1
|
Su Q, Tang Q, Ma C, Wang K. Advances in the study of the relationship between gut microbiota and erectile dysfunction. Sex Med Rev 2024; 12:664-669. [PMID: 38984896 DOI: 10.1093/sxmrev/qeae049] [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: 04/17/2024] [Revised: 06/15/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024]
Abstract
INTRODUCTION In recent years, in-depth research has revealed that gut microbiota has an inseparable relationship with erectile dysfunction (ED) in men. OBJECTIVES (1) To review the correlation between gut microbiota and ED from the perspective of its impact on men's mental health, metabolism, immunity, and endocrine regulation and (2) to provide reference to further explore the pathogenesis of ED and the improvement of clinical treatment plans. METHODS PubMed was used for the literature search to identify publications related to ED and gut microbiota. RESULTS Gut microbiota may induce depression and anxiety through the microbiota-gut-brain axis, leading to the occurrence of psychological ED. It may also cause vascular endothelial dysfunction and androgen metabolism disorder by interfering with lipid metabolism, immunity, and endocrine regulation, leading to the occurrence of organic ED. CONCLUSION Gut microbiota and its metabolites play an important role in the occurrence and development of ED. As a new influencing factor of ED, gut microbiota disorder is expected to become a target for treatment.
Collapse
Affiliation(s)
- Quanxin Su
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Qizhen Tang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Chuanyu Ma
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Kenan Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| |
Collapse
|
2
|
Tang L, Liu J, Yang H, Zhao HQ, Hu C, Ma SJ, Qing YH, Yang L, Zhou RR, Zhang SH. Microbiome Metabolomic Analysis of the Anxiolytic Effect of Baihe Dihuang Decoction in a Rat Model of Chronic Restraint Stress. Drug Des Devel Ther 2024; 18:2227-2248. [PMID: 38882046 PMCID: PMC11180446 DOI: 10.2147/dddt.s458983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/03/2024] [Indexed: 06/18/2024] Open
Abstract
Purpose The Baihe Dihuang decoction (BDD) is a representative traditional Chinese medicinal formula that has been used to treat anxiety disorders for thousands of years. This study aimed to reveal mechanisms of anxiolytic effects of BDD with multidimensional omics. Methods First, 28-day chronic restraint stress (CRS) was used to create a rat model of anxiety, and the open field test and elevated plus maze were used to assess anxiety-like behavior. Enzyme-linked immunosorbent assay (ELISA), hematoxylin-eosin staining, and immunofluorescence staining were used to evaluate inflammatory response. Besides, 16S rRNA gene sequencing assessed fecal microbiota composition and differential microbiota. Non-targeted metabolomics analysis of feces was performed to determine fecal biomarkers, and targeted metabolomics was used to observe the levels of hippocampus neurotransmitters. Finally, Pearson correlation analysis was used to examine relationships among gut microbiota, fecal metabolites, and neurotransmitters. Results BDD significantly improved anxiety-like behaviors in CRS-induced rats and effectively ameliorated hippocampal neuronal damage and abnormal activation of hippocampal microglia. It also had a profound effect on the diversity of microbiota, as evidenced by significant changes in the abundance of 10 potential microbial biomarkers at the genus level. Additionally, BDD led to significant alterations in 18 fecal metabolites and 12 hippocampal neurotransmitters, with the majority of the metabolites implicated in amino acid metabolism pathways such as D-glutamine and D-glutamate, alanine, arginine and proline, and tryptophan metabolism. Furthermore, Pearson analysis showed a strong link among gut microbiota, metabolites, and neurotransmitters during anxiety and BDD treatment. Conclusion BDD can effectively improve anxiety-like behaviors by regulating the gut-brain axis, including gut microbiota and metabolite modification, suppression of hippocampal neuronal inflammation, and regulation of neurotransmitters.
Collapse
Affiliation(s)
- Lin Tang
- Hospital-Made Preparations Center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, People's Republic of China
| | - Jian Liu
- Medical Innovation Experiment Center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, People's Republic of China
| | - Hui Yang
- Medical Innovation Experiment Center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, People's Republic of China
| | - Hong-Qing Zhao
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan Province, People's Republic of China
| | - Chao Hu
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, People's Republic of China
| | - Si-Jing Ma
- The Affiliated Hospital, Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan Province, People's Republic of China
| | - Yu-Hui Qing
- Institute of Chinese Medicine Resources, Hunan Academy of Chinese Medicine, Changsha, Hunan Province, People's Republic of China
| | - Lei Yang
- Hospital-Made Preparations Center, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, People's Republic of China
| | - Rong-Rong Zhou
- The Affiliated Hospital, Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan Province, People's Republic of China
| | - Shui-Han Zhang
- Institute of Chinese Medicine Resources, Hunan Academy of Chinese Medicine, Changsha, Hunan Province, People's Republic of China
| |
Collapse
|
3
|
Xia M, Xu Y, Li H, Huang J, Zhou H, Gao C, Han J. Structural and functional alteration of the gut microbiota in elderly patients with hyperlipidemia. Front Cell Infect Microbiol 2024; 14:1333145. [PMID: 38812752 PMCID: PMC11133514 DOI: 10.3389/fcimb.2024.1333145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Objective To investigate the structure, composition, and functions of the gut microbiota in elderly patients with hyperlipidemia. Methods Sixteen older patients diagnosed with hyperlipidemia (M group) and 10 healthy, age-matched normal volunteers (N group) were included. These groups were further subdivided by sex into the male normal (NM, n = 5), female normal (NF, n = 5), male hyperlipidemia (MM, n = 8), and female hyperlipidemia (MF, n = 8) subgroups. Stool samples were collected for high-throughput sequencing of 16S rRNA genes. Blood samples were collected for clinical biochemical index testing. Results Alpha- and beta-diversity analyses revealed that the structure and composition of the gut microbiota were significantly different between the M and N groups. The relative abundances of Bacteroides, Parabacteroides, Blautia, Peptococcus, and Bifidobacterium were significantly decreased, while those of Lactobacillus, Helicobacter, and Desulfovibrio were significantly higher in the M group. There were also significant sex-related differences in microbial structure between the NM and NF groups, and between the MM and MF groups. Through functional prediction with PICRUSt 2, we observed distinct between-group variations in metabolic pathways associated with the gut microbiota and their impact on the functionality of the nervous system. Pearson's correlation coefficient was used as a distance metric to build co-abundance networks. A hypergeometric test was used to detect taxonomies with significant enrichment in specific clusters. We speculated that modules with Muribaculaceae and Lachnospiraceae as the core microbes play an important ecological role in the intestinal microbiota of the M group. The relative intestinal abundances of Agathobacter and Faecalibacterium in the M group were positively correlated with serum triglyceride and low-density lipoprotein levels, while the relative abundance of Bifidobacterium was negatively correlated with the serum lipoprotein a level.
Collapse
Affiliation(s)
- Meng Xia
- Department of Clinical Laboratory, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yafang Xu
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Huajun Li
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Juan Huang
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Haolin Zhou
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Chuanzhou Gao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jingyi Han
- Department of Clinical Laboratory, First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
4
|
Wang N, Li C, Gao X, Huo Y, Li Y, Cheng F, Jiang F, Zhang Z. Co-exposure to lead and high-fat diet aggravates systemic inflammation in mice by altering gut microbiota and the LPS/TLR4 pathway. Metallomics 2024; 16:mfae022. [PMID: 38658185 DOI: 10.1093/mtomcs/mfae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/23/2024] [Indexed: 04/26/2024]
Abstract
This study reports the toxicity of Pb exposure on systemic inflammation in high-fat-diet (HFD) mice and the potential mechanisms. Results indicated that Pb exacerbated intestinal barrier damage and increased serum levels of lipopolysaccharide (LPS) and diamine oxidase in HFD mice. Elevated LPS activates the colonic and ileal LPS-TLR4 inflammatory signaling pathway and further induces hepatic and adipose inflammatory expression. The 16S rRNA gene sequencing results showed that Pb promoted the abundance of potentially harmful and LPS-producing bacteria such as Coriobacteriaceae_UCG-002, Alloprevotella, and Oscillibacter in the intestines of HFD mice, and their abundance was positively correlated with LPS levels. Additionally, Pb inhibited the abundance of the beneficial bacteria Akkermansia, resulting in lower levels of the metabolite short-chain fatty acids (SCFAs). Meanwhile, Pb inhibited adenosine 5'-monophosphate-activated protein kinase signaling-mediated lipid metabolism pathways, promoting hepatic lipid accumulation. The above results suggest that Pb exacerbates systemic inflammation and lipid disorders in HFD mice by altering the gut microbiota, intestinal barrier, and the mediation of metabolites LPS and SCFAs. Our study provides potential novel mechanisms of human health related to Pb-induced metabolic damage and offers new evidence for a comprehensive assessment of Pb risk.
