1
|
Luo J, Wang C, Ye W, He R, Huang L, Fang Z, Deng Q, Qiu M, Sun L, Gooneratne R. Effects of Lutjanus erythropterus Protein on Depression-like Behavior and Gut Microbiota in Stressed Juvenile Mice. Foods 2025; 14:330. [PMID: 39856996 PMCID: PMC11765133 DOI: 10.3390/foods14020330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025] Open
Abstract
Lutjanus erythropterus protein (Lep) exhibits anti-inflammatory effects, but its antidepressant activity is unknown. This study used a 44-day chronic unpredictable mild stress (CUMS) model to determine whether Lep has a beneficial effect through the gut-brain axis in 3-week-old male C57BL/6 mice. Gavaging with Lep solution alleviated the depression-like behavior and anxiety symptoms in CUMS growing mice. Administration of Lep decreased serum IL-1β, IL-2, IL-6, and TNF-α levels and restored colonic mucosal damage. In addition, Lep improved the disturbance of 5-hydroxytryptamine (5-HT) secretion in the gut-brain axis. Pearson analysis revealed that gut short-chain fatty acid (SCFAs) concentration significantly (p < 0.05) correlated with mucosal damage scores and the depression-like behavior index. Lep was able to prevent the gut SCFA enrichment. Lep upregulated gut Muribaculaceae and downregulated SCFA-producing bacteria by replenishing deficient amino acid (AA) (tryptophan, alanine, aspartate, glutamate) and decreased (p < 0.01) the gene abundance of the AA metabolism pathway of SCFA-producing bacteria, thereby preventing gut SCFA enrichment and alleviating associated depression-like behavior. These findings indicate that Lep could attenuate depression in CUMS juvenile mice via the gut microbiota-SCFA-brain axis.
Collapse
Affiliation(s)
- Jinjin Luo
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (C.W.); (W.Y.); (R.H.); (L.H.); (Z.F.); (Q.D.); (L.S.)
| | - Chen Wang
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (C.W.); (W.Y.); (R.H.); (L.H.); (Z.F.); (Q.D.); (L.S.)
| | - Weichang Ye
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (C.W.); (W.Y.); (R.H.); (L.H.); (Z.F.); (Q.D.); (L.S.)
| | - Ruiyang He
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (C.W.); (W.Y.); (R.H.); (L.H.); (Z.F.); (Q.D.); (L.S.)
| | - Ling Huang
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (C.W.); (W.Y.); (R.H.); (L.H.); (Z.F.); (Q.D.); (L.S.)
| | - Zhijia Fang
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (C.W.); (W.Y.); (R.H.); (L.H.); (Z.F.); (Q.D.); (L.S.)
| | - Qi Deng
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (C.W.); (W.Y.); (R.H.); (L.H.); (Z.F.); (Q.D.); (L.S.)
| | - Mei Qiu
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (C.W.); (W.Y.); (R.H.); (L.H.); (Z.F.); (Q.D.); (L.S.)
| | - Lijun Sun
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (C.W.); (W.Y.); (R.H.); (L.H.); (Z.F.); (Q.D.); (L.S.)
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, P.O. Box 85084, Lincoln 7647, New Zealand;
| |
Collapse
|
2
|
Wang XW, Ding YL, Li CL, Ma Q, Shi YG, Liu GE, Li CJ, Kang XL. Effects of rumen metabolite butyric acid on bovine skeletal muscle satellite cells proliferation, apoptosis and transcriptional states during myogenic differentiation. Domest Anim Endocrinol 2025; 90:106892. [PMID: 39418766 DOI: 10.1016/j.domaniend.2024.106892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Butyric acid, a pivotal short-chain fatty acid in rumen digestion, profoundly influences animal digestive and locomotor systems. Extensive research indicates its direct or indirect involvement in the growth and development of muscle and fat cells. However, the impact of butyric acid on the proliferation and differentiation of bovine skeletal muscle satellite cells (SMSCs) remains unclear. This study aimed to elucidate the effects of butyrate on SMSCs proliferation and differentiation. After isolating, SMSCs were subjected to varying concentrations of sodium butyrate (NaB) during the proliferation and differentiation stages. Optimal treatment conditions (1 mM NaB for 2 days) were determined based on proliferative force, cell viability, and mRNA expression of proliferation and differentiation marker genes. Transcriptome sequencing was employed to screen for differential gene expression between 1 mM NaB-treated and untreated groups during SMSCs differentiation. Results indicated that lower NaB concentrations (≤1.0 mM) inhibited proliferation while promoting differentiation and apoptosis after a 2-day treatment. Conversely, higher NaB concentrations (≥2.0 mM) suppressed proliferation and differentiation and induced apoptosis. Transcriptome sequencing revealed differential expression of genes(ND1, ND3, CYTB, COX2, ATP6, MYOZ2, MYOZ3, MYBPC1 and ATP6V0A4,etc.) were associated with SMSCs differentiation and energy metabolism, enriching pathways such as Oxidative phosphorylation, MAPK, and Wnt signaling. These findings offer valuable insights into the molecular mechanisms underlying butyrate regulation of bovine SMSCs proliferation and differentiation, as well as muscle fiber type conversion in the future study.
Collapse
Affiliation(s)
- Xiao-Wei Wang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Institute of Animal Science, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan750002, China
| | - Yan-Ling Ding
- Key Laboratory of Ruminant Molecular and Cellular Breeding, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Cheng-Long Li
- Key Laboratory of Ruminant Molecular and Cellular Breeding, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Qing Ma
- Institute of Animal Science, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan750002, China
| | - Yuan-Gang Shi
- Key Laboratory of Ruminant Molecular and Cellular Breeding, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - George E Liu
- Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, MD 20705, United States
| | - Cong-Jun Li
- Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, MD 20705, United States.
| | - Xiao-Long Kang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
3
|
Wang F, Xu J, Hu C, Lai J, Shen P, Lu Y, Jiang F. β-glucan improves intestinal health of pearl gentian grouper via activation of the p38 mitogen-activated protein kinase signaling pathway. FISH & SHELLFISH IMMUNOLOGY 2024; 153:109868. [PMID: 39216713 DOI: 10.1016/j.fsi.2024.109868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/04/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Our previous study has demonstrated that supplementation of yeast β-glucan improves intestinal health in pearl gentian grouper (Epinephelus lanceolatus♂ × Epinephelus fuscoguttatus♀), accompanied by the activation of the mitogen-activated protein kinase (MAPK) signaling pathway. In this study, we investigated the effects of perturbing p38 MAPK activity using an inhibitor on the intestinal health of β-glucan-injected pearl gentian grouper to elucidate the potential molecular mechanism underlying the protective effects of β-glucan on the fish gut. The pearl gentian grouper was categorized into four groups: PBS injected (CD group), β-glucan injected at a dose of 80 mg/kg (βG group), p38 MAPK inhibitor SB203580 injected at a dose of 1 mg/kg (SB203580 group), and a combination of β-glucan (80 mg/kg) and SB203580 (1 mg/kg) injected together (βG + SB203580 group). The results revealed that the introduction of SB203580 significantly suppressed the β-glucan-induced increase in p38α and p38β mRNA expression, as well as the phosphorylation of p38 MAPK. Both the βG group and SB203580 group exhibited reduced plica height and muscularis thickness. The βG + SB203580 group displayed a significant reduction in mucin cell level; interleukin 1β (il1β) mRNA expression; induced nitric oxide synthase, tumor necrosis factor α, and IL1β concentration; catalase and total antioxidant capacity activities. Additionally, there was a significant increase in the levels of intestinal malondialdehyde in the βG + SB203580 group compared to the βG group. The inhibition of the p38 MAPK signaling halted the trend of apoptosis-related caspase molecular expression induced by β-glucan. In conclusion, β-glucan injection resulted in elevated levels of mucous cells, nonspecific immunity, antioxidant capacity, and anti-apoptosis in grouper by modulating the p38 MAPK pathway. This study offers insights into the potential molecular mechanism underlying the protective effects of β-glucan on intestinal health in pearl gentian grouper.
