines. Overall, this research revealed the underlying digestive and fermentation mechanisms of BAP and provided new insights into the usage of edible mushroom polysaccharides in functional food.
Collapse
Affiliation(s)
- Shixiang Wei
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University Nanjing 210023, China
| | - Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Xiaodie Chen
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University Nanjing 210023, China
| | - Yue Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University Nanjing 210023, China
| | - Lingling Tong
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University Nanjing 210023, China
| | - Qianyun Han
- BIOSYST-MeBioS, Faculty of Bioscience Engineering, KU Leuven, Leuven 3000, Belgium
- College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Beijing 100083, China
| | - Bo Ren
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University Nanjing 210023, China
| | - Dongsheng Guo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University Nanjing 210023, China
| |
Collapse
|
8
|
Vagnerová K, Hudcovic T, Vodička M, Ergang P, Klusoňová P, Petr Hermanová P, Šrůtková D, Pácha J. The effect of oral butyrate on colonic short-chain fatty acid transporters and receptors depends on microbial status. Front Pharmacol 2024; 15:1341333. [PMID: 38595917 PMCID: PMC11002167 DOI: 10.3389/fphar.2024.1341333] [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: 11/20/2023] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
Butyrate, a metabolite produced by gut bacteria, has demonstrated beneficial effects in the colon and has been used to treat inflammatory bowel diseases. However, the mechanism by which butyrate operates remains incompletely understood. Given that oral butyrate can exert either a direct impact on the gut mucosa or an indirect influence through its interaction with the gut microbiome, this study aimed to investigate three key aspects: (1) whether oral intake of butyrate modulates the expression of genes encoding short-chain fatty acid (SCFA) transporters (Slc16a1, Slc16a3, Slc16a4, Slc5a8, Abcg2) and receptors (Hcar2, Ffar2, Ffar3, Olfr78, Olfr558) in the colon, (2) the potential involvement of gut microbiota in this modulation, and (3) the impact of oral butyrate on the expression of colonic SCFA transporters and receptors during colonic inflammation. Specific pathogen-free (SPF) and germ-free (GF) mice with or without DSS-induced inflammation were provided with either water or a 0.5% sodium butyrate solution. The findings revealed that butyrate decreased the expression of Slc16a1, Slc5a8, and Hcar2 in SPF but not in GF mice, while it increased the expression of Slc16a3 in GF and the efflux pump Abcg2 in both GF and SPF animals. Moreover, the presence of microbiota was associated with the upregulation of Hcar2, Ffar2, and Ffar3 expression and the downregulation of Slc16a3. Interestingly, the challenge with DSS did not alter the expression of SCFA transporters, regardless of the presence or absence of microbiota, and the effect of butyrate on the transporter expression in SPF mice remained unaffected by DSS. The expression of SCFA receptors was only partially affected by DSS. Our results indicate that (1) consuming a relatively low concentration of butyrate can influence the expression of colonic SCFA transporters and receptors, with their expression being modulated by the gut microbiota, (2) the effect of butyrate does not appear to result from direct substrate-induced regulation but rather reflects an indirect effect associated with the gut microbiome, and (3) acute colon inflammation does not lead to significant changes in the transcriptional regulation of most SCFA transporters and receptors, with the effect of butyrate in the inflamed colon remaining intact.
Collapse
Affiliation(s)
- Karla Vagnerová
- Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| | - Tomáš Hudcovic
- Institute of Microbiology, Czech Academy of Sciences, Nový Hrádek, Czechia
| | - Martin Vodička
- Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| | - Peter Ergang
- Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| | - Petra Klusoňová
- Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| | | | - Dagmar Šrůtková
- Institute of Microbiology, Czech Academy of Sciences, Nový Hrádek, Czechia
| | - Jiří Pácha
- Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| |
Collapse
|
9
|
Tsugami Y, Suzuki N, Nii T, Isobe N. Effect of sodium butyrate treatment at the basolateral membranes on the tight junction barrier function via a monocarboxylate transporter in goat mammary epithelial cells. Exp Cell Res 2024; 436:113944. [PMID: 38296017 DOI: 10.1016/j.yexcr.2024.113944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 02/09/2024]
Abstract
In lactating mammary glands, tight junctions (TJs) prevent blood from mixing with milk and maintain epithelial cell polarity, which is important for milk production. This study aimed to investigate the effect of sodium acetate and sodium butyrate (SB) stimulation direction on the TJ barrier function, which is measured with regard to transepithelial electrical resistance and fluorescein flux, in goat mammary epithelial cells. The expression and localization of the TJ proteins claudin-3 and claudin-4 were examined using Western blotting and immunofluorescence. SB treatment in the lower chamber of cell culture inserts adversely affected the TJ barrier function, whereas sodium acetate barely had any effect, regardless of stimulation direction. In addition, SB treatment in the lower chamber significantly upregulated claudin-3 and claudin-4, whereas TJ proteins showed intermittent localization. Moreover, SB induced endoplasmic reticulum (ER) stress. ARC155858, a monocarboxylate transporter-1 inhibitor, alleviated the adverse impact of SB on TJs and the associated ER stress. Interestingly, sodium β-hydroxybutyrate, a butyrate metabolite, did not affect the TJ barrier function. Our findings indicate that sodium acetate and SB influence the TJ barrier function differently, and excessive cellular uptake of SB can disrupt TJs and induce ER stress.
Collapse
Affiliation(s)
- Yusaku Tsugami
- National Institute of Animal Health, National Agriculture and Food Research Organization, 4 Hitsujigaoka, Toyohira, Sapporo, Hokkaido, 062-0045, Japan; Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, Hiroshima, 739-8528, Japan.
| | - Naoki Suzuki
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, Hiroshima, 739-8528, Japan.
| | - Takahiro Nii
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, Hiroshima, 739-8528, Japan.
| | - Naoki Isobe
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, Hiroshima, 739-8528, Japan.
| |
Collapse
|
10
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Viral Liver Disease and Intestinal Gut–Liver Axis. GASTROINTESTINAL DISORDERS 2024; 6:64-93. [DOI: 10.3390/gidisord6010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
The intestinal microbiota is closely related to liver diseases via the intestinal barrier and bile secretion to the gut. Impairment of the barrier can translocate microbes or their components to the liver where they can contribute to liver damage and fibrosis. The components of the barrier are discussed in this review along with the other elements of the so-called gut–liver axis. This bidirectional relation has been widely studied in alcoholic and non-alcoholic liver disease. However, the involvement of microbiota in the pathogenesis and treatment of viral liver diseases have not been extensively studied, and controversial data have been published. Therefore, we reviewed data regarding the integrity and function of the intestinal barrier and the changes of the intestinal microbioma that contribute to progression of Hepatitis B (HBV) and Hepatitis C (HCV) infection. Their consequences, such as cirrhosis and hepatic encephalopathy, were also discussed in connection with therapeutic interventions such as the effects of antiviral eradication and the use of probiotics that may influence the outcome of liver disease. Profound alterations of the microbioma with significant reduction in microbial diversity and changes in the abundance of both beneficial and pathogenic bacteria were found.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Department of Gastroenterology, Medical School, University of Crete, 71500 Heraklion, Greece
| | - Ioannis Tsomidis
- Department of Gastroenterology, Medical School, University of Crete, 71500 Heraklion, Greece
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece
| |
Collapse
|
11
|
Chaves Júnior JV, Ayala AP, Pontes DDL, de Souza FS, Aragão CFS. A Metformin-Ferulic Acid Salt with Improved Biopharmaceutical Parameters. J Pharm Sci 2023; 112:3120-3130. [PMID: 37451318 DOI: 10.1016/j.xphs.2023.07.009] [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: 03/06/2023] [Revised: 06/27/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
Though ferulic acid presents great hypoglycemic potential, it possesses limited aqueous solubility, and low oral bioavailability. When associated with metformin, the first-choice drug in Type 2 diabetes treatment, FA demonstrates synergistic hypoglycemic effects, however, it also causes certain undesirable dose-related effects. This study aimed to develop a new ferulic acid - metformin multicomponent system, and incorporate it into a solid dosage form with improved biopharmaceutical parameters. A novel metformin: ferulate (1:1) salt (MFS) was produced, which was properly characterized using differing analytical techniques, including single crystal analysis. Also during the course of the study, a new polymorph of the metformin free base was observed. The MFS was obtained using solvent evaporation methods, which achieved high yields in reproducible process, as well as a 740-fold increase in ferulic acid aqueous solubility. The MFS tablets developed met quality control requirements for this dosage form, as well as revealing excellent performance in vitro dissolution tests, presenting dissolution efficiency values of 95.4 ± 0.5%. Additionally, physicochemical instability was not observed in a study at 40 °C for 3 months for both MFS powder and its tablet form. The MFS product developed is a promising candidate for further Type 2 diabetes clinical study.