Collapse
Affiliation(s)
- Nana Wang
- School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Changhao Li
- School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Xue Gao
- School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Yuan Huo
- School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Yuting Li
- School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Fangru Cheng
- School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Fei Jiang
- School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Zengli Zhang
- School of Public Health, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| |
Collapse
|
5
|
Xiao H, Yin D, Du L, Li G, Lin J, Fang C, Shen S, Xiao G, Fang R. Effects of pork sausage on intestinal microecology and metabolism in mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:3413-3427. [PMID: 38111159 DOI: 10.1002/jsfa.13227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/01/2023] [Accepted: 12/16/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Processed meat, as an important part of the human diet, has been recognized as a carcinogen by the International Agency for Research on Cancer (IARC). Although numerous epidemiological reports supported the IARC's view, the relevant evidence of a direct association between processed meat and carcinogenicity has been insufficient and the mechanism has been unclear. This study aims to investigate the effects of pork sausage (as a representative example of processed meat) intake on gut microbial communities and metabolites of mice. Microbial communities and metabolites from all groups were analyzed using 16S rRNA gene sequencing and Ultra performance liquid chromatography-quadrupole-time of flight-mass spectrometer (UPLC-Q-TOF/MS), respectively. RESULTS The levels of Bacteroidetes, Bacteroides, Alloprevotella, Lactobacillus, Prevotella_9, Lachnospiraceae_NK4A136_group, Alistipes, Blautia, Proteobacteria, Firmicutes, Allobaculum, Helicobacter, Desulfovibrio, Clostridium_sensu_stricto_1, Ruminococcaceae_UCG-014, Lachnospiraceae_UCG-006 and Streptococcus (P < 0.05) were obviously altered in the mice fed a pork sausage diet. Twenty-seven metabolites from intestinal content samples and fourteen matabolites from whole blood samples were identified as potential biomarkers from multivariate analysis, including Phosphatidic acid (PA), Sphingomyelin (SM), Lysophosphatidylcholine (LysoPC), Diglyceride (DG), D-maltose, N-acylamides and so forth. The significant changes in these biomarkers demonstrate metabonomic variations in pork sausage treated rats, especially carbohydrate metabolism, lipid metabolism, and amino acid metabolism. CONCLUSION The present study provided evidence that a processed meat diet can increase the risk of colorectal cancer and other diseases significantly by altering the microbial community structure and disrupting the body's metabolic pathways. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Hailong Xiao
- Key Laboratory of Agricultural Products Chemical and Biological Processing Technology, Zhejiang University of Science and Technology, Hangzhou, China
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Danhan Yin
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Lidan Du
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Gaotian Li
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Jie Lin
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Chenyu Fang
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Shaolin Shen
- Hangzhou Xiaoshan Institute of Measurement for Quality and Technique Supervision, Hangzhou, China
| | - Gongnian Xiao
- Key Laboratory of Agricultural Products Chemical and Biological Processing Technology, Zhejiang University of Science and Technology, Hangzhou, China
| | - Ruosi Fang
- Key Laboratory of Agricultural Products Chemical and Biological Processing Technology, Zhejiang University of Science and Technology, Hangzhou, China
| |
Collapse
|
6
|
Wu S, Du W, Wu Z, Wen F, Zhong X, Huang X, Gu H, Wang J. Effect of chronic noise exposure on glucose and lipid metabolism in mice via modulating gut microbiota and regulating CREB/CRTC2 and SREBP1/SCD pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115887. [PMID: 38157803 DOI: 10.1016/j.ecoenv.2023.115887] [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: 09/29/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Chronic noise exposure is correlated with gut microbiota dysbiosis and glucose and lipid metabolism disorders. However, evidence on the mechanisms underlying of gut microbiota alterations in chronic noise induced glucose and lipid metabolism disorders is limited, and the potential aftereffects of chronic noise exposure on metabolic disorders remain unclear. In present study, we established chronic daytime and nighttime noise exposure mice models to explore the effects and underlying mechanism of gut microbiota on chronic noise-induced glucose and lipid metabolism disorders. The results showed that exposure to chronic daytime or nighttime noise significantly increased the fasting blood glucose, serum and liver TG levels, impaired glucose tolerance, and decreased serum HDL-C levels and liver TC levels in mice. However, after 4 weeks of recovery, only serum TG of mice in nighttime noise recovery group remained elevated. Besides, exposure to chronic noise reduced the intestinal tight junction protein levels and increased intestinal permeability, while this effect did not completely dissipate even after the recovery period. Moreover, chronic noise exposure changed the gut microbiota and significantly regulated metabolites and metabolic pathways, and further activate hepatic gluconeogenesis CRTC2/CREB-PCK1 signaling pathway and lipid synthesis SREBP1/SCD signaling pathway through intestinal hepatic axis. Together, our findings demonstrated that chronic daytime and nighttime noise exposure could cause the glucose and lipid metabolism disorder by modulating the gut microbiota and serum metabolites, and activating hepatic gluconeogenic CREB/CRTC2-PCK1 signaling and lipid synthesis SREBP1/SCD signaling pathway. The potential aftereffects of noise exposure during wakefulness on metabolic disorders are more significant than that of noise exposure during sleep.
Collapse
Affiliation(s)
- Shan Wu
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510000, China
| | - Wenjing Du
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510000, China
| | - Zhidan Wu
- Guangzhou Baiyun District Center for Disease Control and Prevention, Guangzhou 510445, China
| | - Fei Wen
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510000, China
| | - Xiangbin Zhong
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510000, China
| | - Xin Huang
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510000, China
| | - Haoyan Gu
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510000, China
| | - Junyi Wang
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510000, China.
| |
Collapse
|
7
|
Zeng W, Yang B, Wang Y, Sun M, Yang W, Cui H, Jin J, Zhao Z. Rotundic acid alleviates hyperlipidemia in rats by regulating lipid metabolism and gut microbiota. Phytother Res 2023; 37:5958-5973. [PMID: 37776121 DOI: 10.1002/ptr.8008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 10/01/2023]
Abstract
Disturbances in lipid metabolism and dysbiosis of the gut microbiota play an important role in the progression of hyperlipidemia. Previous study indicated that Ilicis Rotundae Cortex possesses anti-hyperlipidemic activity, and rotundic acid (RA) identified as a key active compound to be incorporated into the body. The study aimed to evaluate the anti-hyperlipidemia effects of RA and explored its impact on gut microbiota and lipid metabolism, as well as its possible mechanisms for improving hyperlipidemia. The study methodology included a comprehensive evaluation of the effects of RA on steatosis markers of hyperlipidemia, lipid metabolism, and gut microbiota by assessing biochemical parameters and histopathology, lipidomics, 16S rRNA gene sequencing, and short-chain fatty acid (SCFA) assays. The results showed that RA effectively reduced body weight and the steatosis markers in serum and liver. Moreover, the lipidomic analysis revealed significant changes in plasmatic and hepatic lipid levels, and these were restored by RA. According to the results of 16S rRNA gene sequencing, RA supplementation raised the relative abundance of Bacteroidetes and Proteobacteria while decreasing the relative abundance of Firmicutes. RA significantly boosted the relative abundance of SCFAs by increasing SCFAs-producing bacteria such as Bacteroides, Alloprevotella, Desulfovibrio, etc. In summary, RA could regulate triglyceride metabolism and glycerophospholipid metabolism, restore gut microbiota structure, and increase the relative abundance of SCFAs-producing bacteria to exert its hypolipidemic effects. These findings suggest RA to be a promising therapeutic agent for hyperlipidemia.