Collapse
Affiliation(s)
- Fan Wang
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, China; College of Life Science and Technology of Guangxi University, Nanning 530005, Guangxi, China
| | - Jia Xu
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, China
| | - Chaoqun Hu
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, China
| | - Junxiang Lai
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, China
| | - Peihong Shen
- College of Life Science and Technology of Guangxi University, Nanning 530005, Guangxi, China
| | - Yishan Lu
- College of Fishery, Guangdong Ocean University, Zhanjiang 524088, China.
| | - Fajun Jiang
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, China.
| |
Collapse
|
4
|
Gao Y, Yao Q, Meng L, Wang J, Zheng N. Double-side role of short chain fatty acids on host health via the gut-organ axes. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 18:322-339. [PMID: 39290857 PMCID: PMC11406094 DOI: 10.1016/j.aninu.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 01/29/2024] [Accepted: 05/14/2024] [Indexed: 09/19/2024]
Abstract
Short chain fatty acids (SCFA) exist in dietary foods and are produced by the fermentation of gut microbiota, and are considered an important element for regulating host health. Through blood circulation, SCFA produced in the gut and obtained from foods have an impact on the intestinal health as well as vital organs of the host. It has been recognized that the gut is the "vital organ" in the host. As the gut microbial metabolites, SCFA could create an "axis" connecting the gut and to other organs. Therefore, the "gut-organ axes" have become a focus of research in recent years to analyze organism health. In this review, we summarized the sources, absorption properties, and the function of SCFA in both gut and other peripheral tissues (brain, kidney, liver, lung, bone and cardiovascular) in the way of "gut-organ axes". Short chain fatty acids exert both beneficial and pathological role in gut and other organs in various ways, in which the beneficial effects are more pronounced. In addition, the beneficial effects are reflected in both preventive and therapeutic effects. More importantly, the mechanisms behinds the gut and other tissues provided insight into the function of SCFA, assisting in the development of novel preventive and therapeutic strategies for maintaining the host health.
Collapse
Affiliation(s)
- Yanan Gao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qianqian Yao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Department of Food Science, Faculty of Veterinary Medicine, University of Liège, Liège 4000, Belgium
| | - Lu Meng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jiaqi Wang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
5
|
Huang Y, Wang YF, Miao J, Zheng RF, Li JY. Short-chain fatty acids: Important components of the gut-brain axis against AD. Biomed Pharmacother 2024; 175:116601. [PMID: 38749177 DOI: 10.1016/j.biopha.2024.116601] [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: 01/20/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 06/03/2024] Open
Abstract
Alzheimer's disease (AD) comprises a group of neurodegenerative disorders with some changes in the brain, which could lead to the deposition of certain proteins and result in the degeneration and death of brain cells. Patients with AD manifest primarily as cognitive decline, psychiatric symptoms, and behavioural disorders. Short-chain fatty acids (SCFAs) are a class of saturated fatty acids (SFAs) produced by gut microorganisms through the fermentation of dietary fibre ingested. SCFAs, as a significant mediator of signalling, can have diverse physiological and pathological roles in the brain through the gut-brain axis, and play a positive effect on AD via multiple pathways. Firstly, differences in SCFAs and microbial changes have been stated in AD cases of humans and mice in this paper. And then, mechanisms of three main SCFAs in treating with AD have been summarized, as well as differences of gut bacteria. Finally, functions of SCFAs played in regulating intestinal flora homeostasis, modulating the immune system, and the metabolic system, which were considered to be beneficial for the treatment of AD, have been elucidated, and the key roles of gut bacteria and SCFAs were pointed out. All in all, this paper provides an overview of SCFAs and gut bacteria in AD, and can help people to understand the importance of gut-brain axis in AD.
Collapse
Affiliation(s)
- Yan Huang
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China
| | - Yi Feng Wang
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China
| | - Jing Miao
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830004, China.
| | - Rui Fang Zheng
- Xinjiang Key Laboratory of Uygur Medical Research, Xinjiang Institute of Materia Medica, Urumqi 830004, China.
| | - Jin Yao Li
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830004, China.
| |
Collapse
|
6
|
Elford JD, Becht N, Garssen J, Kraneveld AD, Perez-Pardo P. Buty and the beast: the complex role of butyrate in Parkinson's disease. Front Pharmacol 2024; 15:1388401. [PMID: 38694925 PMCID: PMC11061429 DOI: 10.3389/fphar.2024.1388401] [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: 02/19/2024] [Accepted: 04/02/2024] [Indexed: 05/04/2024] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disease which is often associated with gastrointestinal (GI) dysfunction. The GI tract is home to a wide range of microorganisms, among which bacteria, that can influence the host through various mechanisms. Products produced by these bacteria can act in the gut but can also exert effects in the brain via what is now well established to be the microbiota-gut-brain axis. In those with PD the gut-bacteria composition is often found to be different to that of non-PD individuals. In addition to compositional changes, the metabolic activity of the gut-microbiota is also changed in PD. Specifically, it is often reported that key producers of short chain fatty acids (SCFAs) as well as the concentration of SCFAs themselves are altered in the stool and blood of those with PD. These SCFAs, among which butyrate, are essential nutrients for the host and are a major energy source for epithelial cells of the GI tract. Additionally, butyrate plays a key role in regulating various host responses particularly in relation to inflammation. Studies have demonstrated that a reduction in butyrate levels can have a critical role in the onset and progression of PD. Furthermore, it has been shown that restoring butyrate levels in those with PD through methods such as probiotics, prebiotics, sodium butyrate supplementation, and fecal transplantation can have a beneficial effect on both motor and non-motor outcomes of the disease. This review presents an overview of evidence for the altered gut-bacteria composition and corresponding metabolite production in those with PD, with a particular focus on the SCFA butyrate. In addition to presenting current studies regarding SCFA in clinical and preclinical reports, evidence for the possibility to target butyrate production using microbiome based approaches in a therapeutic context is discussed.
Collapse
Affiliation(s)
- Joshua D. Elford
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Nanette Becht
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Department of Neuroscience, Faculty of Science, Vrije Universiteit, Amsterdam, Netherlands
| | - Paula Perez-Pardo
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
7
|
Wang Z, Jiang D, Zhang M, Teng Y, Huang Y. Causal association between gut microbiota and fibromyalgia: a Mendelian randomization study. Front Microbiol 2024; 14:1305361. [PMID: 38260871 PMCID: PMC10800605 DOI: 10.3389/fmicb.2023.1305361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Background Fibromyalgia (FM) is a syndrome characterized by chronic and widespread musculoskeletal pain. A number of studies have implied a potential association between gut microbiota and FM. However, the casual association between gut microbiota and FM remains unknown. Method Mendelian randomization (MR) study was conducted using the summary statistics of genetic variants from the genome-wide association study (GWAS). Inverse variance weighted (IVW), combined with MR-Egger and weighted median were used to investigate the causal association between 119 gut microbiota genera and FM. Sensitivity analyses were performed on the MR results, including heterogeneity test, leave-one-out test and pleiotropy test. Results A total of 1,295 single nucleotide polymorphism (SNPs) were selected as instrumental variables (IVs), with no significant heterogeneity and pleiotropy according to the sensitivity analyses. Five gut microbiota genera were found to have significant casual association with FM. Coprococcus2 (OR = 2.317, p-value = 0.005, 95% CI: 1.289-4.167), Eggerthella (OR = 1.897, p-value = 0.001, 95% CI: 1.313-2.741) and Lactobacillus (OR = 1.576, p-value =0.020, 95% CI: 1.073-2.315) can increase the risk of FM. FamillyXIIIUCG001 (OR = 0.528, p-value = 0.038, 95% CI: 0.289-0.964) and Olsenella (OR = 0.747, p-value = 0.050, 95% CI: 0.557-1.000) can decrease the risk of FM. Conclusion This MR study found that gut microbiota is casually associated with FM. New insights into the mechanisms of FM mediated by gut microbiota are provided.