Collapse
Affiliation(s)
- José Venâncio Chaves Júnior
- Pharmacy Department, Federal University of Rio Grande do Norte, 59010-115, Natal, Brazil; Pharmaceutical Sciences Department, Federal University of Paraíba, 58051-970, João Pessoa, Brazil.
| | | | - Daniel de Lima Pontes
- Institute of Chemistry, Federal University of Rio Grande do Norte, 59010-115, Natal, Brazil
| | - Fábio Santos de Souza
- Pharmaceutical Sciences Department, Federal University of Paraíba, 58051-970, João Pessoa, Brazil
| | | |
Collapse
|
12
|
Sun Y, Song J, Lan X, Ma F, Jiang M, Jiang C. Calcium-Sensitive Receptors Alters Intestinal Microbiota Metabolites Especially SCFAs and Ameliorates Intestinal Barrier Damage in Neonatal Rat Endotoxemia. Infect Drug Resist 2023; 16:5707-5717. [PMID: 37667808 PMCID: PMC10475303 DOI: 10.2147/idr.s420689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/17/2023] [Indexed: 09/06/2023] Open
Abstract
Purpose The calcium-sensing receptor (CaSR) acts as a major modulator of tissue responses related to calcium homeostasis and expresses highly in the mammalian intestine. Endotoxemia tends to impair intestinal barrier function and poses significant obstacles in clinical treatment. This work is designed to decipher whether CaSR can protect lipopolysaccharide (LPS)-induced intestinal barrier dysfunction in neonatal rats by targeting intestinal metabolites. Patient and Methods In this study, we utilized gas chromatography (GC) combined with liquid chromatography-mass spectrometry (LC-MS) to quantitatively analyze SCFAs and metabolites in fecal samples of 24 neonatal rats with LPS induced endotoxemia. Results Our results showed that CaSR alleviated endotoxin damage to the intestinal tight junction structure and upregulated the levels of butyric acid, propionic acid, valeric acid, and isovaleric acid in short-chain fatty acids (SCFAs). Non-targeted metabolomics analysis indicated that CaSR improved intestinal metabolic disorders by regulating glycerophospholipid metabolism, α-linolenic acid metabolism, as well as sphingolipids metabolism. Conclusion CaSR can alter intestinal microbiota metabolites, especially SCFAs, and improve intestinal barrier damage in neonatal rat endotoxemia.
Collapse
Affiliation(s)
- Yan Sun
- Department of Neonatology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People’s Republic of China
| | - Jiayu Song
- Department of Neonatology, Zhuhai Women and Children’s Hospital, Zhuhai, Guangdong, 519060, People’s Republic of China
| | - Xue Lan
- Department of Neonatology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People’s Republic of China
| | - Fei Ma
- Department of Neonatology, Zhuhai Women and Children’s Hospital, Zhuhai, Guangdong, 519060, People’s Republic of China
| | - Mingyu Jiang
- Department of Pediatrics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People’s Republic of China
| | - Chunming Jiang
- Department of Neonatology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People’s Republic of China
- Department of Neonatology, Zhuhai Women and Children’s Hospital, Zhuhai, Guangdong, 519060, People’s Republic of China
| |
Collapse
|
13
|
Hu W, Di Q, Liang T, Zhou N, Chen H, Zeng Z, Luo Y, Shaker M. Effects of in vitro simulated digestion and fecal fermentation of polysaccharides from straw mushroom (Volvariella volvacea) on its physicochemical properties and human gut microbiota. Int J Biol Macromol 2023; 239:124188. [PMID: 36996950 DOI: 10.1016/j.ijbiomac.2023.124188] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/06/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023]
Abstract
Herein, the fermentation and digestion behavior of Volvariella volvacea polysaccharide (VVP) were examined through the in vitro simulation experiment. The results revealed that succeeding the simulated salivary gastrointestinal digestion, the molecular weight of VVP was reduced by only 8.9 %. In addition, the reducing sugar, uronic acid, monosaccharide composition and Fourier transform infrared spectroscopy characteristics of VVP did not change significantly, which indicate that saliva-gastrointestinal could not digest VVP. However, 48 h of fecal fermentation of VVP dramatically reduced its molecular weight by 40.4 %. Furthermore, the molar ratios of the monosaccharide composition altered considerably due to the degradation of VVP by microorganisms and the metabolysis into different short-chain fatty acids (SCFAs). Meanwhile, the VVP also raised the proportion of Bacteroidetes to Firmicutes and promoted the proliferation of some beneficial bacteria including Bacteroides and Phascolarctobacterium, whereas it inhibited the growth of unfavorable bacteria such as Escherichia-shigella. Therefore, VVP has the potential to have a positive influence on health and hinder diseases by improving the intestinal microbial environment. These findings provide a theoretical foundation to further develop Volvariella volvacea as a healthy functional food.
Collapse
Affiliation(s)
- Wei Hu
- Chongqing Academy of Agricultural Sciences, Chongqing 401329, China
| | - Qing Di
- Chongqing Academy of Agricultural Sciences, Chongqing 401329, China
| | - Tao Liang
- Chongqing Academy of Agricultural Sciences, Chongqing 401329, China
| | - Na Zhou
- Chongqing Academy of Agricultural Sciences, Chongqing 401329, China
| | - Hongxia Chen
- Chongqing Academy of Agricultural Sciences, Chongqing 401329, China
| | - Zhihong Zeng
- Chongqing Academy of Agricultural Sciences, Chongqing 401329, China.
| | - Yang Luo
- The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Majid Shaker
- Chongqing 2D Materials Institute, Chongqing 400714, China; Lehrstuhl für Physikalische Chemie II, Friedrich-Alexander-Universität Erlangen-Nürnberg, Egerlandstr. 3, Erlangen 91058, Germany
| |
Collapse
|
14
|
Zhang F, Xiao Y, Pan L, Yu L, Liu Y, Li D, Liu X. Effects of polysaccharides from Lyophyllum decastes (Fr.) Singer on gut microbiota via in vitro-simulated digestion and fermentation. Front Microbiol 2023; 14:1083917. [PMID: 36860492 PMCID: PMC9969080 DOI: 10.3389/fmicb.2023.1083917] [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: 10/29/2022] [Accepted: 01/09/2023] [Indexed: 02/17/2023] Open
Abstract
Introduction Lyophyllum decastes (Fr.) Singer polysaccharides (LDSPs) have been verified to possess strong biological properties. However, the effects of LDSPs on intestinal microbes and their metabolites have rarely been addressed. Methods The in vitro-simulated saliva-gastrointestinal digestion and human fecal fermentation were used to evaluate the effects of LDSPs on non-digestibility and intestinal microflora regulation in the present study. Results The results showed a slight increase in the content of the reducing end of the polysaccharide chain and no obvious change in the molecular weight during in vitro digestion. After 24 h in vitro fermentation, LDSPs were degraded and utilized by human gut microbiota, and LDSPs could be transformed into short-chain fatty acids leading to significant (p < 0.05) decrease in the pH of the fermentation solution. The digestion did not remarkably affect the overall structure of LDSPs and 16S rRNA analysis revealed distinct shifts in the gut microbial composition and community diversity of the LDSPs-treated cultures, compared with the control group. Notably, the LDSPs group directed a targeted promotion of the abundance of butyrogenic bacteria, including Blautia, Roseburia, and Bacteroides, and an increase in the n-butyrate level. Discussion These findings suggest that LDSPs might be a potential prebiotic to provide a health benefit.
Collapse
Affiliation(s)
- Fangfang Zhang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, China
| | - Ying Xiao
- School of Health and Social Care, Shanghai Urban Construction Vocational College, Shanghai, China,*Correspondence: Ying Xiao, ✉
| | - Liang Pan
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, China
| | - Ling Yu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, China,Ling Yu, ✉
| | - Yanfang Liu
- National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture and Rural Affairs, Shanghai, China,Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai, China,Yanfang Liu, ✉
| | - Deshun Li
- National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture and Rural Affairs, Shanghai, China,Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Xiaojie Liu
- School of Health and Social Care, Shanghai Urban Construction Vocational College, Shanghai, China
| |
Collapse
|
15
|
Xu M, Pan L, Wang B, Zou X, Zhang A, Zhou Z, Han Y. Simulated Digestion and Fecal Fermentation Behaviors of Levan and Its Impacts on the Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1531-1546. [PMID: 36622938 DOI: 10.1021/acs.jafc.2c06897] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Levan is a microbial fructan widely explored in various fields owing to its excellent physical and biochemical properties. However, little is known about its digestion and fermentation characteristics in vitro. This study evaluated the potential prebiotic properties of levan obtained by enzymatic synthesis. Scanning electron microscopy, Fourier transform infrared spectroscopy, and nuclear magnetic resonance spectroscopy showed that the primary structures of levan remained stable after saliva-gastrointestinal digestion. The microtopography, molecular weight, and functional group of levan were seriously damaged during fecal fermentation. Moreover, the total short-chain fatty acid levels increased significantly, especially for propionic acid, butyric acid, and valeric acid. The 16S rDNA sequencing showed that levan mainly increased the abundance of Firmicutes; in genus levels, certain beneficial bacteria such as Megasphaera and Megamonas genera were remarkably promoted, and the proliferation of harmful genera was inhibited (such as Cedecea and Klebsiella). Overall, this study provided new insights into the potential probiotic mechanism of levan.