Collapse
Affiliation(s)
- Wei Zeng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bao Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, Enshi, China
| | - Yuanyuan Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mengjia Sun
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiqun Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui Cui
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Jin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Zhongxiang Zhao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
8
|
Ismail MF, Lim SM, Lim FT, Ramasamy K. In Vitro and In Vivo Characterisation of Lactiplantibacillus plantarum LAB12 in Pea Protein-Alginate Microcapsules. Probiotics Antimicrob Proteins 2023:10.1007/s12602-023-10171-6. [PMID: 37816988 DOI: 10.1007/s12602-023-10171-6] [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] [Accepted: 09/27/2023] [Indexed: 10/12/2023]
Abstract
The susceptibility of probiotics to high temperature and low pH remains a major challenge in food industries. Numerous commercially available probiotic products were reportedly presented with lower probiotic viability than claimed. To confer health benefits to the host, it is essential that probiotic strain remains viable at optimal amount during food processing procedures, storage and passage through the gastrointestinal tract. This study addressed these issues by immobilising Lactiplantibacillus plantarum LAB12 isolated from tempeh (fermented soybean) in a polymeric matrix made up of alginate (Alg, 0.5% w/v) and denatured pea protein isolate (PPi, 1-10% w/v) using the emulsion/acidification technique. Alg supplemented with 10% PPi (Alg-PPi10) appeared to be optimally small (< 350 µm), substantiated by the improved surface smoothness and uniform dispersion of probiotics in the Alg-PPi core. The findings indicated that microencapsulation enhanced thermal stability of L. plantarum LAB12. The microencapsulated L. plantarum LAB12 remained highly viable (80%) despite exposure to 100 °C for 5 min. The microencapsulated cell number during storage at 4 and 25 °C for 8 weeks was greater than 7 log CFU g-1. L. plantarum LAB12 encapsulated in Alg-PPi10 exhibited high viability (96%) in simulated gastric juice (at pH 1.8 for 120 min) and facilitated maximum release of probiotics (> 9 log CFU g-1) in simulated intestinal fluid (at pH 6.8 for 240 min). Whilst retaining their intrinsic cholesterol lowering effect, microencapsulation conferred additional advantages to L. plantarum LAB12 in terms of lowering serum triglyceride and increasing HDL cholesterol in zebrafish fed with high-cholesterol diet (HCD). Overall, our findings strongly imply the potential use of Alg-PPi10 as an effective medium that confers thermal protection and facilitates pH-sensitive release of cholesterol-reducing L. plantarum LAB12. This will allow the diverse applications L. plantarum LAB12 across health, food and agro-feed industries amongst others.
Collapse
Affiliation(s)
- Muhamad Fareez Ismail
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
- School of Biology, Faculty of Applied Sciences, Universiti Teknologi MARA (UiTM), Shah Alam Campus, 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Fei Tieng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
9
|
Wang Y, Rong X, Guan H, Ouyang F, Zhou X, Li F, Tan X, Li D. The Potential Effects of Isoleucine Restricted Diet on Cognitive Impairment in High-Fat-Induced Obese Mice via Gut Microbiota-Brain Axis. Mol Nutr Food Res 2023; 67:e2200767. [PMID: 37658490 DOI: 10.1002/mnfr.202200767] [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: 11/06/2022] [Revised: 05/15/2023] [Indexed: 09/03/2023]
Abstract
SCOPE Obesity induced by high-fat diet (HFD) can cause lipid metabolism disorders and cognitive impairment. Isoleucine restriction can effectively alleviate lipid metabolism disorders caused by HFD but the underlying mechanisms on cognition are unknown. METHODS AND RESULTS Thirty 3-month-old C57BL/6J mice are divided equally into the following groups: the control group, HFD group, and HFD Low Ile group (67% reduction in isoleucine in high fat feeds). Feeding for 11 weeks with behavioral testing, which shows that isoleucine restriction attenuates HFD-induced cognitive dysfunction. As observed by staining, isoleucine restriction inhibits HFD-induced neuronal damage and microglia activation. Furthermore, isoleucine restriction significantly increases the relative abundance of gut microbiota, decreases the proportion of Proteobacteria, and reduces the levels of lipopolysaccharide (LPS) in serum and brain. Isoleucine restriction reduces protein expression of TLR4/MyD88/NF-κB signaling pathway and inhibits upregulation of proinflammatory cytokine genes and protein expression in mice brain. In addition, isoleucine restriction significantly improves insulin resistance in the brain as well as synaptic plasticity impairment. CONCLUSION Isoleucine restriction may be a potential intervention to reduce HFD-induced cognitive impairment by altering gut microbiota, reducing neuroinflammation, insulin resistance, and improving synaptic plasticity in mice brain.
Collapse
Affiliation(s)
- Yuli Wang
- College of Food Science and Engineering, Shandong Agricultural University Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, 271018, China
| | - Xue Rong
- College of Food Science and Engineering, Shandong Agricultural University Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, 271018, China
| | - Hui Guan
- College of Food Science and Engineering, Shandong Agricultural University Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, 271018, China
| | - Fangxin Ouyang
- College of Food Science and Engineering, Shandong Agricultural University Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, 271018, China
| | - Xing Zhou
- College of Food Science and Engineering, Shandong Agricultural University Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, 271018, China
| | - Feng Li
- College of Food Science and Engineering, Shandong Agricultural University Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, 271018, China
| | - Xintong Tan
- College of Food Science and Engineering, Shandong Agricultural University Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, 271018, China
| | - Dapeng Li
- College of Food Science and Engineering, Shandong Agricultural University Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, 271018, China
| |
Collapse
|
10
|
SHI J, XIE Y, LI Y, REN D, ZHANG Y, SHAO H, LIU Y, WANG X, LI Y. Effects of food-grade iron(III) oxide nanoparticles on cecal digesta- and mucosa-associated microbiota and short-chain fatty acids in rats. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2023; 43:43-54. [PMID: 38188661 PMCID: PMC10767317 DOI: 10.12938/bmfh.2023-012] [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: 02/15/2023] [Accepted: 08/21/2023] [Indexed: 01/09/2024]
Abstract
Although iron(III) oxide nanoparticles (IONPs) are widely used in diverse applications ranging from food to biomedicine, the effects of IONPs on different locations of gut microbiota and short-chain fatty acids (SCFAs) are unclear. So, a subacute repeated oral toxicity study on Sprague Dawley (SD) rats was performed, administering low (50 mg/kg·bw), medium (100 mg/kg·bw), and high (200 mg/kg·bw) doses of IONPs. In this study, we found that a high dose of IONPs increased animal weight, and 16S rRNA sequencing revealed that IONPs caused intestinal flora disorders in both the cecal digesta- and mucosa-associated microbiota. However, only high-dose IONP exposure changed the abundance and composition of the mucosa-associated microbiota. IONPs increased the relative abundances of Firmicutes, Ruminococcaceae_UCG-014, Ruminiclostridium_9, Romboutsia, and Bilophila and decreased the relative abundance of Bifidobacterium, and many of these microorganisms are associated with weight gain, obesity, inflammation, diabetes, and mucosal damage. Functional analysis showed that changes in the gut microbiota induced by a high dose of IONPs were mainly related to metabolism, infection, immune, and endocrine disease functions. IONPs significantly elevated the levels of valeric, isobutyric, and isovaleric acid, promoting the absorption of iron. This is the first description of intestinal microbiota dysbiosis in SD rats caused by IONPs, and the effects and mechanisms of action of IONPs on intestinal and host health need to be further studied and confirmed.