Collapse
Affiliation(s)
- Zhaohua Wang
- Beijing Engineering Research Center of Food Environment and Public Health, Minzu University of China, Beijing, China
- College of life and Environmental Science, Minzu University of China, Beijing, China
| | - Dan Jiang
- College of Food Science and Engineering, Dalian Ocean University, Dalian, China
| | - Min Zhang
- Beijing Engineering Research Center of Food Environment and Public Health, Minzu University of China, Beijing, China
- College of life and Environmental Science, Minzu University of China, Beijing, China
| | - Yu Teng
- Beijing Engineering Research Center of Food Environment and Public Health, Minzu University of China, Beijing, China
- College of life and Environmental Science, Minzu University of China, Beijing, China
| | - Yaojiang Huang
- Beijing Engineering Research Center of Food Environment and Public Health, Minzu University of China, Beijing, China
- College of life and Environmental Science, Minzu University of China, Beijing, China
| |
Collapse
|
8
|
Sun M, Ji W, Ye H, Cai Y, Yun Y, Wei X, Wang C, Mao H. Sodium butyrate administration improves intestinal development of suckling lambs. J Anim Sci 2024; 102:skae028. [PMID: 38285605 PMCID: PMC10889743 DOI: 10.1093/jas/skae028] [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: 11/06/2023] [Accepted: 01/25/2024] [Indexed: 01/31/2024] Open
Abstract
This study was conducted to investigate the effects of sodium butyrate (SB) supplementation on growth performance, intestinal barrier functions, and intestinal bacterial communities in sucking lambs. Forty lambs of 7 d old, with an average body weight (BW) of 4.46 ± 0.45 kg, were allocated into the control (CON) or SB group, with each group having five replicate pens (n = 5). Lambs were orally administered SB at 1.8 mL/kg BW in the SB group or the same volume of saline in the CON group. Treatments were administered from 7 to 35 d of age, when one lamb from each replicate was slaughtered to obtain intestinal tissues and contents. The results showed that supplementation with SB tended to increase the BW (P = 0.079) and the starter intake (P = 0.089) of lambs at 35 d of age. The average daily gain of lambs in the SB group was significantly greater than that in the CON group (P < 0.05). The villus height of jejunum in the SB group was markedly higher (P < 0.05) than that in the CON group. In ileum, lambs in the SB group had lower (P < 0.05) crypt depth and greater (P < 0.05) villus-to-crypt ratio than those in the CON group. Compared with the CON group, the mRNA and protein expressions of Claudin-1 and Occludin were increased (P < 0.05) in the SB group. Supplementation with SB decreased the relative abundances of pathogenic bacteria, including Clostridia_UCG-014 (P = 0.094) and Romboutsia (P < 0.05), which were negatively associated with the intestinal barrier function genes (P < 0.05). The relative abundance of Succiniclasticum (P < 0.05) was higher in the SB group, and it was positively correlated with the ratio of villi height to crypt depth in the jejunum (P < 0.05). Compared with the CON group, the function "Metabolism of Cofactors and Vitamins" was increased in the SB group lambs (P < 0.05). In conclusion, SB orally administration during suckling period could improve the small intestine development and growth performance of lambs by inhibiting the harmful bacteria (Clostridia_UCG-014, Romboutsia) colonization, and enhancing intestinal barrier functions.
Collapse
Affiliation(s)
- Mengzhen Sun
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University; Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Lin’an 311300, China
| | - Wenwen Ji
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University; Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Lin’an 311300, China
| | - Hongwei Ye
- Hangzhou Lin ‘an District Agroforestry Technology Extension Center, Lin’an 311300, China
| | - Yitao Cai
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University; Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Lin’an 311300, China
| | - Yan Yun
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University; Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Lin’an 311300, China
| | - Xiaoshi Wei
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University; Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Lin’an 311300, China
| | - Chong Wang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University; Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Lin’an 311300, China
| | - Huiling Mao
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University; Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Lin’an 311300, China
| |
Collapse
|
9
|
Guo B, Zhang J, Zhang W, Chen F, Liu B. Gut microbiota-derived short chain fatty acids act as mediators of the gut-brain axis targeting age-related neurodegenerative disorders: a narrative review. Crit Rev Food Sci Nutr 2023; 65:265-286. [PMID: 37897083 DOI: 10.1080/10408398.2023.2272769] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
Neurodegenerative diseases associated with aging are often accompanied by cognitive decline and gut microbiota disorder. But the impact of gut microbiota on these cognitive disturbances remains incompletely understood. Short chain fatty acids (SCFAs) are major metabolites produced by gut microbiota during the digestion of dietary fiber, serving as an energy source for gut epithelial cells and/or circulating to other organs, such as the liver and brain, through the bloodstream. SCFAs have been shown to cross the blood-brain barrier and played crucial roles in brain metabolism, with potential implications in mediating Alzheimer's disease (AD) and Parkinson's disease (PD). However, the underlying mechanisms that SCFAs might influence psychological functioning, including affective and cognitive processes and their neural basis, have not been fully elucidated. Furthermore, the dietary sources which determine these SCFAs production was not thoroughly evaluated yet. This comprehensive review explores the production of SCFAs by gut microbiota, their transportation through the gut-brain axis, and the potential mechanisms by which they influence age-related neurodegenerative disorders. Also, the review discusses the importance of dietary fiber sources and the challenges associated with harnessing dietary-derived SCFAs as promoters of neurological health in elderly individuals. Overall, this study suggests that gut microbiota-derived SCFAs and/or dietary fibers hold promise as potential targets and strategies for addressing age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Bingbing Guo
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Jingyi Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Weihao Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Feng Chen
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Innovative Development of Food Industry, Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Shenzhen University, Shenzhen, China
| | - Bin Liu
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Innovative Development of Food Industry, Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
10
|
Yang N, Lan T, Han Y, Zhao H, Wang C, Xu Z, Chen Z, Tao M, Li H, Song Y, Ma X. Tributyrin alleviates gut microbiota dysbiosis to repair intestinal damage in antibiotic-treated mice. PLoS One 2023; 18:e0289364. [PMID: 37523400 PMCID: PMC10389721 DOI: 10.1371/journal.pone.0289364] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 07/17/2023] [Indexed: 08/02/2023] Open
Abstract
Tributyrin (TB) is a butyric acid precursor and has a key role in anti-inflammatory and intestinal barrier repair effects by slowly releasing butyric acid. However, its roles in gut microbiota disorder caused by antibiotics remain unclear. Herein, we established an intestinal microbiota disorder model using ceftriaxone sodium via gavage to investigate the effects of different TB doses for restoring gut microbiota and intestinal injury. First, we divided C57BL/6 male mice into two groups: control (NC, n = 8) and experimental (ABx, n = 24) groups, receiving gavage with 0.2 mL normal saline and 400 mg/mL ceftriaxone sodium solution for 7 d (twice a day and the intermediate interval was 6 h), respectively. Then, mice in the ABx group were randomly split into three groups: model (M, 0.2 mL normal saline), low TB group (TL, 0.3 g/kg BW), and high TB group (TH, 3 g/kg BW) for 11 d. We found that TB supplementation alleviated antibiotics-induced weight loss, diarrhea, and intestinal tissue damage. The 16S rRNA sequence analysis showed that TB intervention increased the α diversity of intestinal flora, increased potential short-chain fatty acids (SCFAs)-producing bacteria (such as Muribaculaceae and Bifidobacterium), and inhibited the relative abundance of potentially pathogenic bacteria (such as Bacteroidetes and Enterococcus) compared to the M group. TB supplementation reversed the reduction in SCFAs production in antibiotic-treated mice. Additionally, TB downregulated the levels of serum LPS and zonulin, TNF-α, IL-6, IL-1β and NLRP3 inflammasome-related factors in intestinal tissue and upregulated tight junction proteins (such as ZO-1 and Occludin) and MUC2. Overall, the adjustment ability of low-dose TB to the above indexes was stronger than high-dose TB. In conclusion, TB can restore the dysbiosis of gut microbiota, increase SCFAs, suppress inflammation, and ameliorate antibiotic-induced intestinal damage, indicating that TB might be a potential gut microbiota modulator.
Collapse
Affiliation(s)
- Ning Yang
- Department of Nutrition and Food Hygiene, School of Public Health, College of Medicine, Qingdao University, Qingdao, China
| | - Tongtong Lan
- Department of Nutrition and Food Hygiene, School of Public Health, College of Medicine, Qingdao University, Qingdao, China
| | - Yisa Han
- Department of Nutrition and Food Hygiene, School of Public Health, College of Medicine, Qingdao University, Qingdao, China
| | - Haifeng Zhao
- Qingdao Institute of Food and Drug Control, Key Laboratory of Quality Research and Evaluation of Marine Traditional Chinese Medicine, State Medical Products Administration, Qingdao, China
| | - Chuhui Wang
- Department of Nutrition and Food Hygiene, School of Public Health, College of Medicine, Qingdao University, Qingdao, China
| | - Zhen Xu
- Department of Nutrition and Food Hygiene, School of Public Health, College of Medicine, Qingdao University, Qingdao, China
| | - Zhao Chen
- Department of Nutrition and Food Hygiene, School of Public Health, College of Medicine, Qingdao University, Qingdao, China
| | - Meng Tao
- Department of Nutrition and Food Hygiene, School of Public Health, College of Medicine, Qingdao University, Qingdao, China
| | - Hui Li
- Department of Nutrition and Food Hygiene, School of Public Health, College of Medicine, Qingdao University, Qingdao, China
| | - Yang Song
- Department of Nutrition and Food Hygiene, School of Public Health, College of Medicine, Qingdao University, Qingdao, China
| | - Xuezhen Ma
- The Affiliated Qingdao Central Hospital of Qingdao University, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, China
| |
Collapse
|
11
|
Garofalo C, Cristiani CM, Ilari S, Passacatini LC, Malafoglia V, Viglietto G, Maiuolo J, Oppedisano F, Palma E, Tomino C, Raffaeli W, Mollace V, Muscoli C. Fibromyalgia and Irritable Bowel Syndrome Interaction: A Possible Role for Gut Microbiota and Gut-Brain Axis. Biomedicines 2023; 11:1701. [PMID: 37371796 DOI: 10.3390/biomedicines11061701] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Fibromyalgia (FM) is a serious chronic pain syndrome, characterised by muscle and joint stiffness, insomnia, fatigue, mood disorders, cognitive dysfunction, anxiety, depression and intestinal irritability. Irritable Bowel Syndrome (IBS) shares many of these symptoms, and FM and IBS frequently co-exist, which suggests a common aetiology for the two diseases. The exact physiopathological mechanisms underlying both FM and IBS onset are unknown. Researchers have investigated many possible causes, including alterations in gut microbiota, which contain billions of microorganisms in the human digestive tract. The gut-brain axis has been proven to be the link between the gut microbiota and the central nervous system, which can then control the gut microbiota composition. In this review, we will discuss the similarities between FM and IBS. Particularly, we will focus our attention on symptomatology overlap between FM and IBS as well as the similarities in microbiota composition between FM and IBS patients. We will also briefly discuss the potential therapeutic approaches based on microbiota manipulations that are successfully used in IBS and could be employed also in FM patients to relieve pain, ameliorate the rehabilitation outcome, psychological distress and intestinal symptoms.