Collapse
Affiliation(s)
- Min Xu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| | - Lei Pan
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| | - Binbin Wang
- School of Life Science, Shanxi Normal University, Taiyuan 030000, China
| | - Xuan Zou
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| | - Aihua Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| | - Zhijiang Zhou
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| | - Ye Han
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| |
Collapse
|
16
|
Clifford MN, King LJ, Kerimi A, Pereira-Caro MG, Williamson G. Metabolism of phenolics in coffee and plant-based foods by canonical pathways: an assessment of the role of fatty acid β-oxidation to generate biologically-active and -inactive intermediates. Crit Rev Food Sci Nutr 2022; 64:3326-3383. [PMID: 36226718 DOI: 10.1080/10408398.2022.2131730] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
ω-Phenyl-alkenoic acids are abundant in coffee, fruits, and vegetables. Along with ω-phenyl-alkanoic acids, they are produced from numerous dietary (poly)phenols and aromatic amino acids in vivo. This review addresses how phenyl-ring substitution and flux modulates their gut microbiota and endogenous β-oxidation. 3',5'-Dihydroxy-derivatives (from alkyl-resorcinols, flavanols, proanthocyanidins), and 4'-hydroxy-phenolic acids (from tyrosine, p-coumaric acid, naringenin) are β-oxidation substrates yielding benzoic acids. In contrast, 3',4',5'-tri-substituted-derivatives, 3',4'-dihydroxy-derivatives and 3'-methoxy-4'-hydroxy-derivatives (from coffee, tea, cereals, many fruits and vegetables) are poor β-oxidation substrates with metabolism diverted via gut microbiota dehydroxylation, phenylvalerolactone formation and phase-2 conjugation, possibly a strategy to conserve limited pools of coenzyme A. 4'-Methoxy-derivatives (citrus fruits) or 3',4'-dimethoxy-derivatives (coffee) are susceptible to hepatic "reverse" hydrogenation suggesting incompatibility with enoyl-CoA-hydratase. Gut microbiota-produced 3'-hydroxy-4'-methoxy-derivatives (citrus fruits) and 3'-hydroxy-derivatives (numerous (poly)phenols) are excreted as the phenyl-hydracrylic acid β-oxidation intermediate suggesting incompatibility with hydroxy-acyl-CoA dehydrogenase, albeit with considerable inter-individual variation. Further investigation is required to explain inter-individual variation, factors determining the amino acid to which C6-C3 and C6-C1 metabolites are conjugated, the precise role(s) of l-carnitine, whether glycine might be limiting, and whether phenolic acid-modulation of β-oxidation explains how phenolic acids affect key metabolic conditions, such as fatty liver, carbohydrate metabolism and insulin resistance.
Collapse
Affiliation(s)
- Michael N Clifford
- School of Bioscience and Medicine, University of Surrey, Guildford, UK
- Department of Nutrition, Dietetics and Food, Monash University, Clayton, Australia
| | - Laurence J King
- School of Bioscience and Medicine, University of Surrey, Guildford, UK
| | - Asimina Kerimi
- Department of Nutrition, Dietetics and Food, Monash University, Clayton, Australia
| | - Maria Gema Pereira-Caro
- Department of Food Science and Health, Instituto Andaluz de Investigacion y Formacion Agraria Pesquera Alimentaria y de la Produccion Ecologica, Sevilla, Spain
| | - Gary Williamson
- Department of Nutrition, Dietetics and Food, Monash University, Clayton, Australia
| |
Collapse
|
17
|
Dardi P, dos Santos-Eichler RA, de Oliveira S, Vinolo MAR, Câmara NOS, Rossoni LV. Reduced intestinal butyrate availability is associated with the vascular remodeling in resistance arteries of hypertensive rats. Front Physiol 2022; 13:998362. [PMID: 36246106 PMCID: PMC9558208 DOI: 10.3389/fphys.2022.998362] [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: 07/19/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
During hypertension an unbalance of short-chain fatty acids (SCFAs) production by intestinal bacteria is described. However, no data evaluate the association of SCFAs and vascular remodeling in hypertension, which is an important hallmark of this disease. Thus, the present study aims to evaluate the correlations between SCFAs availability and the resistance arteries remodeling in hypertension, as well as to identify the possible pathway by which the SCFAs could exert a structural and mechanical influence. Hence, male spontaneously hypertensive rats (SHR) and normotensive Wistar rats had blood pressure measured by tail-cuff plethysmography; fecal SCFAs content assessed by gas chromatography; gene expression of SCFAs-transporters in gut epithelium and SCFAs-sensing receptors on mesenteric resistance arteries (MRA) quantified by PCR; and MRA structural and mechanical parameters analyzed by pressure myograph. Reduced butyrate fecal content was found in SHR, with no changes in propionate and acetate, as well as decreased mRNA levels of SCFAs-transporters (MCT1, MCT4, and SMCT1) in the intestinal epithelium. In addition, lower gene expression of SCFAs-sensing receptors (GPR41, GPR43, and GPR109a, but not Olfr78) was identified in MRAs of SHR, which also shows inward eutrophic remodeling with stiffness. Butyrate content presented a negative correlation with systolic blood pressure and with the structural alterations found on MRAs, while a positive correlation between butyrate content and mechanical parameters was detected. Altogether the present study suggests that lower butyrate content due to ineffective SCFA bioavailability, associated with lower SCFAs-sensing receptors expression, could favor MRA remodeling, increasing peripheral vascular resistance and worsening hypertension prognosis.
Collapse
Affiliation(s)
- Patrizia Dardi
- Laboratory of Vascular Physiology, Institute of Biomedical Science, Department of Physiology and Biophysics, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Sarah de Oliveira
- Laboratory of Immunoinflammation, Institute of Biology, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Marco Aurélio Ramirez Vinolo
- Laboratory of Immunoinflammation, Institute of Biology, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Niels Olsen Saraiva Câmara
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Science, Department of Immunology, University of Sao Paulo, Sao Paulo, Brazil
| | - Luciana Venturini Rossoni
- Laboratory of Vascular Physiology, Institute of Biomedical Science, Department of Physiology and Biophysics, University of Sao Paulo, Sao Paulo, Brazil
- *Correspondence: Luciana Venturini Rossoni,
| |
Collapse
|
18
|
Zhou W, Tao W, Wang M, Liu W, Xing J, Yang Y. Dendrobium officinale Xianhu 2 polysaccharide helps forming a healthy gut microbiota and improving host immune system: An in vitro and in vivo study. Food Chem 2022; 401:134211. [PMID: 36122490 DOI: 10.1016/j.foodchem.2022.134211] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 01/08/2023]
Abstract
Dendrobium officinale is widely consumed owing to its numerous beneficial effects. We aimed to characterize polysaccharides of Dendrobium officinale (DOP) from the stems of Dendrobium officinale Xianhu 2 and clarify whether it benefit the intestinal microbiota and the immune system. The DOP weighed 291 kDa and comprised mannose, glucose, galactose, and rhamnose at 59.31:33.31:1.00:0.51 M ratio. In in vitro/vivo studies, DOP significantly increased benign intestinal microbe proportion (Lactobacillus, etc.), but reduced harmful bacteria (Escherichia_Shigella) (P < 0.05), and significantly increased butyric acid production (P < 0.05). Concentrations of 2 g/L DOP for in vitro fermentation and 100 mg/kg body weight for the mouse model were effective. In mice, DOP significantly reduced CRP, CD3, CD4, and TNF-α levels and increased C4 levels (P < 0.05). DOP might influence the immune system indirectly through regulation of the gut microbiota. Its possible regulation mechanism was that DOP reduced CD4+ Th cells proliferation so that reduced the secretion of TNF-α.