Collapse
Affiliation(s)
- Jiangchun SHI
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yumeng XIE
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yulin LI
- Department of Hospital-acquired Infection Management, Guizhou
Provincial People’s Hospital, Guiyang 550002, China
| | - Dongxia REN
- Department of Blood Transfusion, Tangdu Hospital, Fourth
Military Medical University, Xi’an 710032, China
| | - Yiqi ZHANG
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Huangfang SHAO
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yang LIU
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Xue WANG
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yun LI
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
- Provincial Key Laboratory of Food Safety Monitoring and Risk
Assessment of Sichuan, Chengdu 610041, China
| |
Collapse
|
11
|
Liu Y, Xie T, Wu S, Yang G, Zhang J, Song J, Yang G. Effect of macadamia oil cake on blood lipid characteristics and intestinal microbiota in hyperlipidemic rat. Food Sci Nutr 2023; 11:5318-5324. [PMID: 37701238 PMCID: PMC10494627 DOI: 10.1002/fsn3.3490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 09/14/2023] Open
Abstract
Macadamia oil cake (MOC) is a type of macadamia nut by-product, that is extremely rich in amino acids and has beneficial health effects. It lowers blood lipid levels and regulates the intestinal microbiota. MOC effectively attenuated total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) levels in model rats. Depending on the morphology of the colon, MOC can effectively attenuate damage to the tissue structure. The 16S rDNA gene of the rat intestinal microbiota was sequenced using Illumina PE250 high-throughput sequencing technology, and the changes in the intestinal microbiota in each group are discussed. Supplementing MOC at different doses significantly increased the microbiota of Dorea, Erysipelotrichaceae, Stercoris, etc. in the intestinal tracts of rats fed a high-fat diet. Therefore, MOC can be included in lipid healthy dietary patterns to lower lipid characteristics and restructure the intestinal microbiota. Future clinical trials are required to determine the therapeutic effects and mechanisms of hypolipidemia.
Collapse
Affiliation(s)
- Yao Liu
- Guangdong Eco‐engineering PolytechnicGuangzhouChina
| | - Tengfei Xie
- Guangdong Eco‐engineering PolytechnicGuangzhouChina
| | - Shijun Wu
- Guangdong Eco‐engineering PolytechnicGuangzhouChina
| | - Guang Yang
- Guangdong Eco‐engineering PolytechnicGuangzhouChina
| | - Jinyun Zhang
- Guangdong Eco‐engineering PolytechnicGuangzhouChina
| | - Jie Song
- Guangdong Eco‐engineering PolytechnicGuangzhouChina
| | - Guifang Yang
- Guangdong Eco‐engineering PolytechnicGuangzhouChina
| |
Collapse
|
12
|
Fan X, Zhang Q, Guo W, Wu Q, Hu J, Cheng W, Lü X, Rao P, Ni L, Chen Y, Chen L. The protective effects of Levilactobacillus brevis FZU0713 on lipid metabolism and intestinal microbiota in hyperlipidemic rats. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
13
|
Modulatory effects of Lactiplantibacillus plantarum on chronic metabolic diseases. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
14
|
Zhao D, Zhang R, Wang J, Zhang X, Liu K, Zhang H, Liu H. Effect of Limosilactobacillus reuteri ZJF036 on Growth Performance and Gut Microbiota in Juvenile Beagle Dogs. Curr Microbiol 2023; 80:155. [PMID: 36995478 DOI: 10.1007/s00284-023-03276-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/18/2023] [Indexed: 03/31/2023]
Abstract
This experiment investigated the effects of Limosilactobacillus reuteri ZJF036 on growth performance, serum biochemical parameters, and gut microbiota in beagle dogs. Sixteen 75 ± 5-day-old healthy male beagles (4.51 ± 1.37 kg) were randomly divided into two groups; the experimental group (L1) and the control group (L0), and then fed with or without a basal diet containing L. reuteri ZJF036 (109 CFU/g), respectively. The results showed that there was no significant difference in daily weight gain between the two groups (P > 0.05). However, we found that L. reuteri ZJF036 decreased Chao1 index and ACE index and increased the relative abundance of Firmicutes and Fusobacteria (P < 0.05) compared to the L0 group. In addition, we also found that the ratio of Firmicutes to Bacteroidetes was decreased in L1 group. Furthermore, the relative abundance of Lactobacillus increased, while that of Turicibacter and Blautia decreased in L1 group (P < 0.05). In conclusion, L. reuteri ZJF036 appeared to regulate the intestinal microbiota of beagle dogs. This study revealed the potential use of L. reuteri ZJBF036 as a probiotic supplement for beagle dogs.
Collapse
Affiliation(s)
- Dehui Zhao
- College of Agriculture, Chifeng University, Chifeng, 024000, People's Republic of China
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation, Hebei Normal University of Science and Technology, Qinhuangdao, 066004, People's Republic of China
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, 130112, People's Republic of China
| | - Ruchun Zhang
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, 130112, People's Republic of China
| | - Jinming Wang
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, 130112, People's Republic of China
| | - Xinyu Zhang
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, 130112, People's Republic of China
| | - Keyuan Liu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Haihua Zhang
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation, Hebei Normal University of Science and Technology, Qinhuangdao, 066004, People's Republic of China
| | - Hanlu Liu
- College of Agriculture, Chifeng University, Chifeng, 024000, People's Republic of China.
| |
Collapse
|
15
|
Salman MK, Abuqwider J, Mauriello G. Anti-Quorum Sensing Activity of Probiotics: The Mechanism and Role in Food and Gut Health. Microorganisms 2023; 11:microorganisms11030793. [PMID: 36985366 PMCID: PMC10056907 DOI: 10.3390/microorganisms11030793] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
Background: Quorum sensing (QS) is a cell-to-cell communication mechanism that occurs between inter- and intra-bacterial species and is regulated by signaling molecules called autoinducers (AIs). It has been suggested that probiotics can exert a QS inhibitory effect through their metabolites. Purpose: To provide an overview of (1) the anti-QS activity of probiotics and its mechanism against foodborne pathogenic and spoilage bacteria; (2) the potential role of the QS of probiotics in gut health; and (3) the impact of microencapsulation on QS. Results: Lactobacillus species have been extensively studied for their anti-QS activity and have been found to effectively disrupt QS in vitro. However, their effectiveness in a food matrix is yet to be determined as they interfere with the AI receptor or its synthesis. QS plays an important role in both the biofilm formation of probiotics and pathogenic bacteria. Moreover, in vitro and animal studies have shown that QS molecules can modulate cytokine responses and gut dysbiosis and maintain intestinal barrier function. In this scenario, microencapsulation was found to enhance AI activity. However, its impact on the anti-QS activity of probiotics and its underlying mechanism remains unclear. Conclusions: Probiotics are potential candidates to block QS activity in foodborne pathogenic and food spoilage bacteria. Microencapsulation increases QS efficacy. However, more research is still needed for the identification of the QS inhibitory metabolites from probiotics and for the elucidation of the anti-QS mechanism of probiotics (microcapsules and free cells) in food and the human gut.