Collapse
Affiliation(s)
- Cinzia Garofalo
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Costanza Maria Cristiani
- Department of Medical and Surgical Sciences, Neuroscience Research Center, "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Sara Ilari
- Physiology and Pharmacology of Pain, IRCCS San Raffaele Roma, 00166 Rome, Italy
| | - Lucia Carmela Passacatini
- Department of Health Science, Institute of Research for Food Safety & Health (IRC-FSH), "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | | | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Jessica Maiuolo
- Department of Health Science, Institute of Research for Food Safety & Health (IRC-FSH), "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Francesca Oppedisano
- Department of Health Science, Institute of Research for Food Safety & Health (IRC-FSH), "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Ernesto Palma
- Department of Health Science, Institute of Research for Food Safety & Health (IRC-FSH), "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Carlo Tomino
- Scientific Direction, IRCCS San Raffaele Roma, 00166 Rome, Italy
| | - William Raffaeli
- Institute for Research on Pain, ISAL Foundation, Torre Pedrera, 47922 Rimini, Italy
| | - Vincenzo Mollace
- Department of Health Science, Institute of Research for Food Safety & Health (IRC-FSH), "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Carolina Muscoli
- Department of Health Science, Institute of Research for Food Safety & Health (IRC-FSH), "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
12
|
Rahman MM, Islam F, -Or-Rashid MH, Mamun AA, Rahaman MS, Islam MM, Meem AFK, Sutradhar PR, Mitra S, Mimi AA, Emran TB, Fatimawali, Idroes R, Tallei TE, Ahmed M, Cavalu S. The Gut Microbiota (Microbiome) in Cardiovascular Disease and Its Therapeutic Regulation. Front Cell Infect Microbiol 2022; 12:903570. [PMID: 35795187 PMCID: PMC9251340 DOI: 10.3389/fcimb.2022.903570] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/09/2022] [Indexed: 12/11/2022] Open
Abstract
In the last two decades, considerable interest has been shown in understanding the development of the gut microbiota and its internal and external effects on the intestine, as well as the risk factors for cardiovascular diseases (CVDs) such as metabolic syndrome. The intestinal microbiota plays a pivotal role in human health and disease. Recent studies revealed that the gut microbiota can affect the host body. CVDs are a leading cause of morbidity and mortality, and patients favor death over chronic kidney disease. For the function of gut microbiota in the host, molecules have to penetrate the intestinal epithelium or the surface cells of the host. Gut microbiota can utilize trimethylamine, N-oxide, short-chain fatty acids, and primary and secondary bile acid pathways. By affecting these living cells, the gut microbiota can cause heart failure, atherosclerosis, hypertension, myocardial fibrosis, myocardial infarction, and coronary artery disease. Previous studies of the gut microbiota and its relation to stroke pathogenesis and its consequences can provide new therapeutic prospects. This review highlights the interplay between the microbiota and its metabolites and addresses related interventions for the treatment of CVDs.
Collapse
|
13
|
Amoah K, Dong XH, Tan BP, Zhang S, Chi SY, Yang QH, Liu HY, Yan XB, Yang YZ, Zhang H. Ultra-Performance Liquid Chromatography-Mass Spectrometry-Based Untargeted Metabolomics Reveals the Key Potential Biomarkers for Castor Meal-Induced Enteritis in Juvenile Hybrid Grouper (Epinephelus fuscoguttatus♀ × E. lanceolatus♂). Front Nutr 2022; 9:847425. [PMID: 35811940 PMCID: PMC9261911 DOI: 10.3389/fnut.2022.847425] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 05/03/2022] [Indexed: 12/18/2022] Open
Abstract
The intensification of aquaculture to help kerb global food security issues has led to the quest for more economical new protein-rich ingredients for the feed-based aquaculture since fishmeal (FM, the ingredient with the finest protein and lipid profile) is losing its acceptability due to high cost and demand. Although very high in protein, castor meal (CM), a by-product after oil-extraction, is disposed-off due to the high presence of toxins. Concurrently, the agro-industrial wastes’ consistent production and disposal are of utmost concern; however, having better nutritional profiles of these wastes can lead to their adoption. This study was conducted to identify potential biomarkers of CM-induced enteritis in juvenile hybrid-grouper (Epinephelus fuscoguttatus♀ × Epinephelus lanceolatus♂) using ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) alongside their growth and distal intestinal (DI) health evaluation. A total of 360 fish (initial weight = 9.13 ± 0.01g) were randomly assigned into three groups, namely, fish-meal (FM) (control), 4% CM (CM4), and 20% CM (CM20). After the 56-days feeding-trial, the DI tissues of FM, CM4, and CM20 groups were collected for metabolomics analysis. Principal components analysis and partial least-squares discriminant-analysis (PLS-DA, used to differentiate the CM20 and CM4, from the FM group with satisfactory explanation and predictive ability) were used to analyze the UPLC-MS data. The results revealed a significant improvement in the growth, DI immune responses and digestive enzyme activities, and DI histological examinations in the CM4 group than the others. Nonetheless, CM20 replacement caused DI physiological damage and enteritis in grouper as shown by AB-PAS staining and scanning electron microscopy examinations, respectively. The most influential metabolites in DI contents identified as the potential biomarkers in the positive and negative modes using the metabolomics UPLC-MS profiles were 28 which included five organoheterocyclic compounds, seven lipids, and lipid-like molecules, seven organic oxygen compounds, two benzenoids, five organic acids and derivatives, one phenylpropanoids and polyketides, and one from nucleosides, nucleotides, and analogues superclass. The present study identified a broad array of DI tissue metabolites that differed between FM and CM diets, which provides a valuable reference for further managing fish intestinal health issues. A replacement level of 4% is recommended based on the growth and immunity of fish.
Collapse
Affiliation(s)
- Kwaku Amoah
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Zhanjiang, China
| | - Xiao-hui Dong
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Zhanjiang, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China
- *Correspondence: Xiao-hui Dong,
| | - Bei-ping Tan
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Zhanjiang, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China
| | - Shuang Zhang
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Zhanjiang, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China
| | - Shu-yan Chi
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Zhanjiang, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China
| | - Qi-hui Yang
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Zhanjiang, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China
| | - Hong-yu Liu
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Zhanjiang, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China
| | - Xiao-bo Yan
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Zhanjiang, China
| | - Yuan-zhi Yang
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Haitao Zhang
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China
| |
Collapse
|
14
|
Fortea M, Albert-Bayo M, Abril-Gil M, Ganda Mall JP, Serra-Ruiz X, Henao-Paez A, Expósito E, González-Castro AM, Guagnozzi D, Lobo B, Alonso-Cotoner C, Santos J. Present and Future Therapeutic Approaches to Barrier Dysfunction. Front Nutr 2021; 8:718093. [PMID: 34778332 PMCID: PMC8582318 DOI: 10.3389/fnut.2021.718093] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
There is converging and increasing evidence, but also uncertainty, for the role of abnormal intestinal epithelial barrier function in the origin and development of a growing number of human gastrointestinal and extraintestinal inflammatory disorders, and their related complaints. Despite a vast literature addressing factors and mechanisms underlying changes in intestinal permeability in humans, and its connection to the appearance and severity of clinical symptoms, the ultimate link remains to be established in many cases. Accordingly, there are no directives or clinical guidelines related to the therapeutic management of intestinal permeability disorders that allow health professionals involved in the management of these patients to carry out a consensus treatment based on clinical evidence. Instead, there are multiple pseudoscientific approaches and commercial propaganda scattered on the internet that confuse those affected and health professionals and that often lack scientific rigor. Therefore, in this review we aim to shed light on the different therapeutic options, which include, among others, dietary management, nutraceuticals and medical devices, microbiota and drugs, and epigenetic and exosomes-manipulation, through an objective evaluation of the scientific publications in this field. Advances in the knowledge and management of intestinal permeability will sure enable better options of dealing with this group of common disorders to enhance quality of life of those affected.