Collapse
Affiliation(s)
- Wanyi Zhou
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Wenyang Tao
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Mingzhe Wang
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Wei Liu
- Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Jianrong Xing
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Ying Yang
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China.
| |
Collapse
|
19
|
Liu G, Duan Y, Yang S, Yu M, Lv Z. Simultaneous quantification of marine neutral neoagaro-oligosaccharides and agar-oligosaccharides by the UHPLC-MS/MS method: application to the intestinal transport study by using the Caco-2 cell monolayer. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:2227-2234. [PMID: 35616101 DOI: 10.1039/d2ay00700b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A sensitive and robust ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method was established for the first time to simultaneously quantify marine neutral neoagaro-oligosaccharides (NAOS) and agar-oligosaccharides (AOS) with different degrees of polymerization (DP) in Hanks' balanced salt solution (HBSS). The separation was achieved on a BEH amide column using a mobile phase of acetonitrile-10 mmol L-1 ammonium acetate (58 : 42, v/v) with an isocratic elution program. The total analysis time was 3.5 min. The mass spectra were acquired in the multiple reaction monitoring (MRM) pattern by using a heated-electrospray ionization (H-ESI) source operating in the positive ionization mode. The linear range was 40-20 000 nmol L-1. The accuracy and precision ranged from 91.5 to 110.0% and 0.9 to 10.4%, respectively. The extraction recovery was consistent and reproducible. The stability was within 90.3-110.8%. The matrix effect, carryover, and dilution integrity were all satisfactory. Moreover, the validated method was successfully applied to the intestinal transport study by using the Caco-2 cell monolayer in vitro. The results revealed that neoagarobiose, neoagarotetraose, neoagarohexaose, agarotriose, agaropentose, and agaroheptose were transported by a paracellular pathway.
Collapse
Affiliation(s)
- Guilin Liu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China.
| | - Yunhai Duan
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China.
| | - Shuang Yang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, People's Republic of China
- Key Laboratory of Glycoscience & Glycotechnology of Shandong Province, Qingdao 266003, People's Republic of China
- Key Laboratory of Marine Drugs, Ministry of Education of China, Qingdao 266003, People's Republic of China
| | - Mingming Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, People's Republic of China
- Key Laboratory of Glycoscience & Glycotechnology of Shandong Province, Qingdao 266003, People's Republic of China
- Key Laboratory of Marine Drugs, Ministry of Education of China, Qingdao 266003, People's Republic of China
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, People's Republic of China
- Key Laboratory of Glycoscience & Glycotechnology of Shandong Province, Qingdao 266003, People's Republic of China
- Key Laboratory of Marine Drugs, Ministry of Education of China, Qingdao 266003, People's Republic of China
| |
Collapse
|
20
|
Pan Y, Li H, Shahidi F, Luo T, Deng Z. Interactions among dietary phytochemicals and nutrients: Role of cell membranes. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2022.03.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
21
|
Xu Y, Li T, Yang W, Sun M, An R, Du W. The ratio of xylooligosaccharide to ferulic acid affects faecal ferulic acid content, short chain fatty acid output, and gut stress. ACTA ALIMENTARIA 2021. [DOI: 10.1556/066.2021.00058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Abstract
There have been contradicting observations regarding the prebiotic efficacy of feruloylated oligosaccharides (FOs) extracted from different varieties of cereals with varying oligosaccharides and ferulic acid (FA) levels. The present study was performed to determine whether the mass ratio of xylooligosaccharide (XOS) to FA influences their combined effects on faecal FA content, short chain fatty acid (SCFA) output, and gut stress of d-galactose-treated aging rats. The results show that there was no significant difference in the faecal FA levels of rats fed with 5:1 and 10:1 XOS:FA diet, although the FA level in the 5:1-supplemented diet was twice as much as in the 10:1 diet. More utilisation of FA decreased butyric acid and SCFA output in the faeces for diet 5:1 compared with diets 10:1 XOS:FA or XOS alone. Furthermore, compared with 10:1 XOS:FA or XOS alone treatments, the 5:1 XOS:FA diet resulted in increased 1-diphenyl-2-picrylhydrazyl activity and higher ratios of Bifidobacterium or Lactobacillus to Escherichia coli (P < 0.05), while not increasing the number of probiotic Bifidobacterium and Lactobacillus. These findings suggest that under the specific stress level set for this study, the sufficient amount of FA added to XOS (5:1) can stimulate FA utilisation to modify gut redox balance, while reducing faecal SCFA output.
Collapse
Affiliation(s)
- Y.H. Xu
- 1 School of Ecology and Nature Conservation, Beijing Forestry University, Beijing 100083, China
| | - T. Li
- 2 Department of Food Science and Engineering, Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
| | - W.L. Yang
- 2 Department of Food Science and Engineering, Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
| | - M.Y. Sun
- 2 Department of Food Science and Engineering, Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
| | - R.Z. An
- 2 Department of Food Science and Engineering, Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
| | - W.M. Du
- 2 Department of Food Science and Engineering, Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, China
| |
Collapse
|
22
|
Magaña AA, Kamimura N, Soumyanath A, Stevens JF, Maier CS. Caffeoylquinic acids: chemistry, biosynthesis, occurrence, analytical challenges, and bioactivity. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2021; 107:1299-1319. [PMID: 34171156 PMCID: PMC9084498 DOI: 10.1111/tpj.15390] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/15/2021] [Accepted: 06/19/2021] [Indexed: 05/02/2023]
Abstract
Caffeoylquinic acids (CQAs) are specialized plant metabolites we encounter in our daily life. Humans consume CQAs in mg-to-gram quantities through dietary consumption of plant products. CQAs are considered beneficial for human health, mainly due to their anti-inflammatory and antioxidant properties. Recently, new biosynthetic pathways via a peroxidase-type p-coumaric acid 3-hydroxylase enzyme were discovered. More recently, a new GDSL lipase-like enzyme able to transform monoCQAs into diCQA was identified in Ipomoea batatas. CQAs were recently linked to memory improvement; they seem to be strong indirect antioxidants via Nrf2 activation. However, there is a prevalent confusion in the designation and nomenclature of different CQA isomers. Such inconsistencies are critical and complicate bioactivity assessment since different isomers differ in bioactivity and potency. A detailed explanation regarding the origin of such confusion is provided, and a recommendation to unify nomenclature is suggested. Furthermore, for studies on CQA bioactivity, plant-based laboratory animal diets contain CQAs, which makes it difficult to include proper control groups for comparison. Therefore, a synthetic diet free of CQAs is advised to avoid interferences since some CQAs may produce bioactivity even at nanomolar levels. Biotransformation of CQAs by gut microbiota, the discovery of new enzymatic biosynthetic and metabolic pathways, dietary assessment, and assessment of biological properties with potential for drug development are areas of active, ongoing research. This review is focused on the chemistry, biosynthesis, occurrence, analytical challenges, and bioactivity recently reported for mono-, di-, tri-, and tetraCQAs.
Collapse
Affiliation(s)
- Armando Alcázar Magaña
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, USA
| | - Naofumi Kamimura
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Niigata, Japan
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, USA
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, USA
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
23
|
Li R, Mao Z, Ye X, Zuo T. Human Gut Microbiome and Liver Diseases: From Correlation to Causation. Microorganisms 2021; 9:1017. [PMID: 34066850 PMCID: PMC8151257 DOI: 10.3390/microorganisms9051017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/25/2021] [Accepted: 04/30/2021] [Indexed: 02/06/2023] Open
Abstract
The important role of human gut microbiota in liver diseases has long been recognized as dysbiosis and the translocation of certain microbes from the gut to liver. With the development of high-throughput DNA sequencing, the complexity and integrity of the gut microbiome in the whole spectrum of liver diseases is emerging. Specific patterns of gut microbiota have been identified in liver diseases with different causes, including alcoholic, non-alcoholic, and virus induced liver diseases, or even at different stages, ranging from steatohepatitis, fibrosis, cirrhosis, to hepatocellular carcinoma. At the same time, the mechanism of how microbiota contributes to liver diseases goes beyond the traditional function of the gut-liver axis which could lead to liver injury and inflammation. With the application of proteomics, metabolomics, and modern molecular technologies, more microbial metabolites and the complicated interaction of microbiota with host immunity come into our understanding in the liver pathogenesis. Germ-free animal models serve as a workhorse to test the function of microbiota and their derivatives in liver disease models. Here, we review the current evidence on the relationship between gut microbiota and liver diseases, and the mechanisms underlying this phenotype. In addition to original liver diseases, gut microbiota might also affect liver injury in systemic disorders involving multiple organs, as in the case of COVID-19 at a severe state. A better understanding of the gut microbial contribution to liver diseases might help us better benefit from this guest-host relationship and pave the way for novel therapies.