Collapse
|
16
|
Niu K, Bai P, Zhang J, Feng X, Qiu F. Cytidine Alleviates Dyslipidemia and Modulates the Gut Microbiota Composition in ob/ob Mice. Nutrients 2023; 15:nu15051147. [PMID: 36904146 PMCID: PMC10005144 DOI: 10.3390/nu15051147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Cytidine and uridine are endogenous metabolites in the pyrimidine metabolism pathway, and cytidine is a substrate that can be metabolized into uridine via cytidine deaminase. Uridine has been widely reported to be effective in regulating lipid metabolism. However, whether cytidine could ameliorate lipid metabolism disorder has not yet been investigated. In this research, ob/ob mice were used, and the effect of cytidine (0.4 mg/mL in drinking water for five weeks) on lipid metabolism disorder was evaluated in terms of an oral glucose tolerance test, serum lipid levels, liver histopathological analysis and gut microbiome analysis. Uridine was used as a positive control. Our findings reveal that cytidine could alleviate certain aspects of dyslipidemia and improve hepatic steatosis via modulating the gut microbiota composition in ob/ob mice, especially increasing the abundance of short-chain fatty acids-producing microbiota. These results suggest that cytidine supplementation could be a potential therapeutic approach for dyslipidemia.
Collapse
Affiliation(s)
| | | | | | - Xinchi Feng
- Correspondence: (X.F.); (F.Q.); Tel.: +86-22-595-6223 (X.F.)
| | - Feng Qiu
- Correspondence: (X.F.); (F.Q.); Tel.: +86-22-595-6223 (X.F.)
| |
Collapse
|
17
|
Tian G, Wang W, Xia E, Chen W, Zhang S. Dendrobium officinale alleviates high-fat diet-induced nonalcoholic steatohepatitis by modulating gut microbiota. Front Cell Infect Microbiol 2023; 13:1078447. [PMID: 36860985 PMCID: PMC9968977 DOI: 10.3389/fcimb.2023.1078447] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction The gut microbiota plays an important role in the development of nonalcoholic steatohepatitis (NASH). This study investigated the preventive effect of Dendrobium officinale (DO), including whether its effect was related to the gut microbiota, intestinal permeability and liver inflammation. Methods A NASH model was established in rats using a high-fat diet (HFD) and gavage with different doses of DO or Atorvastatin Calcium (AT) for 10 weeks. Body weight and body mass index along with liver appearance, weight, index, pathology, and biochemistry were measured to assess the preventive effects of DO on NASH rats. Changes in the gut microbiota were analyzed by 16S rRNA sequencing, and intestinal permeability and liver inflammation were determined to explore the mechanism by which DO treatment prevented NASH. Results Pathological and biochemical indexes showed that DO was able to protect rats against HFD-induced hepatic steatosis and inflammation. Results of 16S rRNA sequencing showed that Proteobacteria, Romboutsia, Turicibacter, Lachnoclostridium, Blautia, Ruminococcus_torques_group, Sutterella, Escherichia-Shigella, Prevotella, Alistipes, and Lactobacillus_acidophilus differed significantly at the phylum, genus, and species levels. DO treatment modulated the diversity, richness, and evenness of gut microbiota, downregulated the abundance of the Gram-negative bacteria Proteobacteria, Sutterella, and Escherichia-Shigella, and reduced gut-derived lipopolysaccharide (LPS) levels. DO also restored expression of the tight junction proteins, zona occludens-1 (ZO-1), claudin-1, and occludin in the intestine and ameliorated the increased intestinal permeability caused by HFD, gut microbiota such as Turicibacter, Ruminococcus, Escherichia-Shigella, and Sutterella, and LPS. Lower intestinal permeability reduced LPS delivery to the liver, thus inhibiting TLR4 expression and nuclear factor-kappaB (NF-κB) nuclear translocation, improving liver inflammation. Discussion These results suggest that DO may alleviate NASH by regulating the gut microbiota, intestinal permeability, and liver inflammation.
Collapse
Affiliation(s)
- Gege Tian
- College of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China,The Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Yunnan University of Chinese Medicine, Kunming, China
| | - Wei Wang
- The Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Yunnan University of Chinese Medicine, Kunming, China,College of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Enrui Xia
- College of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China,The Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Yunnan University of Chinese Medicine, Kunming, China
| | - Wenhui Chen
- The Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Yunnan University of Chinese Medicine, Kunming, China,College of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, China,*Correspondence: Shunzhen Zhang, ; Wenhui Chen,
| | - Shunzhen Zhang
- College of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China,The Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Yunnan University of Chinese Medicine, Kunming, China,*Correspondence: Shunzhen Zhang, ; Wenhui Chen,
| |
Collapse
|
18
|
Transcriptome and Gut Microbiota Profiling Analysis of ANIT-Induced Cholestasis and the Effects of Da-Huang-Xiao-Shi Decoction Intervention. Microbiol Spectr 2022; 10:e0324222. [PMID: 36409145 PMCID: PMC9769994 DOI: 10.1128/spectrum.03242-22] [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] [Indexed: 11/23/2022] Open
Abstract
Cholestasis is characterized by bile acid (BA) circulation disorders, which is usually related to damage of hepatocyte barrier function. Currently, patients with cholestasis face several obstacles in seeking diagnosis and therapy. Da-Huang-Xiao-Shi decoction (DHXSD) is an ancient classic formula that has been used clinically for cholestasis treatment. Nevertheless, the underlying biological activities and therapeutic mechanisms remain unclear. In this study, an alpha-naphthylisothiocyanate (ANIT)-induced cholestasis rat model was established to examine the anticholestatic effects of DHXSD using histopathological and molecular analyses. Transcriptomic analysis combined with 16S rRNA gene sequencing analysis was systematically applied to study the mechanism of action of DHXSD. Simultaneously, the effect of DHXSD on gut microbiota, short-chain fatty acids (SCFAs), and intestinal barrier function were evaluated based on the ANIT-induced cholestasis model in rats. The results showed that DHXSD effectively attenuated ANIT-induced cholestasis by reducing liver function indicators (alanine transaminase [ALT], P < 0.05; alkaline phosphatase [ALP], P < 0.05; total bile acid [TBA], P < 0.01; γ-glutamyl transpeptidase [GGT], P < 0.001) and levels of hepatotoxicity-related enzymes (P < 0.05), thus improving the recovery of histopathological injuries, and regulating levels of inflammatory cytokines (P < 0.05). In addition, 16S rRNA gene sequencing analysis combined with intestinal barrier function analysis revealed that the DHXSD significantly ameliorated ANIT-induced gut microbiota dysbiosis. Significantly altered genes in the model and treatment groups were screened using transcriptomic analysis. Sixty-eight genes and four microbial genera were simultaneously altered with opposing trends in variation after ANIT and DHXSD treatments. We built a framework for predicting targets and host-microbe interaction mechanisms, as well as identifying alternative treatment for cholestasis, which should be validated further for clinical application. In conclusion, DHXSD appears to be a promising agent for protection against liver injury. IMPORTANCE Cholestasis is a serious manifestation of liver diseases resulting in liver injury, fibrosis, and liver failure with limited therapies. To date, only ursodeoxycholic acid (UDCA) has been approved by the U.S. Food and Drug Administration for the treatment of cholestasis. However, approximately one-third of patients with cholestasis are unresponsive to UDCA. Therefore, it is urgent to search for appropriate therapeutic agents for restoring stoppage status of the bile components to treat cholestasis. In this study, we investigated how the microbiome and transcriptome data sets correlated with each other to clarify the role of microbiome alterations in host metabolism. In combination, this research offers potential molecular biomarkers that should be validated for more accurate diagnosis of cholestasis and the clinical utilisation of gut microbiota as a target for treatment.
Collapse
|
19
|
Long R, Shi L, He P, Tian J, Wang S, Zheng J. 3D cell culture based on artificial cells and hydrogel under microgravity for bottom-up microtissue constructs. Front Bioeng Biotechnol 2022; 10:1056652. [DOI: 10.3389/fbioe.2022.1056652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
The use of hydrogel as a filling medium to recombine dispersed microencapsulated cells to form an embedded gel-cell microcapsule complex is a new idea based on bottom-up tissue construction, which is benefit for cell distribution and of great significance for tissue construction research in vitro. In this experiment, sodium alginate and chitosan were used as the main materials, rat normal liver cell BRL-3A was used as the model cell to prepare “artificial cells”. Silkworm pupa was used as raw material to extract silk fibroin solution, which was prepared by ultrasound to be the silk fibroin gel; silk fibroin hydrogel-microencapsulated hepatocyte embedded complex was then prepared by using silk fibroin gel as filling medium; the complex was cultured under three modes (static, shaking, and 3D microgravity), and the tissue forming ability of rat hepatocytes was investigated. The results showed that the microgravity culture condition can enhance the cell proliferation and promote the formation of cell colonies in the microcapsules; silk fibroin can form an embedded gel-cell microcapsule complex with microencapsulated cells, which provided mechanical support for the structure of the composite. We hope that this bottom-up construction system will have potential applications in the fields of cell culture and tissue construction.