Collapse
Affiliation(s)
- Marina Fortea
- Laboratory for Enteric NeuroScience, Translational Research Center for GastroIntestinal Disorders, University of Leuven, Leuven, Belgium
| | - Mercé Albert-Bayo
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Mar Abril-Gil
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - John-Peter Ganda Mall
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Xavier Serra-Ruiz
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Alejandro Henao-Paez
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Elba Expósito
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Ana María González-Castro
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Danila Guagnozzi
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Facultad de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERHED), Instituto de Salud Carlos III, Madrid, Spain
| | - Beatriz Lobo
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Facultad de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Carmen Alonso-Cotoner
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Facultad de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERHED), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Santos
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Facultad de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERHED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
15
|
Chang MC, Wang TM, Chien HH, Pan YH, Tsai YL, Jeng PY, Lin LD, Jeng JH. Effect of butyrate, a bacterial by-product, on the viability and ICAM-1 expression/production of human vascular endothelial cells: Role in infectious pulpal/periapical diseases. Int Endod J 2021; 55:38-53. [PMID: 34420220 DOI: 10.1111/iej.13614] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 08/19/2021] [Indexed: 01/21/2023]
Abstract
AIM To investigate the effects of butyric acid (BA), a metabolic product generated by pulp and root canal pathogens, on the viability and intercellular adhesion molecule-1 (ICAM-1) production of endothelial cells, which are crucial to angiogenesis and pulpal/periapical wound healing. METHODOLOGY Endothelial cells were exposed to butyrate with/without inhibitors. Cell viability, apoptosis and reactive oxygen species (ROS) were evaluated using an MTT assay, PI/annexin V and DCF fluorescence flow cytometry respectively. RNA and protein expression was determined using a polymerase chain reaction assay and Western blotting or immunofluorescent staining. Soluble ICAM-1 (sICAM-1) was measured using an enzyme-linked immunosorbent assay. The quantitative results were expressed as mean ± standard error (SE) of the mean. The data were analysed using a paired Student's t-test where necessary. A p-value ≤0.05 was considered to indicate a statistically significant difference between groups. RESULTS Butyrate (>4 mM) inhibited cell viability and induced cellular apoptosis and necrosis. It inhibited cyclin B1 but stimulated p21 and p27 expression. Butyrate stimulated ROS production and hemeoxygenase-1 (HO-1) expression as well as activated the Ac-H3, p-ATM, p-ATR, p-Chk1, p-Chk2, p-p38 and p-Akt expression of endothelial cells. Butyrate stimulated ICAM-1 mRNA/protein expression and significant sICAM-1 production (p < .05). Superoxide dismutase, 5z-7oxozeaenol, SB203580 and compound C (p < .05), but not ZnPP, CGK733, AZD7762 or LY294002, attenuated butyrate cytotoxicity to endothelial cells. Notably, little effect on butyrate-stimulated sICAM-1 secretion was found. Valproic acid, phenylbutyrate and trichostatin (three histone deacetylase inhibitors) significantly induced sICAM-1 production (p < .05). CONCLUSION Butyric acid inhibited proliferation, induced apoptosis, stimulated ROS and HO-1 production and increased ICAM-1 mRNA expression and protein synthesis in endothelial cells. Cell viability affected by BA was diminished by some inhibitors; however, the increased sICAM-1 secretion by BA was not affected by any of the tested inhibitors. These results facilitate understanding of the pathogenesis, prevention and treatment of pulpal/periapical diseases.
Collapse
Affiliation(s)
- Mei-Chi Chang
- Chang Gung University of Science and Technology, Taoyuan City, Taiwan.,Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Tong-Mei Wang
- School of Dentistry & Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Hua-Hong Chien
- Division of Periodontology, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - Yu-Hwa Pan
- Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Yi-Ling Tsai
- School of Dentistry & Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Yuan Jeng
- School of Dentistry & Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Deh Lin
- School of Dentistry & Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Jiiang-Huei Jeng
- School of Dentistry & Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan.,School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung City, Taiwan
| |
Collapse
|
16
|
Bredeck G, Kämpfer AAM, Sofranko A, Wahle T, Lison D, Ambroise J, Stahlmecke B, Albrecht C, Schins RPF. Effects of dietary exposure to the engineered nanomaterials CeO 2, SiO 2, Ag, and TiO 2 on the murine gut microbiome. Nanotoxicology 2021; 15:934-950. [PMID: 34380002 DOI: 10.1080/17435390.2021.1940339] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Rodent studies on the effects of engineered nanomaterials (ENM) on the gut microbiome have revealed contradictory results. Our aim was to assess the effects of four well-investigated model ENM using a realistic exposure scenario. Two independent ad libitum feeding studies were performed. In study 1, female mice from the local breeding facility received feed pellets containing 1% CeO2 or 1% SiO2 for three weeks. In study 2, both female and male mice were purchased and exposed to 0.2% Ag-PVP or 1% TiO2 for four weeks. A next generation 16S rDNA sequencing-based approach was applied to assess impacts on the gut microbiome. None of the ENM had an effect on the α- or β-diversity. A decreased relative abundance of the phylum Actinobacteria was observed in SiO2 exposed mice. In female mice, the relative abundance of the genus Roseburia was increased with Ag exposure. Furthermore, in study 2, a sex-related difference in the β-diversity was observed. A difference in the β-diversity was also shown between the female control mice of the two studies. We did not find major effects on the gut microbiome. This contrast to other studies may be due to variations in the study design. Our investigation underlined the important role of the sex of test animals and their microbiome composition prior to ENM exposure initiation. Hence, standardization of microbiome studies is strongly required to increase comparability. The ENM-specific effects on Actinobacteria and Roseburia, two taxa pivotal for the human gut homeostasis, warrant further research on their relevance for health.
Collapse
Affiliation(s)
- Gerrit Bredeck
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Angela A M Kämpfer
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Adriana Sofranko
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Tina Wahle
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Dominique Lison
- Louvain Centre for Toxicology and Applied Pharmacology, Université Catholique de Louvain, Brussels, Belgium
| | - Jérôme Ambroise
- Centre de Technologies Moléculaires Appliquées, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Burkhard Stahlmecke
- Institute for Energy and Environmental Technology e.V. (IUTA), Duisburg, Germany
| | - Catrin Albrecht
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Roel P F Schins
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| |
Collapse
|
17
|
Liu W, Luo X, Tang J, Mo Q, Zhong H, Zhang H, Feng F. A bridge for short-chain fatty acids to affect inflammatory bowel disease, type 1 diabetes, and non-alcoholic fatty liver disease positively: by changing gut barrier. Eur J Nutr 2021; 60:2317-2330. [PMID: 33180143 DOI: 10.1007/s00394-020-02431-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE In previous studies, short-chain fatty acids (SCFAs) have been found to regulate gut microbiota and change gut barrier status, and the potential positive effects of SCFAs on inflammatory bowel disease (IBD), type 1 diabetes mellitus (T1D), and non-alcoholic fatty liver disease (NAFLD) have also been found, but the role of SCFAs in these three diseases is not clear. This review aims to summarize existing evidence on the effects of SCFAs on IBD, T1D, and NHFLD, and correlates them with gut barrier and gut microbiota (gut microbiota barrier). METHODS A literature search in PubMed, Web of Science, Springer, and Wiley Online Library up to October 2020 was conducted for all relevant studies published. RESULTS This is a retrospective review of 150 applied research articles or reviews. The destruction of gut barrier may promote the development of IBD, T1D, and NAFLD. SCFAs seem to maintain the gut barrier by promoting the growth of intestinal epithelial cells, strengthening the intestinal tight connection, and regulating the activities of gut microbiota and immune cells, which might result possible beneficial effects on the above three diseases at a certain dose. CONCLUSIONS Influencing gut barrier health may be a bridge for SCFAs (especially butyrate) to have positive effects on IBD, T1D, and NAFLD. It is expected that this article can provide new ideas for the subsequent research on the treatment of diseases by SCFAs and help SCFAs be better applied to precise and personalized treatment.