Collapse
Affiliation(s)
- Rui Li
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan 430070, China;
| | - Zhengsheng Mao
- Department of Neurology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China;
| | - Xujun Ye
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan 430070, China;
| | - Tao Zuo
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510000, China
| |
Collapse
|
24
|
Wu DT, Nie XR, Gan RY, Guo H, Fu Y, Yuan Q, Zhang Q, Qin W. In vitro digestion and fecal fermentation behaviors of a pectic polysaccharide from okra (Abelmoschus esculentus) and its impacts on human gut microbiota. Food Hydrocoll 2021. [DOI: 10.1016/j.foodhyd.2020.106577] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
25
|
Coppola S, Avagliano C, Calignano A, Berni Canani R. The Protective Role of Butyrate against Obesity and Obesity-Related Diseases. Molecules 2021; 26:molecules26030682. [PMID: 33525625 PMCID: PMC7865491 DOI: 10.3390/molecules26030682] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 12/14/2022] Open
Abstract
Worldwide obesity is a public health concern that has reached pandemic levels. Obesity is the major predisposing factor to comorbidities, including type 2 diabetes, cardiovascular diseases, dyslipidemia, and non-alcoholic fatty liver disease. The common forms of obesity are multifactorial and derive from a complex interplay of environmental changes and the individual genetic predisposition. Increasing evidence suggest a pivotal role played by alterations of gut microbiota (GM) that could represent the causative link between environmental factors and onset of obesity. The beneficial effects of GM are mainly mediated by the secretion of various metabolites. Short-chain fatty acids (SCFAs) acetate, propionate and butyrate are small organic metabolites produced by fermentation of dietary fibers and resistant starch with vast beneficial effects in energy metabolism, intestinal homeostasis and immune responses regulation. An aberrant production of SCFAs has emerged in obesity and metabolic diseases. Among SCFAs, butyrate emerged because it might have a potential in alleviating obesity and related comorbidities. Here we reviewed the preclinical and clinical data that contribute to explain the role of butyrate in this context, highlighting its crucial contribute in the diet-GM-host health axis.
Collapse
Affiliation(s)
- Serena Coppola
- Department of Translational Medical Science, University of Naples Federico II, 80131 Naples, Italy;
- ImmunoNutriton Lab at CEINGE Advanced Biotechnologies, University of Naples Federico II, 80131 Naples, Italy
| | - Carmen Avagliano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (C.A.); (A.C.)
| | - Antonio Calignano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (C.A.); (A.C.)
| | - Roberto Berni Canani
- Department of Translational Medical Science, University of Naples Federico II, 80131 Naples, Italy;
- ImmunoNutriton Lab at CEINGE Advanced Biotechnologies, University of Naples Federico II, 80131 Naples, Italy
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples Federico II, 80131 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-081-7462680
| |
Collapse
|
26
|
A Role for Gut Microbiome Fermentative Pathways in Fatty Liver Disease Progression. J Clin Med 2020; 9:jcm9051369. [PMID: 32392712 PMCID: PMC7291163 DOI: 10.3390/jcm9051369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/24/2020] [Accepted: 05/03/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifactorial disease in which environmental and genetic factors are involved. Although the molecular mechanisms involved in NAFLD onset and progression are not completely understood, the gut microbiome (GM) is thought to play a key role in the process, influencing multiple physiological functions. GM alterations in diversity and composition directly impact disease states with an inflammatory course, such as non-alcoholic steatohepatitis (NASH). However, how the GM influences liver disease susceptibility is largely unknown. Similarly, the impact of strategies targeting the GM for the treatment of NASH remains to be evaluated. This review provides a broad insight into the role of gut microbiota in NASH pathogenesis, as a diagnostic tool, and as a therapeutic target in this liver disease. We highlight the idea that the balance in metabolic fermentations can be key in maintaining liver homeostasis. We propose that an overabundance of alcohol-fermentation pathways in the GM may outcompete healthier, acid-producing members of the microbiota. In this way, GM ecology may precipitate a self-sustaining vicious cycle, boosting liver disease progression.
Collapse
|
27
|
Clifford MN, Kerimi A, Williamson G. Bioavailability and metabolism of chlorogenic acids (acyl‐quinic acids) in humans. Compr Rev Food Sci Food Saf 2020; 19:1299-1352. [DOI: 10.1111/1541-4337.12518] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 10/08/2019] [Accepted: 11/13/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Michael N. Clifford
- School of Bioscience and Medicine, Faculty of Health and Medical SciencesUniversity of Surrey Guildford UK
- Department of Nutrition, Dietetics, and Food, School of Clinical Sciences at Monash HealthFaculty of Medicine Nursing and Health SciencesMonash University Notting Hill Victoria Australia
| | - Asimina Kerimi
- Department of Nutrition, Dietetics, and Food, School of Clinical Sciences at Monash HealthFaculty of Medicine Nursing and Health SciencesMonash University Notting Hill Victoria Australia
| | - Gary Williamson
- Department of Nutrition, Dietetics, and Food, School of Clinical Sciences at Monash HealthFaculty of Medicine Nursing and Health SciencesMonash University Notting Hill Victoria Australia
| |
Collapse
|
28
|
Wang M, Chen G, Chen D, Ye H, Sun Y, Zeng X, Liu Z. Purified fraction of polysaccharides from Fuzhuan brick tea modulates the composition and metabolism of gut microbiota in anaerobic fermentation in vitro. Int J Biol Macromol 2019; 140:858-870. [PMID: 31446105 DOI: 10.1016/j.ijbiomac.2019.08.187] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/17/2019] [Accepted: 08/21/2019] [Indexed: 12/18/2022]
Abstract
One purified fraction from crude Fuzhuan brick tea polysaccharides (FBTPS), FBTPS-3, was obtained through column chromatography of DEAE Sepharose Fast Flow. The chemical properties and probiotic effects of FBTPS-3 were evaluated by fermentation in vitro. Moreover, the effects of FBTPS-3 on the function and metabolic pathway of gut microbiota were investigated by metagenomic sequencing. The results showed that FBTPS-3 was an heteropolysaccharide with molecular weight of 741 kDa, which was mainly composed of Man, Rha, GalA, Gal and Ara in molar ratio of 8.7:15.5:42.2:19.7:13.9. The contents of carbohydrates and uronic acid in FBTPS-3 were 44.78 ± 2.85% and 40.4 ± 2.11%, respectively. After fermentation, the molecular weight of FBTPS-3 and content of carbohydrates were significantly decreased, indicating that FBTPS-3 could be utilized by gut microbiota. Furthermore, the relative abundances of Bacteroides, Megasphaera and Prevotella were significantly increased by FBTPS-3. FBTPS-3 also significantly promoted the production of acetic, propionic and n-butyric acids. Based on the metagenomic sequencing, it was found that FBTPS-3 significantly enriched the metabolic pathway of starch and sucrose. All the results suggest that FBTPS-3 is expected to be developed as functional ingredients or foods to improve the host health through regulating the gut microbiota and physiological metabolic functions.
Collapse
Affiliation(s)
- Mingjia Wang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Guijie Chen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Dan Chen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Hong Ye
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Yi Sun
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | - Zhonghua Liu
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| |
Collapse
|
29
|
Xu F, Zhu L, Qian C, Zhou J, Geng D, Li P, Xuan W, Wu F, Zhao K, Kong W, Qin Y, Liang L, Liu L, Liu X. Impairment of Intestinal Monocarboxylate Transporter 6 Function and Expression in Diabetic Rats Induced by Combination of High-Fat Diet and Low Dose of Streptozocin: Involvement of Butyrate-Peroxisome Proliferator-Activated Receptor- γ Activation. Drug Metab Dispos 2019; 47:556-566. [PMID: 30923035 DOI: 10.1124/dmd.118.085803] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Generally, diabetes remarkably alters the expression and function of intestinal drug transporters. Nateglinide and bumetanide are substrates of monocarboxylate transporter 6 (MCT6). We investigated whether diabetes down-regulated the function and expression of intestinal MCT6 and the possible mechanism in diabetic rats induced by a combination of high-fat diet and low-dose streptozocin. Our results indicated that diabetes significantly decreased the oral plasma exposure of nateglinide. The plasma peak concentration and area under curve in diabetic rats were 16.9% and 28.2% of control rats, respectively. Diabetes significantly decreased the protein and mRNA expressions of intestinal MCT6 and oligopeptide transporter 1 (PEPT1) but up-regulated peroxisome proliferator-activated receptor γ (PPARγ) protein level. Single-pass intestinal perfusion demonstrated that diabetes prominently decreased the absorption of nateglinide and bumetanide. The MCT6 inhibitor bumetanide, but not PEPT1 inhibitor glycylsarcosine, significantly inhibited intestinal absorption of nateglinide in rats. Coadministration with bumetanide remarkably decreased the oral plasma exposure of nateglinide in rats. High concentrations of butyrate were detected in the intestine of diabetic rats. In Caco-2 cells (a human colorectal adenocarcinoma cell line), bumetanide and MCT6 knockdown remarkably inhibited the uptake of nateglinide. Butyrate down-regulated the function and expression of MCT6 in a concentration-dependent manner but increased PPARγ expression. The decreased expressions of MCT6 by PPARγ agonist troglitazone or butyrate were reversed by both PPARγ knockdown and PPARγ antagonist 2-chloro-5-nitro-N-phenylbenzamide (GW9662). Four weeks of butyrate treatment significantly decreased the oral plasma concentrations of nateglinide in rats, accompanied by significantly higher intestinal PPARγ and lower MCT6 protein levels. In conclusion, diabetes impaired the expression and function of intestinal MCT6 partly via butyrate-mediated PPARγ activation, decreasing the oral plasma exposure of nateglinide.