Collapse
|
20
|
Gao J, Sadiq FA, Zheng Y, Zhao J, He G, Sang Y. Biofilm-based delivery approaches and specific enrichment strategies of probiotics in the human gut. Gut Microbes 2022; 14:2126274. [PMID: 36175161 PMCID: PMC9542427 DOI: 10.1080/19490976.2022.2126274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The use of probiotics has been one of the effective strategies to restructure perturbed human gut microbiota following a disease or metabolic disorder. One of the biggest challenges associated with the use of probiotic-based gut modulation strategies is to keep the probiotic cells viable and stable during the gastrointestinal transit. Biofilm-based probiotics delivery approaches have emerged as fascinating modes of probiotic delivery in which probiotics show significantly greater tolerance and biotherapeutic potential, and interestingly probiotic biofilms can be developed on food-grade surfaces too, which is ideal for the growth and proliferation of bacterial cells for incorporation into food matrices. In addition, biofilms can be further encapsulated with food-grade materials or with bacterial self-produced biofilms. This review presents a newly emerging and unprecedently discussed techniques for the safe delivery of probiotics based on biofilms and further discusses newly emerging prebiotic materials which target specific gut microbiota groups for growth and proliferation.
Collapse
Affiliation(s)
- Jie Gao
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Faizan Ahmed Sadiq
- Flanders Research Institute for Agriculture, Fisheries and Food (ILVO), Technology & Food Sciences Unit, Melle, Belgium
| | - Yixin Zheng
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Jinrong Zhao
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Guoqing He
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China,CONTACT Guoqing He College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Yaxin Sang
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China,Yaxin Sang Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| |
Collapse
|
21
|
Deng L, Yang Y, Xu G. Empagliflozin ameliorates type 2 diabetes mellitus-related diabetic nephropathy via altering the gut microbiota. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159234. [PMID: 36185030 DOI: 10.1016/j.bbalip.2022.159234] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/22/2022] [Accepted: 09/04/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND The dysregulation of gut microbiota can be found in patients with type 2 diabetes mellitus (T2DM)-related diabetic nephropathy (DN). Inhibitors of sodium-glucose co-transporter 2 (SGLT2) were reported to affect gut microbiota. This study aimed to identify whether empagliflozin (EMPA) attenuated DN via regulating gut microbiota. MATERIALS AND METHODS The high-fat diet (HFD) combining streptozocin (STZ) injection was performed to induce DN in mice. The therapeutic effects of EMPA were observed by staining of renal tissues and urine albumin/creatinine ratio (UACR). Mouse feces were collected for 16S rRNA sequencing. Fecal short-chain fatty acids (SCFAs) and fecal and serum lipopolysaccharide (LPS) were determined. An antibiotic-ablated model was established to confirm the role of the gut microbiota in the actions of EMPA. RESULTS EMPA reduced the elevation of blood glucose and UACR caused by HFD/STZ. It inhibited the thickening of the colonic crypt and restored goblet cells and the expressions of ZO-1 and Occludin. The 16S rRNA sequencing showed that the diversity of gut microbiota was reduced after HFD/STZ treatment, while it was restored after EMPA treatment. The LPS-producing bacteria, Oscillibacter, and the SCFA-producing bacteria, Bateroid and Odoribacter, were changed after EMPA administration. The therapeutic effects of EMPA on ABX-treated mice were reduced. Meanwhile, the level of fecal SCFAs was decreased, while the levels of fecal and serum LPS were elevated, in T2DM mice, and they were negated by the administration of EMPA. CONCLUSION EMPA ameliorates T2DM-related DN via altering the gut microbiota, especially reducing LPS-producing bacteria and increasing SCFA-producing bacteria.
Collapse
Affiliation(s)
- Le Deng
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Jiangxi 330006, China
| | - Yang Yang
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Jiangxi 330006, China
| | - Gaosi Xu
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Jiangxi 330006, China.
| |
Collapse
|
22
|
Talebian S, Schofield T, Valtchev P, Schindeler A, Kavanagh JM, Adil Q, Dehghani F. Biopolymer-Based Multilayer Microparticles for Probiotic Delivery to Colon. Adv Healthc Mater 2022; 11:e2102487. [PMID: 35189037 PMCID: PMC11468821 DOI: 10.1002/adhm.202102487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/16/2022] [Indexed: 11/06/2022]
Abstract
The potential health benefits of probiotics may not be realized because of the substantial reduction in their viability during food storage and gastrointestinal transit. Microencapsulation has been successfully utilized to improve the resistance of probiotics to critical conditions. Owing to the unique properties of biopolymers, they have been prevalently used for microencapsulation of probiotics. However, majority of microencapsulated products only contain a single layer of protection around probiotics, which is likely to be inferior to more sophisticated approaches. This review discusses emerging methods for the multilayer encapsulation of probiotic using biopolymers. Correlations are drawn between fabrication techniques and the resultant microparticle properties. Subsequently, multilayer microparticles are categorized based on their layer designs. Recent reports of specific biopolymeric formulations are examined regarding their physical and biological properties. In particular, animal models of gastrointestinal transit and disease are highlighted, with respect to trials of multilayer microencapsulated probiotics. To conclude, novel materials and approaches for fabrication of multilayer structures are highlighted.
Collapse
Affiliation(s)
- Sepehr Talebian
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - Timothy Schofield
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
| | - Peter Valtchev
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Centre for Advanced Food EngineeringThe University of SydneySydneyNSW2006Australia
| | - Aaron Schindeler
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Centre for Advanced Food EngineeringThe University of SydneySydneyNSW2006Australia
- Bioengineering & Molecular Medicine LaboratoryThe Children's Hospital at Westmead and the Westmead Institute for Medical ResearchWestmeadNSW2145Australia
| | - John M. Kavanagh
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
| | - Qayyum Adil
- PharmaCare Laboratories18 Jubilee AveWarriewoodNSW2102Australia
| | - Fariba Dehghani
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Centre for Advanced Food EngineeringThe University of SydneySydneyNSW2006Australia
| |
Collapse
|
23
|
Influence of Pholiota adiposa on gut microbiota and promote tumor cell apoptosis properties in H22 tumor-bearing mice. Sci Rep 2022; 12:8589. [PMID: 35597811 PMCID: PMC9124200 DOI: 10.1038/s41598-022-11041-x] [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: 11/12/2021] [Accepted: 04/06/2022] [Indexed: 11/08/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a common type of cancer-prevalent worldwide-and one of the causes of cancer-related deaths. In this study, ethanol extracts from Pholiota adiposa (EPA) were used to identify possible targets for HCC treatment and their effects on intestinal microflora were analyzed. Methods: Male mice were randomly assigned to groups-the model group, cyclophosphamide (25 mg/kg/d), and EPA groups, in which the mice were categorized based on the different concentrations of each compound (100, 200, and 300 mg/kg/day). Relevant biochemical indicators were detected using ELISA, H&E staining, and TUNEL assay. Four tumor apoptosis-related proteins and genes, Cleaved Caspases, BAX, Bcl-2, and VEGF, were detected by immunohistochemical staining, western blotting, and RT-PCR. The total genomic DNA was obtained from the contents of the small intestine and colon and was sequenced. The V3 + V4 regions of bacterial 16 s rDNA (from 341 to 806) were amplified. Results: The tests revealed that EPA exhibited antitumor activity in vivo by promoting apoptosis and inhibiting angiogenesis. Moreover, EPA treatment could increase beneficial and decrease harmful microflorae. These results demonstrate that EPA may be a potential therapy for HCC.