Collapse
Affiliation(s)
- Wangxin Liu
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture, Zhejiang University, No. 866, Yuhangtang Road, Hangzhou, 310058, China
| | - Xianliang Luo
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture, Zhejiang University, No. 866, Yuhangtang Road, Hangzhou, 310058, China
| | - Jun Tang
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture, Zhejiang University, No. 866, Yuhangtang Road, Hangzhou, 310058, China
| | - Qiufen Mo
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture, Zhejiang University, No. 866, Yuhangtang Road, Hangzhou, 310058, China
| | - Hao Zhong
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture, Zhejiang University, No. 866, Yuhangtang Road, Hangzhou, 310058, China
| | - Hui Zhang
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture, Zhejiang University, No. 866, Yuhangtang Road, Hangzhou, 310058, China
| | - Fengqin Feng
- College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture, Zhejiang University, No. 866, Yuhangtang Road, Hangzhou, 310058, China.
| |
Collapse
|
18
|
Climent E, Martinez-Blanch JF, Llobregat L, Ruzafa-Costas B, Carrión-Gutiérrez MÁ, Ramírez-Boscá A, Prieto-Merino D, Genovés S, Codoñer FM, Ramón D, Chenoll E, Navarro-López V. Changes in Gut Microbiota Correlates with Response to Treatment with Probiotics in Patients with Atopic Dermatitis. A Post Hoc Analysis of a Clinical Trial. Microorganisms 2021; 9:microorganisms9040854. [PMID: 33921166 PMCID: PMC8071520 DOI: 10.3390/microorganisms9040854] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic recurrent inflammatory skin disease with a high impact on the comfort of those who are affected and long-term treated with corticosteroids with limited efficacy and a high prevalence of relapses. Because of the limited effectiveness of these treatments, new strategies for recovery from AD lesions are continually being explored. In this article, we describe the gut microbiome changes achieved in a recently published clinical trial with the probiotic formulation Bifidobacterium animalis subsp. lactis CECT 8145, Bifidobacterium longum CECT 7347, and Lacticaseibacillus casei CECT 9104 (formerly Lactobacillus casei CECT 9104), showing a significant improvement in SCORAD (scoring atopic dermatitis) index in children (4-17 years) with AD (Clinicaltrials.gov identifier: NCT02585986). The present gut microbiome post hoc study showed no significant changes in diversity (Shannon and Simpson indexes) after probiotic consumption. In the probiotic group, genera Bacteroides, Ruminococcus, and Bifidobacterium significantly increased their levels while Faecalibacterium decreased, compared to the placebo group. Faecalibacterium showed the highest presence and significant positive correlation with AD severity (SCORAD index), whereas Abyssivirga, Bifidobacterium, and Lactococcus were inversely correlated. The results suggest that the consumption of the probiotic formulation here assayed modulates the gut microbiome with significant changes in genera Bacteroides and Faecalibacterium. In turn, the improvement in SCORAD correlates with a decrease in Faecalibacterium and an increase in Bifidobacterium, among others.
Collapse
Affiliation(s)
- Eric Climent
- Biopolis S.L.-ADM, Catedrático Agustín Escardino Benlloch 9 Edif. 2, 46980 Paterna, Spain; (E.C.); (J.F.M.-B.); (L.L.); (S.G.); (F.M.C.); (D.R.)
| | - Juan Francisco Martinez-Blanch
- Biopolis S.L.-ADM, Catedrático Agustín Escardino Benlloch 9 Edif. 2, 46980 Paterna, Spain; (E.C.); (J.F.M.-B.); (L.L.); (S.G.); (F.M.C.); (D.R.)
| | - Laura Llobregat
- Biopolis S.L.-ADM, Catedrático Agustín Escardino Benlloch 9 Edif. 2, 46980 Paterna, Spain; (E.C.); (J.F.M.-B.); (L.L.); (S.G.); (F.M.C.); (D.R.)
| | - Beatriz Ruzafa-Costas
- Department of Clinical Medicine, Universidad Católica San Antonio de Murcia (UCAM), 30107 Murcia, Spain; (B.R.-C.); (A.R.-B.); (V.N.-L.)
| | | | - Ana Ramírez-Boscá
- Department of Clinical Medicine, Universidad Católica San Antonio de Murcia (UCAM), 30107 Murcia, Spain; (B.R.-C.); (A.R.-B.); (V.N.-L.)
- Department of Dermatology, Hospital Universitario Vinalopó, 03293 Elche, Spain
| | - David Prieto-Merino
- Applied Statistical Methods in Medical Research Group, Universidad Católica San Antonio de Murcia (UCAM), 30107 Murcia, Spain;
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London 400706, UK
| | - Salvador Genovés
- Biopolis S.L.-ADM, Catedrático Agustín Escardino Benlloch 9 Edif. 2, 46980 Paterna, Spain; (E.C.); (J.F.M.-B.); (L.L.); (S.G.); (F.M.C.); (D.R.)
| | - Francisco M. Codoñer
- Biopolis S.L.-ADM, Catedrático Agustín Escardino Benlloch 9 Edif. 2, 46980 Paterna, Spain; (E.C.); (J.F.M.-B.); (L.L.); (S.G.); (F.M.C.); (D.R.)
| | - Daniel Ramón
- Biopolis S.L.-ADM, Catedrático Agustín Escardino Benlloch 9 Edif. 2, 46980 Paterna, Spain; (E.C.); (J.F.M.-B.); (L.L.); (S.G.); (F.M.C.); (D.R.)
| | - Empar Chenoll
- Biopolis S.L.-ADM, Catedrático Agustín Escardino Benlloch 9 Edif. 2, 46980 Paterna, Spain; (E.C.); (J.F.M.-B.); (L.L.); (S.G.); (F.M.C.); (D.R.)
- Correspondence:
| | - Vicente Navarro-López
- Department of Clinical Medicine, Universidad Católica San Antonio de Murcia (UCAM), 30107 Murcia, Spain; (B.R.-C.); (A.R.-B.); (V.N.-L.)
- Clinical Microbiology and Infectious Disease Unit, Hospital Universitario Vinalopó, 03293 Elche, Spain
| |
Collapse
|
19
|
Shi Z, Fang ZY, Gao XX, Yu H, Zhu YW, Ouyang HL, Song YX, Du XL, Wang Z, Li XW, Liu GW. Nuciferine improves high-fat diet-induced obesity via reducing intestinal permeability by increasing autophagy and remodeling the gut microbiota. Food Funct 2021; 12:5850-5861. [PMID: 34018523 DOI: 10.1039/d1fo00367d] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Nuciferine (NF) has received extensive attention due to its medicinal value in the treatment of metabolic diseases, such as obesity; however, to date, the effects of NF on obesity-related intestinal permeability, autophagy and the gut microbiota have not been investigated. Herein, C57BL/6J mice were fed either a chow or a high-fat diet (HFD) with or without NF for 8 weeks. The results showed that NF supplement reduced weight gain, fat accumulation and intestinal permeability in the HFD mice accompanied by improved autophagy. Subsequently, an in vitro experiment was performed using Caco-2 and HT-29 cells, which showed that NF supplement not only promoted the formation of autophagosomes and autophagolysosomes, but also alleviated LPS-increased intestinal permeability. Importantly, NF supplement protected from LPS-induced paracellular permeability impairment after the administration of autophagy-related gene (Atg) 5 small-interfering RNA (siRNA). These results demonstrate that NF exerts beneficial effects on the intestinal permeability by improving autophagy. Furthermore, we also found that NF supplement lowered the abundance of Butyricimonas and increased the abundance of Akkermansia, an anti-obesity bacterium. Thus, overall, we demonstrated that NF supplement confers reduced intestinal permeability by improving autophagy and alters the composition of the gut microbiota in HFD-fed mice, thereby producing an anti-obesity effect.