Collapse
Affiliation(s)
- Feng Xu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Liang Zhu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Chaoqun Qian
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Junjie Zhou
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Donghao Geng
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ping Li
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Wenjing Xuan
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Fangge Wu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Kaijing Zhao
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Weimin Kong
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yuanyuan Qin
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Limin Liang
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
30
|
Vitaglione P, Mennella I, Ferracane R, Goldsmith F, Guice J, Page R, Raggio AM, Coulon D, Martin R, Keenan M. Gut fermentation induced by a resistant starch rich whole grain diet explains serum concentration of dihydroferulic acid and hippuric acid in a model of ZDF rats. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.12.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
31
|
Oat bran, but not its isolated bioactive β-glucans or polyphenols, have a bifidogenic effect in an in vitro fermentation model of the gut microbiota. Br J Nutr 2019; 121:549-559. [PMID: 30688188 DOI: 10.1017/s0007114518003501] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Wholegrain oats are known to modulate the human gut microbiota and have prebiotic properties (increase the growth of some health-promoting bacterial genera within the colon). Research to date mainly attributes these effects to the fibre content; however, oat is also a rich dietary source of polyphenols, which may contribute to the positive modulation of gut microbiota. In vitro anaerobic batch-culture experiments were performed over 24 h to evaluate the impact of two different doses (1 and 3 % (w/v)) of oat bran, matched concentrations of β-glucan extract or polyphenol mix, on the human faecal microbiota composition using 16S RNA gene sequencing and SCFA analysis. Supplementation with oats increased the abundance of Proteobacteria (P <0·01) at 10 h, Bacteroidetes (P <0·05) at 24 h and concentrations of acetic and propionic acid increased at 10 and 24 h compared with the NC. Fermentation of the 1 % (w/v) oat bran resulted in significant increase in SCFA production at 24 h (86 (sd 27) v. 28 (sd 5) mm; P <0·05) and a bifidogenic effect, increasing the relative abundance of Bifidobacterium unassigned at 10 h and Bifidobacterium adolescentis (P <0·05) at 10 and 24 h compared with NC. Considering the β-glucan treatment induced an increase in the phylum Bacteroidetes at 24 h, it explains the Bacteriodetes effects of oats as a food matrix. The polyphenol mix induced an increase in Enterobacteriaceae family at 24 h. In conclusion, in this study, we found that oats increased bifidobacteria, acetic acid and propionic acid, and this is mediated by the synergy of all oat compounds within the complex food matrix, rather than its main bioactive β-glucan or polyphenols. Thus, oats as a whole food led to the greatest impact on the microbiota.
Collapse
|
32
|
Han F, Yang P, Wang H, Fernandes I, Mateus N, Liu Y. Digestion and absorption of red grape and wine anthocyanins through the gastrointestinal tract. Trends Food Sci Technol 2019. [DOI: 10.1016/j.tifs.2018.11.025] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Tripathi A, Debelius J, Brenner DA, Karin M, Loomba R, Schnabl B, Knight R. Publisher Correction: The gut-liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol 2018; 15:785. [PMID: 29785003 PMCID: PMC7133393 DOI: 10.1038/s41575-018-0031-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the original version of Table 1 published online, upward arrows to indicate increased translocation of PAMPs were missing from the row entitled 'Translocation' for both the column on alcoholic liver disease and nonalcoholic fatty liver disease. This error has now been updated in the PDF and HTML version of the article.
Collapse
|
34
|
Kristek A, Schär MY, Soycan G, Alsharif S, Kuhnle GGC, Walton G, Spencer JPE. The gut microbiota and cardiovascular health benefits: A focus on wholegrain oats. NUTR BULL 2018. [DOI: 10.1111/nbu.12354] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- A. Kristek
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - M. Y. Schär
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - G. Soycan
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - S. Alsharif
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - G. G. C. Kuhnle
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - G. Walton
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| | - J. P. E. Spencer
- Department of Food and Nutritional Sciences; University of Reading; Reading UK
| |
Collapse
|
35
|
Xu GD, Cai L, Ni YS, Tian SY, Lu YQ, Wang LN, Chen LL, Ma WY, Deng SP. Comparisons of Effects on Intestinal Short-Chain Fatty Acid Concentration after Exposure of Two Glycosidase Inhibitors in Mice. Biol Pharm Bull 2018; 41:1024-1033. [PMID: 29962399 DOI: 10.1248/bpb.b17-00978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acarbose and voglibose are the most widely used diabetes drugs as glycosidase inhibitors. In this study, the use of these two inhibitors significantly increased the content of starch in large intestine, and altered the concentration of short-chain fatty acids (SCFAs) by affecting the intestinal microbiota. However, there are some differences in the intestinal microbiome of the two groups of mice, mainly in bacteria such as Bacteroidaceae bacteroides and Desulfovibrionaceae desulfovibrio. The productions of acetate and propionate in caecum in voglibose group were significantly higher than those in acarbose group and two kinds of glycosidase inhibitors were close in the production of butyrate in caecum. The Tax4Fun analysis based on Kyoto Encyclopedia of Genes and Genomes (KEGG) data indicated that different productions of acetate and propionate between acarbose group and voglibose group may be related to 2-oxoisovalerate dehydrogenase and pyruvate oxidase. In addition, in-vitro experiments suggested that voglibose had less effect on epithelial cells than acarbose after direct stimulation. According to the recent researches of SCFAs produced by intestinal microbiota, our comparative study shown higher concentration of these beneficial fatty acids in the lumen of voglibose-treated mice, which implied a lower level of inflammation.
Collapse
Affiliation(s)
- Guo-Dong Xu
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Lei Cai
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Yi-Shu Ni
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Shi-Yi Tian
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Ying-Qi Lu
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Li-Na Wang
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Lian-Lian Chen
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Wen-Ya Ma
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Shao-Ping Deng
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| |
Collapse
|
36
|
Tripathi A, Debelius J, Brenner DA, Karin M, Loomba R, Schnabl B, Knight R. The gut-liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol 2018; 15:397-411. [PMID: 29748586 PMCID: PMC6319369 DOI: 10.1038/s41575-018-0011-z] [Citation(s) in RCA: 864] [Impact Index Per Article: 123.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the past decade, an exciting realization has been that diverse liver diseases - ranging from nonalcoholic steatohepatitis, alcoholic steatohepatitis and cirrhosis to hepatocellular carcinoma - fall along a spectrum. Work on the biology of the gut-liver axis has assisted in understanding the basic biology of both alcoholic fatty liver disease and nonalcoholic fatty liver disease (NAFLD). Of immense importance is the advancement in understanding the role of the microbiome, driven by high-throughput DNA sequencing and improved computational techniques that enable the complexity of the microbiome to be interrogated, together with improved experimental designs. Here, we review gut-liver communications in liver disease, exploring the molecular, genetic and microbiome relationships and discussing prospects for exploiting the microbiome to determine liver disease stage and to predict the effects of pharmaceutical, dietary and other interventions at a population and individual level. Although much work remains to be done in understanding the relationship between the microbiome and liver disease, rapid progress towards clinical applications is being made, especially in study designs that complement human intervention studies with mechanistic work in mice that have been humanized in multiple respects, including the genetic, immunological and microbiome characteristics of individual patients. These 'avatar mice' could be especially useful for guiding new microbiome-based or microbiome-informed therapies.