Collapse
|
24
|
Proteomics analysis of the hypothalamus of high-fat diet fed mice after Lactiplantibacillus plantarum Y44 administration. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
25
|
Curcumin Supplementation Ameliorates Bile Cholesterol Supersaturation in Hamsters by Modulating Gut Microbiota and Cholesterol Absorption. Nutrients 2022; 14:nu14091828. [PMID: 35565795 PMCID: PMC9100705 DOI: 10.3390/nu14091828] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 01/27/2023] Open
Abstract
Curcumin is a polyphenol that has been shown to have prebiotic and cholesterol-lowering properties. This study aimed to investigate the impact of curcumin on bile cholesterol supersaturation and the potential mechanistic role of intestinal microbiota and cholesterol absorption. Male hamsters (n = 8) were fed a high-fat diet (HFD) supplemented with or without curcumin for 12 weeks. Results showed that curcumin significantly decreased cholesterol levels in the serum (from 5.10 to 4.10 mmol/L) and liver (from 64.60 to 47.72 nmol/mg protein) in HFD-fed hamsters and reduced the bile cholesterol saturation index (CSI) from 1.64 to 1.08 due to the beneficial modifications in the concentration of total bile acids (BAs), phospholipids and cholesterol (p < 0.05). Gut microbiota analysis via 16S rRNA sequencing revealed that curcumin modulated gut microbiota, predominantly increasing microbiota associated with BA metabolism and short-chain fatty acid production, which subsequently up-regulated the expression of hepatic cholesterol 7-alpha hydroxylase and increased the synthesis of bile acids (p < 0.05). Furthermore, curcumin significantly down-regulated the expression of intestinal Niemann−Pick C1-like protein 1(NPC1L1) in hamsters and reduced cholesterol absorption in Caco-2 cells (p < 0.05). Our results demonstrate that dietary curcumin has the potential to prevent bile cholesterol supersaturation through modulating the gut microbiota and inhibiting intestinal cholesterol absorption.
Collapse
|
26
|
Wang Y, Xu Y, Xu X, Wang H, Wang D, Yan W, Zhu J, Hao H, Wang G, Cao L, Zhang J. Ginkgo biloba extract ameliorates atherosclerosis via rebalancing gut flora and microbial metabolism. Phytother Res 2022; 36:2463-2480. [PMID: 35312112 DOI: 10.1002/ptr.7439] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/17/2022] [Accepted: 02/24/2022] [Indexed: 12/31/2022]
Abstract
The Ginkgo biloba leave extract (GbE) is widely applied in the prevention and treatment of atherosclerotic cardiovascular diseases in clinical practice. However, its mechanism of actions has not been totally elucidated. In this study, we confirmed the beneficial effects of GbE in alleviating hypercholesterolemia, inflammation and atherosclerosis in Ldlr-/- mice, which were fed 12 weeks of Western diet (WD). Moreover, 16S rRNA sequencing revealed that GbE treatment reshaped the WD-perturbed intestinal microbiota, particularly decreased the Firmicutes/Bacteroidetes ratio and elevated the abundance of Akkermansia, Alloprevotella, Alistipes, and Parabacteroides. Furthermore, GbE treatment downregulated the intestinal transcriptional levels of proinflammatory cytokines and enhanced the expression of tight junction proteins, exerting the roles of attenuating the intestinal inflammation as well as repairing the gut barrier. Meanwhile, the targeted metabolomic analysis displayed that GbE treatment significantly reversed the dysfunction of the microbial metabolic phenotypes, including promoting the production of short chain fatty acids, indole-3-acetate and secondary bile acids, which were correlated with the atherosclerotic plaque areas. Finally, we confirmed GbE-altered gut microbiota was sufficient to alleviate atherosclerosis by fecal microbiota transplantation. In summary, our findings provide important insights into the pharmacological mechanism underlying the antiatherogenic efficacy of GbE.
Collapse
Affiliation(s)
- Yun Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Yuanyuan Xu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Xiaowei Xu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Hong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Dong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Wenchao Yan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Jiaying Zhu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Lijuan Cao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Jun Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China.,School of Pharmacy, Nanjing Medical University, Nanjing, China
| |
Collapse
|
27
|
Zhang P, Lu G, Sun Y, Yan Z, Dang T, Liu J. Metagenomic analysis explores the interaction of aged microplastics and roxithromycin on gut microbiota and antibiotic resistance genes of Carassius auratus. JOURNAL OF HAZARDOUS MATERIALS 2022; 425:127773. [PMID: 34802820 DOI: 10.1016/j.jhazmat.2021.127773] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/02/2021] [Accepted: 11/10/2021] [Indexed: 06/13/2023]
Abstract
The aging process changes the physicochemical structure of microplastics and affects environmental behaviors and toxicological effects of coexisting pollutants, thereby posing ecological risks. In this study, the effects of aged polystyrene microplastics alone or in combination with the 100 μg/L roxithromycin (ROX) on intestines of Carassius auratus were investigated. The carrier effect of microplastics was enhanced by aging due to changes in functional groups and surface area, which led to an increase in the bioaccumulation of ROX. The combined exposure of aged microplastics (APS) and ROX caused more inflammatory cell infiltration and cilia defects, and significantly inhibited the activity of amylase and lipase. Metagenomic sequencing revealed that the combined exposure of microplastics and ROX increased the abundance of Gemmobacter, Bosea, Rhizobium, and Shinella and decreased the abundance of Cetobacterium and Akkermansia (p < 0.05). The presence of APS enhanced the selective enrichment of antibiotic resistance genes. What's more, the influence of microplastics and antibiotics on gut microbiota was closely related to carbohydrate metabolism and amino acid metabolism activities, as well as the abundance of baca and sul1 resistance genes. These results expand our understanding of the interaction mechanism between APS and antibiotics in real aquatic environment.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Integrated Regulation and Resources Development of Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| | - Guanghua Lu
- Key Laboratory of Integrated Regulation and Resources Development of Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China.