Collapse
Affiliation(s)
- Zhen Shi
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130000, China.
| | - Zhi-Yuan Fang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130000, China.
| | - Xin-Xing Gao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130000, China.
| | - Hao Yu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130000, China.
| | - Yi-Wei Zhu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130000, China.
| | - Hui-Lin Ouyang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130000, China.
| | - Yu-Xiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130000, China.
| | - Xi-Liang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130000, China.
| | - Zhe Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130000, China.
| | - Xin-Wei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130000, China.
| | - Guo-Wen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130000, China.
| |
Collapse
|
20
|
Autoimmune Hepatitis: Shifts in Gut Microbiota and Metabolic Pathways among Egyptian Patients. Microorganisms 2020; 8:microorganisms8071011. [PMID: 32640728 PMCID: PMC7409351 DOI: 10.3390/microorganisms8071011] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
Autoimmune hepatitis (AIH) is a chronic inflammatory disorder with complex immunopathogenesis. Dysbiosis has been linked to many autoimmune diseases, but its detailed role in autoimmune hepatitis (AIH) still needs rigorous evaluation, especially in Egypt. We aimed to identify the shift in the gut microbiota profile and resultant metabolic pathways in AIH Egyptian patients compared to healthy individuals. Stool samples were collected from 15 AIH-naive patients and from 10 healthy individuals. The V3-V4 hyper-variable regions in16S rRNA gene was amplified and sequenced using Illumina MiSeq platform. Significantly lower bacterial diversity in AIH patients was found compared to the controls. A phylum-level analysis showed the overrepresentation of Firmicutes, Bacteroides, and Proteobacteria. At the genus level, AIH-associated enrichment of Faecalibacterium, Blautia, Streptococcus, Haemophilus, Bacteroides, Veillonella, Eubacterium, Lachnospiraceae and Butyricicoccus was reported in contrast to Prevotella, Parabacteroides and Dilaster, which were significantly retracted in such patients. Overall, the predicted metabolic pathways associated with dysbiosis in AIH patients could orchestrate the potential pathogenic roles of gut microbiota in autoimmune disease, though not in a disease-specific manner, calling for future large-scale studies.
Collapse
|
21
|
Tsvetikova SA, Koshel EI. Microbiota and cancer: host cellular mechanisms activated by gut microbial metabolites. Int J Med Microbiol 2020; 310:151425. [PMID: 32423739 DOI: 10.1016/j.ijmm.2020.151425] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 03/25/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022] Open
|
22
|
Abstract
Purpose of this Review This review assesses the latest evidence linking short-chain fatty acids (SCFA) with host metabolic health and cardiovascular disease (CVD) risk and presents the latest evidence on possible biological mechanisms. Recent Findings SCFA have a range of effects locally in the gut and at both splanchnic and peripheral tissues which together appear to induce improved metabolic regulation and have direct and indirect effects on markers of CVD risk. Summary SCFA produced primarily from the microbial fermentation of dietary fibre appear to be key mediators of the beneficial effects elicited by the gut microbiome. Not only does dietary fibre fermentation regulate microbial activity in the gut, SCFA also directly modulate host health through a range of tissue-specific mechanisms related to gut barrier function, glucose homeostasis, immunomodulation, appetite regulation and obesity. With the increasing burden of obesity worldwide, the role for gut microbiota-generated SCFA in protecting against the effects of energy dense diets offers an intriguing new avenue for regulating metabolic health and CVD risk.
Collapse
|
23
|
Song J, Li Q, Li P, Liu R, Cui H, Zheng M, Everaert N, Zhao G, Wen J. The effects of inulin on the mucosal morphology and immune status of specific pathogen-free chickens. Poult Sci 2018; 97:3938-3946. [PMID: 29992311 DOI: 10.3382/ps/pey260] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 05/27/2018] [Indexed: 12/18/2022] Open
Abstract
This study investigated the effects of inulin on mucosal morphology and immune function of specific pathogen-free (SPF) chickens. A total of 200 one-day-old White Leghorns SPF chickens were divided into 5 groups of 4 replicates of 10 chickens each. All SPF chickens were fed a basal diet supplemented with 0, 0.25, 0.5, 1.0, or 2.0% inulin. The mucosal morphology and immune indexes were analyzed on days 7, 14, and 21, respectively. Our results showed that the concentrations of acetate and propionate in the cecum and serum had increased with dietary inulin supplementation on day 21 (P < 0.05). Butyrate could not be detected in the cecal digesta, but was increased in the serum of 1 and 2% groups, as compared with the control group (P < 0.05). The villi height was increased (P < 0.05) and the crypt depth was decreased (P < 0.05) in the duodenum and ileum of SPF chickens fed inulin, as compared with the control group. Also, inulin at a low concentration (0.25 or 0.5%) significantly decreased (P < 0.05) the gene expression of nuclear factor-κB (NF-κB), lipopolysaccharide-induced tumor necrosis factor (LITAF) at 7, 14, and 21 d, and of interleukin-6 (IL-6), and inducible nitric oxide synthase (iNOS) at 7 and 14 d, and increased that of mucin 2 (MUC2) and claudin-1 in the ileum of SPF chickens at 7, 14, and 21 d. High inulin supplementation (2%) significantly increased the gene expression of NF-κB, LITAF, IL-6, iNOS, and Claudin-1 at 14 and 21 d compared to low inulin concentration (0.25 or 0.5%). The results indicated that the effects of inulin on mucosal immune function occurred in a dose-dependent manner. A low concentration (0.25 or 0.5%) of inulin may be beneficial in promoting intestinal immune function.
Collapse
Affiliation(s)
- Jiao Song
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, State Key Laboratory of Animal Nutrition, Beijing 100193, People's Republic of China
| | - Qinghe Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, State Key Laboratory of Animal Nutrition, Beijing 100193, People's Republic of China
| | - Peng Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, State Key Laboratory of Animal Nutrition, Beijing 100193, People's Republic of China
| | - RanRan Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, State Key Laboratory of Animal Nutrition, Beijing 100193, People's Republic of China
| | - Huanxian Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, State Key Laboratory of Animal Nutrition, Beijing 100193, People's Republic of China
| | - Maiqing Zheng
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, State Key Laboratory of Animal Nutrition, Beijing 100193, People's Republic of China
| | - Nadia Everaert
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux 5030, Belgium
| | - Guiping Zhao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, State Key Laboratory of Animal Nutrition, Beijing 100193, People's Republic of China
| | - Jie Wen
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, State Key Laboratory of Animal Nutrition, Beijing 100193, People's Republic of China
| |
Collapse
|
24
|
Liu H, Wang J, He T, Becker S, Zhang G, Li D, Ma X. Butyrate: A Double-Edged Sword for Health? Adv Nutr 2018; 9:21-29. [PMID: 29438462 PMCID: PMC6333934 DOI: 10.1093/advances/nmx009] [Citation(s) in RCA: 640] [Impact Index Per Article: 91.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 11/11/2017] [Indexed: 02/06/2023] Open
Abstract
Butyrate, a four-carbon short-chain fatty acid, is produced through microbial fermentation of dietary fibers in the lower intestinal tract. Endogenous butyrate production, delivery, and absorption by colonocytes have been well documented. Butyrate exerts its functions by acting as a histone deacetylase (HDAC) inhibitor or signaling through several G protein-coupled receptors (GPCRs). Recently, butyrate has received particular attention for its beneficial effects on intestinal homeostasis and energy metabolism. With anti-inflammatory properties, butyrate enhances intestinal barrier function and mucosal immunity. However, the role of butyrate in obesity remains controversial. Growing evidence has highlighted the impact of butyrate on the gut-brain axis. In this review, we summarize the present knowledge on the properties of butyrate, especially its potential effects and mechanisms involved in intestinal health and obesity.