Collapse
Affiliation(s)
- Anupriya Tripathi
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, USA
| | - Justine Debelius
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - David A Brenner
- NAFLD Research Center, Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
| | - Michael Karin
- Department of Pediatrics, University of California, San Diego, CA, USA
- Department of Computer Science and Engineering, University of California, San Diego, CA, USA
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, CA, USA
| | - Bernd Schnabl
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego, CA, USA.
- Department of Computer Science and Engineering, University of California, San Diego, CA, USA.
- Center for Microbiome Innovation, University of California, San Diego, CA, USA.
| |
Collapse
|
37
|
Lund J, Aas V, Tingstad RH, Van Hees A, Nikolić N. Utilization of lactic acid in human myotubes and interplay with glucose and fatty acid metabolism. Sci Rep 2018; 8:9814. [PMID: 29959350 PMCID: PMC6026123 DOI: 10.1038/s41598-018-28249-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/11/2018] [Indexed: 01/05/2023] Open
Abstract
Once assumed only to be a waste product of anaerobe glycolytic activity, lactate is now recognized as an energy source in skeletal muscles. While lactate metabolism has been extensively studied in vivo, underlying cellular processes are poorly described. This study aimed to examine lactate metabolism in cultured human myotubes and to investigate effects of lactate exposure on metabolism of oleic acid and glucose. Lactic acid, fatty acid and glucose metabolism were studied in myotubes using [14C(U)]lactic acid, [14C]oleic acid and [14C(U)]glucose, respectively. Myotubes expressed both the MCT1, MCT2, MCT3 and MCT4 lactate transporters, and lactic acid was found to be a substrate for both glycogen synthesis and lipid storage. Pyruvate and palmitic acid inhibited lactic acid oxidation, whilst glucose and α-cyano-4-hydroxycinnamic acid inhibited lactic acid uptake. Acute addition of lactic acid inhibited glucose and oleic acid oxidation, whereas oleic acid uptake was increased. Pretreatment with lactic acid for 24 h did not affect glucose or oleic acid metabolism. By replacing glucose with lactic acid during the whole culturing period, glucose uptake and oxidation were increased by 2.8-fold and 3-fold, respectively, and oleic acid oxidation was increased 1.4-fold. Thus, lactic acid has an important role in energy metabolism of human myotubes.
Collapse
Affiliation(s)
- Jenny Lund
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Vigdis Aas
- Department of Life Sciences and Health, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Ragna H Tingstad
- Department of Life Sciences and Health, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Alfons Van Hees
- Department of Life Sciences and Health, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Nataša Nikolić
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway.
| |
Collapse
|
38
|
Domínguez-Avila JA, Wall-Medrano A, Velderrain-Rodríguez GR, Chen CYO, Salazar-López NJ, Robles-Sánchez M, González-Aguilar GA. Gastrointestinal interactions, absorption, splanchnic metabolism and pharmacokinetics of orally ingested phenolic compounds. Food Funct 2018; 8:15-38. [PMID: 28074953 DOI: 10.1039/c6fo01475e] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The positive health effects of phenolic compounds (PCs) have been extensively reported in the literature. An understanding of their bioaccessibility and bioavailability is essential for the elucidation of their health benefits. Before reaching circulation and exerting bioactions in target tissues, numerous interactions take place before and during digestion with either the plant or host's macromolecules that directly impact the organism and modulate their own bioaccessibility and bioavailability. The present work is focused on the gastrointestinal (GI) interactions that are relevant to the absorption and metabolism of PCs and how these interactions impact their pharmacokinetic profiles. Non-digestible cell wall components (fiber) interact intimately with PCs and delay their absorption in the small intestine, instead carrying them to the large intestine. PCs not bound to fiber interact with digestible nutrients in the bolus where they interfere with the digestion and absorption of proteins, carbohydrates, lipids, cholesterol, bile salts and micronutrients through the inhibition of digestive enzymes and enterocyte transporters and the disruption of micelle formation. PCs internalized by enterocytes may reach circulation (through transcellular or paracellular transport), be effluxed back into the lumen (P-glycoprotein, P-gp) or be metabolized by phase I and phase II enzymes. Some PCs can inhibit P-gp or phase I/II enzymes, which can potentially lead to drug-nutrient interactions. The absorption and pharmacokinetic parameters are modified by all of the interactions within the digestive tract and by the presence of other PCs. Undesirable interactions have promoted the development of nanotechnological approaches to promote the bioaccessibility, bioavailability, and bioefficacy of PCs.
Collapse
Affiliation(s)
- J Abraham Domínguez-Avila
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo, A.C. Carretera a la Victoria Km 0.6. C.P. 83304, Hermosillo, Sonora, Mexico.
| | - Abraham Wall-Medrano
- Departamento de Ciencias Químico-Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Anillo Envolvente del Pronaf y Estocolmo s/n, CP 32310, Cd. Juárez, Chihuahua, Mexico.
| | - Gustavo R Velderrain-Rodríguez
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo, A.C. Carretera a la Victoria Km 0.6. C.P. 83304, Hermosillo, Sonora, Mexico.
| | - C-Y Oliver Chen
- Antioxidants Research Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, 711 Washington Street, Boston, Massachusetts 02111, USA.
| | - Norma Julieta Salazar-López
- Departamento de Investigación y Posgrado en Alimentos, Universidad de Sonora, Blvd. Luis Encinas y Rosales S/N Col. Centro, C.P. 83000, Hermosillo, Sonora, Mexico.
| | - Maribel Robles-Sánchez
- Departamento de Investigación y Posgrado en Alimentos, Universidad de Sonora, Blvd. Luis Encinas y Rosales S/N Col. Centro, C.P. 83000, Hermosillo, Sonora, Mexico.
| | - Gustavo A González-Aguilar
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo, A.C. Carretera a la Victoria Km 0.6. C.P. 83304, Hermosillo, Sonora, Mexico.
| |
Collapse
|
39
|
Gauer JS, Tumova S, Lippiat JD, Kerimi A, Williamson G. Differential patterns of inhibition of the sugar transporters GLUT2, GLUT5 and GLUT7 by flavonoids. Biochem Pharmacol 2018; 152:11-20. [PMID: 29548810 DOI: 10.1016/j.bcp.2018.03.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/09/2018] [Indexed: 11/16/2022]
Abstract
Only limited data are available on the inhibition of the sugar transporter GLUT5 by flavonoids or other classes of bioactives. Intestinal GLUT7 is poorly characterised and no information exists concerning its inhibition. We aimed to study the expression of GLUT7 in Caco-2/TC7 intestinal cells, and evaluate inhibition of glucose transport by GLUT2 and GLUT7, and of fructose transport by GLUT2, GLUT5 and GLUT7, by flavonoids. Differentiated Caco-2/TC7 cell monolayers were used to investigate GLUT7 expression, as well as biotinylation and immunofluorescence to assess GLUT7 location. For mechanistic sugar transport studies, X. laevis oocytes were injected with individual mRNA, and GLUT protein expression on oocyte membranes was confirmed. Oocytes were incubated with D-[14C(U)]-glucose or D-[14C(U)]-fructose in the presence of flavonoids, and uptake was estimated by liquid scintilation counting. In differentiated Caco-2/TC7 cell monolayers, GLUT7 was mostly expressed apically. When applied apically, or to both compartments, sorbitol, galactose, L-glucose or sucrose did not affect GLUT7 mRNA expression. Fructose applied to both sides increased GLUT7 mRNA (13%, p ≤ 0.001) and total GLUT7 protein (2.7-fold, p ≤ 0.05), while the ratio between apical, basolateral and total GLUT7 protein was unchanged. In the X. laevis oocyte model, GLUT2-mediated glucose and fructose transport were inhibited by quercetin, (-)-epigallocatechin gallate (EGCG) and apigenin, GLUT5-mediated fructose transport was inhibited by apigenin and EGCG, but not by quercetin, and GLUT7-mediated uptake of both glucose and fructose was inhibited by apigenin, but not by quercetin nor EGCG. Expression of GLUT7 was increased by fructose, but only when applied to Caco-2/TC7 cells both apically and basolaterally. Since GLUT2, GLUT5 and GLUT7 show different patterns of inhibition by the tested flavonoids, we suggest that they have the potential to be used as investigational tools to distinguish sugar transporter activity in different biological settings.
Collapse
Affiliation(s)
- Julia S Gauer
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK
| | - Sarka Tumova
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK
| | - Jonathan D Lippiat
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Asimina Kerimi
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK
| | - Gary Williamson
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
40
|
Sadeghi Ekbatan S, Iskandar MM, Sleno L, Sabally K, Khairallah J, Prakash S, Kubow S. Absorption and Metabolism of Phenolics from Digests of Polyphenol-Rich Potato Extracts Using the Caco-2/HepG2 Co-Culture System. Foods 2018; 7:foods7010008. [PMID: 29329242 PMCID: PMC5789271 DOI: 10.3390/foods7010008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/23/2017] [Accepted: 01/09/2018] [Indexed: 01/15/2023] Open
Abstract
The bioactivity of dietary polyphenols depends upon gastrointestinal and hepatic metabolism of secondary microbial phenolic metabolites generated via colonic microbiota-mediated biotransformation. A polyphenol-rich potato extract (PRPE) containing chlorogenic, caffeic, and ferulic acids and rutin was digested in a dynamic multi-reactor gastrointestinal simulator of the human intestinal microbial ecosystem (GI model). Simulated digestion showed extensive degradation of the parent compounds and the generation of microbial phenolic metabolites. To characterize the transport and metabolism of microbial phenolic metabolites following digestion, a co-culture of intestinal Caco-2 and hepatic HepG2 cells was exposed to the PRPE-derived digests obtained from the colonic vessels. Following a 2 h incubation of the digesta with the Caco-2/HepG2 co-cultures, approximately 10–15% of ferulic, dihydrocaffeic, and dihydroferulic acids and 3–5% of 3-hydroxybenzoic, 3-hydroxyphenylpropionic, and coumaric acids were observed in the basolateral side, whereas 3-hydroxyphenylacetic acid, phenylpropanoic acid, and cinnamic acid were not detected. Subsequent HepG2 cellular metabolism led to major increases in ferulic, dihydrocaffeic, 3-hydroxyphenylpropionic, and coumaric acids ranging from 160–370%. These findings highlight the importance of hepatic metabolism towards the generation of secondary metabolites of polyphenols despite low selective Caco-2 cellular uptake of microbial phenolic metabolites.
Collapse
Affiliation(s)
- Shima Sadeghi Ekbatan
- School of Human Nutrition, McGill University, 21111 Lakeshore, Ste. Anne de Bellevue, Montreal, QC H9X3V9, Canada.
| | - Michele M Iskandar
- School of Human Nutrition, McGill University, 21111 Lakeshore, Ste. Anne de Bellevue, Montreal, QC H9X3V9, Canada.
| | - Lekha Sleno
- Chemistry Department, Université du Québec à Montréal, 2101 Jeanne-Mance, Montreal, QC H2X2J6, Canada.
| | - Kebba Sabally
- School of Human Nutrition, McGill University, 21111 Lakeshore, Ste. Anne de Bellevue, Montreal, QC H9X3V9, Canada.
| | - Joelle Khairallah
- School of Human Nutrition, McGill University, 21111 Lakeshore, Ste. Anne de Bellevue, Montreal, QC H9X3V9, Canada.
| | - Satya Prakash
- Department of Biomedical Engineering, Duff Medical Building, 3775 Rue University, Montreal, QC H3A2B4, Canada.
| | - Stan Kubow
- School of Human Nutrition, McGill University, 21111 Lakeshore, Ste. Anne de Bellevue, Montreal, QC H9X3V9, Canada.
| |
Collapse
|
41
|
Sivaprakasam S, Bhutia YD, Yang S, Ganapathy V. Short-Chain Fatty Acid Transporters: Role in Colonic Homeostasis. Compr Physiol 2017; 8:299-314. [PMID: 29357130 PMCID: PMC6019286 DOI: 10.1002/cphy.c170014] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Short-chain fatty acids (SCFA; acetate, propionate, and butyrate) are generated in colon by bacterial fermentation of dietary fiber. Though diffusion in protonated form is a significant route, carrier-mediated mechanisms constitute the major route for the entry of SCFA in their anionic form into colonic epithelium. Several transport systems operate in cellular uptake of SCFA. MCT1 (SLC16A1) and MCT4 (SLC16A3) are H+-coupled and mediate electroneutral transport of SCFA (H+: SCFA stoichiometry; 1:1). MCT1 is expressed both in the apical membrane and basolateral membrane of colonic epithelium whereas MCT4 specifically in the basolateral membrane. SMCT1 (SLC5A8) and SMCT2 (SLC5A12) are Na+-coupled; SMCT1-mediated transport is electrogenic (Na+: SCFA stoichiometry; 2:1) whereas SMCT2-mediated transport is electroneutral (Na+: SCFA stoichiometry; 1:1). SMCT1 and SMCT2 are expressed exclusively in the apical membrane. An anion-exchange mechanism also operates in the apical membrane in which SCFA entry in anionic form is coupled to bicarbonate efflux; the molecular identity of this exchanger however remains unknown. All these transporters are subject to regulation, notably by their substrates themselves; this process involves cell-surface receptors with SCFA as signaling molecules. There are significant alterations in the expression of these transporters in ulcerative colitis and colon cancer. The tumor-associated changes occur via transcriptional regulation by p53 and HIF1α and by promoter methylation. As SCFA are obligatory for optimal colonic health, the transporters responsible for the entry and transcellular transfer of these bacterial products in colonic epithelium are critical determinants of colonic function under physiological conditions and in disease states. © 2018 American Physiological Society. Compr Physiol 8:299-314, 2018.
Collapse
Affiliation(s)
- Sathish Sivaprakasam
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Yangzom D. Bhutia
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Shengping Yang
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| |
Collapse
|
42
|
Tumova S, Kerimi A, Porter KE, Williamson G. Transendothelial glucose transport is not restricted by extracellular hyperglycaemia. Vascul Pharmacol 2016; 87:219-229. [DOI: 10.1016/j.vph.2016.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/29/2016] [Accepted: 11/02/2016] [Indexed: 01/18/2023]
|
43
|
Van Rymenant E, Abrankó L, Tumova S, Grootaert C, Van Camp J, Williamson G, Kerimi A. Chronic exposure to short-chain fatty acids modulates transport and metabolism of microbiome-derived phenolics in human intestinal cells. J Nutr Biochem 2016; 39:156-168. [PMID: 27840292 PMCID: PMC5756543 DOI: 10.1016/j.jnutbio.2016.09.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 11/17/2022]
Abstract
Dietary fiber-derived short-chain fatty acids (SCFA) and phenolics produced by the gut microbiome have multiple effects on health. We have tested the hypothesis that long-term exposure to physiological concentrations of SCFA can affect the transport and metabolism of (poly)phenols by the intestinal epithelium using the Caco-2 cell model. Metabolites and conjugates of hesperetin (HT) and ferulic acid (FA), gut-derived from dietary hesperidin and chlorogenic acid, respectively, were quantified by LC-MS with authentic standards following transport across differentiated cell monolayers. Changes in metabolite levels were correlated with effects on mRNA and protein expression of key enzymes and transporters. Propionate and butyrate increased both FA transport and rate of appearance of FA glucuronide apically and basolaterally, linked to an induction of MCT1. Propionate was the only SCFA that augmented the rate of formation of basolateral FA sulfate conjugates, possibly via basolateral transporter up-regulation. In addition, propionate enhanced the formation of HT glucuronide conjugates and increased HT sulfate efflux toward the basolateral compartment. Acetate treatment amplified transepithelial transport of FA in the apical to basolateral direction, associated with lower levels of MCT1 protein expression. Metabolism and transport of both HT and FA were curtailed by the organic acid lactate owing to a reduction of UGT1A1 protein levels. Our data indicate a direct interaction between microbiota-derived metabolites of (poly)phenols and SCFA through modulation of transporters and conjugating enzymes and increase our understanding of how dietary fiber, via the microbiome, may affect and enhance uptake of bioactive molecules.
Collapse
Affiliation(s)
- Evelien Van Rymenant
- Department of Food Safety and Food Quality, Faculty of Bioscience Engineering, University of Ghent, 9000 Gent, Belgium.
| | - László Abrankó
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK.
| | - Sarka Tumova
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK.
| | - Charlotte Grootaert
- Department of Food Safety and Food Quality, Faculty of Bioscience Engineering, University of Ghent, 9000 Gent, Belgium.
| | - John Van Camp
- Department of Food Safety and Food Quality, Faculty of Bioscience Engineering, University of Ghent, 9000 Gent, Belgium.
| | - Gary Williamson
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK.
| | - Asimina Kerimi
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
44
|
Affiliation(s)
- Christine Morand
- INRA, UMR 1019, Human Nutrition Unit, Centre de Recherche Auvergne-Rhone-Alpes, F-63122 Saint-Genès-Champanelle, France.
| | - Helmut Sies
- Heinrich-Heine-Universitaet Duesseldorf, Institute for Biochemistry and Molecular Biology I, D-40225 Duesseldorf, Germany.
| |
Collapse
|