Collapse
Affiliation(s)
- Hu Liu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ji Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ting He
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Sage Becker
- Department of Animal Science, Oklahoma State University, Stillwater, OK; Departments of Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, OK; Departments of Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Defa Li
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX,Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX,Address correspondence to XM (e-mail: )
| |
Collapse
|
25
|
Dun Y, Liu M, Chen J, Peng D, Zhao H, Zhou Z, Wang T, Liu C, Guo Y, Zhang C, Yuan D. Regulatory effects of saponins from Panax japonicus on colonic epithelial tight junctions in aging rats. J Ginseng Res 2016; 42:50-56. [PMID: 29348722 PMCID: PMC5766693 DOI: 10.1016/j.jgr.2016.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 10/27/2016] [Accepted: 12/20/2016] [Indexed: 12/24/2022] Open
Abstract
Background Saponins from Panax japonicus (SPJ) are the most abundant and main active components of P. japonicus, which replaces ginseng roots in treatment for many kinds of diseases in the minority ethnic group in China. Our previous studies have demonstrated that SPJ has the effects of anti-inflammation through the mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) signaling pathways. The present study was designed to investigate whether SPJ can modulate intestinal tight junction barrier in aging rats and further to explore the potential mechanism. Methods Aging rats had been treated with different doses (10 mg/kg, 30 mg/kg, and 60 mg/kg) of SPJ for 6 mo since they were 18 mo old. After the rats were euthanized, the colonic samples were harvested. Levels of tight junctions (claudin-1 and occludin) were determined by immunohistochemical staining. Levels of proinflammatory cytokines (interleukin-1β and tumor necrosis factor-α) were examined by Western blot. NF-κB and phosphorylation of MAPK signaling pathways were also determined by Western blot. Results We found that SPJ increased the expression of the tight junction proteins claudin-1 and occludin in the colon of aging rats. Treatment with SPJ decreased the levels of interleukin-1β and tumor necrosis factor-α, reduced the phosphorylation of three MAPK isoforms, and inhibited the expression of NF-κB in the colon of aging rats. Conclusion The studies demonstrated that SPJ modulates the damage of intestinal epithelial tight junction in aging rats, inhibits inflammation, and downregulates the phosphorylation of the MAPK and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Yaoyan Dun
- Medical College of China Three Gorges University, Yichang, China
| | - Min Liu
- Medical College of China Three Gorges University, Yichang, China
| | - Jing Chen
- Medical College of China Three Gorges University, Yichang, China
| | - Danli Peng
- Medical College of China Three Gorges University, Yichang, China
| | - Haixia Zhao
- Medical College of China Three Gorges University, Yichang, China
| | - Zhiyong Zhou
- Medical College of China Three Gorges University, Yichang, China
| | - Ting Wang
- Medical College of China Three Gorges University, Yichang, China
| | - Chaoqi Liu
- Medical College of China Three Gorges University, Yichang, China
| | - Yuhui Guo
- Medical College of China Three Gorges University, Yichang, China
| | - Changcheng Zhang
- Medical College of China Three Gorges University, Yichang, China
| | - Ding Yuan
- Renhe Hospital of China Three Gorges University, Yichang, China
| |
Collapse
|
26
|
Pulliam SR, Pellom ST, Shanker A, Adunyah SE. Butyrate regulates the expression of inflammatory and chemotactic cytokines in human acute leukemic cells during apoptosis. Cytokine 2016; 84:74-87. [PMID: 27253488 DOI: 10.1016/j.cyto.2016.05.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 02/06/2023]
Abstract
Butyrate is a histone deacetylase inhibitor implicated in many studies as a potential therapy for various forms of cancer. High concentrations of butyrate (>1.5mM) have been shown to activate apoptosis in several cancer cell lines including prostate, breast, and leukemia. Butyrate is also known to influence multiple signaling pathways that are mediators of cytokine production. The purpose of this study was to evaluate the impact of high concentrations of butyrate on the cancer microenvironment vis-à-vis apoptosis, cellular migration, and capacity to modulate cytokine expression in cancer cells. The results indicate that high concentrations of butyrate induced a 2-fold activation of caspase-3 and reduced cell viability by 60% in U937 leukemia cells. Within 24h, butyrate significantly decreased the levels of chemokines CCL2 and CCL5 in HL-60 and U937 cells, and decreased CCL5 in THP-1 leukemia cells. Differential effects were observed in treatments with valproic acid for CCL2 and CCL5 indicating butyrate-specificity. Many of the biological effects examined in this study are linked to activation of the AKT and MAPK signaling pathways; therefore, we investigated whether butyrate alters the levels of phosphorylated forms of these signaling proteins and how it correlated with the expression of chemokines. The results show that butyrate may partially regulate CCL5 production via p38 MAPK. The decrease in p-ERK1/2 and p-AKT levels correlated with the decrease in CCL2 production. These data suggest that while promoting apoptosis, butyrate has the potential to influence the cancer microenvironment by inducing differential expression of cytokines.
Collapse
Affiliation(s)
- Stephanie R Pulliam
- School of Graduate Studies and Research, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA
| | - Samuel T Pellom
- School of Graduate Studies and Research, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA
| | - Anil Shanker
- School of Graduate Studies and Research, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Host-Tumor Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Samuel E Adunyah
- School of Graduate Studies and Research, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA.
| |
Collapse
|
27
|
Liu J, Wang F, Luo H, Liu A, Li K, Li C, Jiang Y. Protective effect of butyrate against ethanol-induced gastric ulcers in mice by promoting the anti-inflammatory, anti-oxidant and mucosal defense mechanisms. Int Immunopharmacol 2016; 30:179-187. [DOI: 10.1016/j.intimp.2015.11.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 11/11/2015] [Accepted: 11/13/2015] [Indexed: 12/24/2022]
|
28
|
Korashy HM, Al-Suwayeh HA, Maayah ZH, Ansari MA, Ahmad SF, Bakheet SA. Mitogen-activated protein kinases pathways mediate the sunitinib-induced hypertrophy in rat cardiomyocyte H9c2 cells. Cardiovasc Toxicol 2015; 15:41-51. [PMID: 24984876 DOI: 10.1007/s12012-014-9266-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Sunitinib (SUN) is a multi-targeted tyrosine kinase inhibitor used for the treatment of gastrointestinal stromal tumors and renal cell carcinoma. Cardiotoxicity has been reported as a significant side effect associated with the SUN treatment, yet the mechanism is poorly understood. The main purpose of this study was to investigate the potential effects of SUN on cardiac hypertrophic genes and the role of mitogen-activated protein kinases (MAPKs) signaling pathway in rat cardiomyocyte H9c2 cell line. In the present study, real-time quantitative polymerase chain reaction showed that the treatment of H9c2 cells with increasing concentrations of SUN (0, 1, 2.5, and 5 µM) significantly induced hypertrophic gene markers, such as brain natriuretic peptides (BNP) and myosin heavy chain (β-MHC and α-MHC) in concentration- and time-dependent manners. The onset of mRNA induction was observed as early as 9 h and remained elevated for at least 18 h after treatment with SUN 5 µM. At the protein level, Western blot analysis showed that SUN increased BNP and β-MHC, while it inhibited α-MHC protein levels in a concentration-dependent manner. These SUN-mediated effects were associated with increase in cell size and hypertrophy by approximately 70 % at the highest concentration, 5 µM. Importantly, inhibition of the MAPK signaling pathway using SB203580 (p38 MAPK inhibitor), U0126 (extracellular signal-regulated kinase inhibitor), and SP600125 (c-Jun NH2-terminal kinase inhibitor) significantly potentiated the SUN-induced BNP and β-MHC mRNA levels, but did alter the α-MHC level. Whereas at the protein level, MAPK inhibitors generally decreased the SUN-induced BNP, whereas only SB and U0 increased β-MHC protein levels with no effect on α-MHC, which were associated with a significant decrease in cell size. Together, these results indicate that SUN induced hypertrophic gene expression through MAPK-dependent mechanisms.
Collapse
Affiliation(s)
- Hesham Mohamed Korashy
- Department of Pharmacology and Toxicology, College of Pharmacology, King Saud University, P.O. Box 2457, Riyadh, 11451, Kingdom of Saudi Arabia,
| | | | | | | | | | | |
Collapse
|
29
|
Supplementation of sodium butyrate protects mice from the development of non-alcoholic steatohepatitis (NASH). Br J Nutr 2015; 114:1745-55. [PMID: 26450277 DOI: 10.1017/s0007114515003621] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Overnutrition, insulin resistance and an impaired intestinal barrier function are discussed as critical factors in the development of non-alcoholic fatty liver disease. Not only butyrate-producing probiotics as well as supplementation of sodium butyrate (SoB) have been suggested to bear protective effects on liver damage of various aetiologies. However, whether an oral consumption of SoB has a protective effect on Western-style diet (WSD)-induced non-alcoholic steatohepatitis (NASH) and if so molecular mechanism involved has not yet been determined. Eight-week-old C57BL/6J mice were pair-fed either a liquid control or WSD±0·6 g/kg body weight SoB. After 6 weeks, markers of liver damage, inflammation, toll-like receptor (TLR)-4 signalling, lipid peroxidation and glucose as well as lipid metabolism were determined in the liver tissue. Tight junction protein levels were determined in the duodenal tissue. SoB supplementation had no effects on the body weight gain or liver weight of WSD-fed mice, whereas liver steatosis and hepatic inflammation were significantly decreased (e.g. less inflammatory foci and neutrophils) when compared with mice fed only a WSD. Tight junction protein levels in duodenum, hepatic mRNA expression of TLR-4 and sterol regulatory element-binding protein 1c were altered similarly in both WSD groups when compared with controls, whereas protein levels of myeloid differentiation primary response gene 88, inducible nitric oxide synthase, 4-hydroxynonenal protein adducts and F4/80 macrophages were only significantly induced in livers of mice fed only the WSD. In summary, these data suggest that an oral supplementation of SoB protects mice from inflammation in the liver and thus from the development of WSD-induced NASH.
Collapse
|