| | - Yu Sun
- Key Laboratory of Integrated Regulation and Resources Development of Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| | - Zhenhua Yan
- Key Laboratory of Integrated Regulation and Resources Development of Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| | - Tianjian Dang
- Key Laboratory of Integrated Regulation and Resources Development of Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| | - Jianchao Liu
- Key Laboratory of Integrated Regulation and Resources Development of Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| |
Collapse
|
28
|
Li Y, Hu H, Yang H, Lin A, Xia H, Cheng X, Kong M, Liu H. Vine Tea (
Ampelopsis grossedentata
) extract attenuates CCl
4
‐induced liver injury by restoring gut microbiota dysbiosis in mice. Mol Nutr Food Res 2022; 66:e2100892. [PMID: 35188709 DOI: 10.1002/mnfr.202100892] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/29/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Ying Li
- College of Basic Medical Sciences Hubei University of Chinese Medicine Huangjiahu West Road 16 Wuhan 430065 PR China
| | - Haiming Hu
- College of Basic Medical Sciences Hubei University of Chinese Medicine Huangjiahu West Road 16 Wuhan 430065 PR China
| | - Huabing Yang
- College of Basic Medical Sciences Hubei University of Chinese Medicine Huangjiahu West Road 16 Wuhan 430065 PR China
| | - Aizhen Lin
- Hubei Provincial Hospital of Traditional Chinese Medicine Wuhan 430061 P.R. China
- Hubei Province Academy of Traditional Chinese Medicine Wuhan 430074 P.R. China
| | - Hui Xia
- College of Basic Medical Sciences Hubei University of Chinese Medicine Huangjiahu West Road 16 Wuhan 430065 PR China
| | - Xue Cheng
- College of Basic Medical Sciences Hubei University of Chinese Medicine Huangjiahu West Road 16 Wuhan 430065 PR China
| | - Mingwang Kong
- College of Basic Medical Sciences Hubei University of Chinese Medicine Huangjiahu West Road 16 Wuhan 430065 PR China
| | - Hongtao Liu
- College of Basic Medical Sciences Hubei University of Chinese Medicine Huangjiahu West Road 16 Wuhan 430065 PR China
| |
Collapse
|
29
|
Yu X, Jin Y, Zhou W, Xiao T, Wu Z, Su J, Gao H, Shen P, Zheng B, Luo Q, Li L, Xiao Y. Rifaximin Modulates the Gut Microbiota to Prevent Hepatic Encephalopathy in Liver Cirrhosis Without Impacting the Resistome. Front Cell Infect Microbiol 2022; 11:761192. [PMID: 35118004 PMCID: PMC8804384 DOI: 10.3389/fcimb.2021.761192] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/22/2021] [Indexed: 12/21/2022] Open
Abstract
The gut microbiota has an important role in the pathogenesis of hepatic encephalopathy(HE). Rifaximin, an intestinal non-absorbable antibacterial agent, is effective in the treatment of HE. However, whether long-term prophylactic use induces antibacterial resistance and its mechanism for treating HE remains unclear. This prospective study assessed the impact of 12 weeks rifaximin administration on the gut microbiota and resistome in cirrhotic patients. Fecal sampling was conducted 1 day before the first rifaximin administration and at Weeks 1, 2, 4, 6, 8, 10, 12 of the study. Thirty cirrhotic patients who were in remission from recurrent HE was enrolled to receive rifaximin (400mg TID for 12 weeks). Rifaximin improved hyperammonemia and cognitive function in the 21 patients who completed rifaximin treatment. The dynamic observations showed the gut microbiota diversity, composition and the number of resistance genes, plasmids, insertion sequences did not change significantly during the period(P>0.05). Metabolic pathways such as aromatic amino acids, tryptophan synthesis, urea cycle, and LPS synthesis reduced. No new antimicrobial resistance genes was emergenced. However, the number of aminoglycosides, rifamycin and phenolic resistance genes increased, whereas tetracycline, fosfomycin and cephamycin decreased (P<0.05). Changes in the abundance of E. coli, K. pneumoniae, and B. longum strains correlated with changes of resistance genes. Prophylactic use of rifaximin for 12 weeks improved hyperammonemia and neurophysiological function, maintained gut microbiota diversity, composition and did not change the overall resistome. Rifaximin altered expression of HE-related metabolic pathways. All of these effects could play a key role in preventing HE. Clinical Trial Registration: ChiCTR1900022234 (registered at the Chinese Clinical Trial Registry).
Collapse
Affiliation(s)
- Xiao Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Respiratory and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ye Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wangxiao Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tingting Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhongwen Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junwei Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hainv Gao
- Department of Infectious Disease, ShuLan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Ping Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Beiwen Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qixia Luo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yonghong Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Yonghong Xiao,
| |
Collapse
|
30
|
Hu B, Li M, He X, Wang H, Huang JA, Liu Z, Mezzenga R. Flavonoid-Amyloid Fibril Hybrid Hydrogels for Obesity Control via Construction of Gut Microbiota. Biomater Sci 2022; 10:3597-3611. [DOI: 10.1039/d2bm00366j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Innovative precise clinical approaches to protect humans from the alarming global growth of epidemics of chronic diseases, such as metabolic syndrome (MetS), are urgently needed. Here, we introduce protein hydrogels...
Collapse
|
31
|
Wang X, Ye P, Fang L, Ge S, Huang F, Polverini PJ, Heng W, Zheng L, Hu Q, Yan F, Wang W. Active Smoking Induces Aberrations in Digestive Tract Microbiota of Rats. Front Cell Infect Microbiol 2021; 11:737204. [PMID: 34917518 PMCID: PMC8668415 DOI: 10.3389/fcimb.2021.737204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
Cigarette smoking could have certain effects on gut microbiota. Some pioneering studies have investigated effects of active smoking on the microbiome in local segments of the digestive tract, while active smoking-induced microbiome alterations in the whole digestive tract have not been fully investigated. Here, we developed a rat model of active smoking and characterized the effects of active smoking on the microbiota within multiple regions along the digestive tract. Blood glucose and some metabolic factors levels, the microbial diversity and composition, relative abundances of taxa, bacterial network correlations and predictive functional profiles were compared between the control group and active smoking group. We found that active smoking induced hyperglycemia and significant reductions in serum insulin and leptin levels. Active smoking induced region-specific shifts in microbiota structure, composition, network correlation and metabolism function along the digestive tract. Our results demonstrated that active smoking resulted in a reduced abundance of some potentially beneficial genera (i.e. Clostridium, Turicibacter) and increased abundance of potentially harmful genera (i.e. Desulfovibrio, Bilophila). Functional prediction suggested that amino acid, lipid, propanoate metabolism function could be impaired and antioxidant activity may be triggered. Active smoking may be an overlooked risk to health through its potential effects on the digestive tract microbiota, which is involved in the cause and severity of an array of chronic diseases.
Collapse
Affiliation(s)
- Xiang Wang
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Periodontics & Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Pei Ye
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Li Fang
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Sheng Ge
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Fan Huang
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Peter J Polverini
- Department of Periodontics & Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Weiwei Heng
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lichun Zheng
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qingang Hu
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Fuhua Yan
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenmei Wang
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
32
|
Pan X, Kaminga AC, Liu A, Wen SW, Luo M, Luo J. Gut Microbiota, Glucose, Lipid, and Water-Electrolyte Metabolism in Children With Nonalcoholic Fatty Liver Disease. Front Cell Infect Microbiol 2021; 11:683743. [PMID: 34778099 PMCID: PMC8581616 DOI: 10.3389/fcimb.2021.683743] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 09/09/2021] [Indexed: 12/15/2022] Open
Abstract
There is evidence that nonalcoholic fatty liver disease (NAFLD) is affected by gut microbiota, glucose, and lipid. However, the function of water-electrolyte metabolism remains undefined in children with NAFLD. Therefore, the aim of this case-control study was to better understand these interactions. The sample consisted of 75 children, aged between 7 and 16, of whom 25 had nonalcoholic fatty liver (NAFL), 25 had nonalcoholic steatohepatitis (NASH), and 25 were obese and without NAFLD. These groups were matched by age, sex, and body mass index. Data were collected between June, 2019 and December, 2019 at the Hunan Children’s Hospital, in China. Microbiome composition in fecal samples was assessed using 16S ribosomal RNA amplicon sequencing. In the clinical indices, 12 glucose and lipid metabolism indices were included, and six water-electrolyte metabolism indices were included. The results indicated that microbiomes of NAFLD children had lower alpha diversity but higher beta diversity index than the other two groups. Specifically, anti-inflammatory and probiotics abundance (e.g., Faecalibacterium, Akkermansia, and Bifidobacterium_adolescentis) was significantly decreased in NAFLD, whereas the abundance of harmful bacteria (e.g., Staphylococcaceae) was increased. Moreover, the abundance of butyrate-producing bacteria (e.g., Faecalibacterium, Roseburia_inulinivorans, Roseburia_intestinalis, and Coprococcus_comes) was significantly decreased in NASH. The abundance of these bacteria were associated with glucose, lipid, and water-electrolyte metabolism (e.g., glucose, triglyceride, cholesterol, inorganic salt, total body water, etc.), implying that the NAFLD and its severity were associated with glucose, lipid, and water-electrolyte metabolism dysbiosis. Therefore, these findings suggest that the gut microbiome, especially butyrate-producing bacteria, play an important role in the development of NAFLD in children.
Collapse
Affiliation(s)
- Xiongfeng Pan
- Xiangya School of Public Health, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, China
| | - Atipatsa C Kaminga
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, China.,Department of Mathematics and Statistics, Mzuzu University, Mzuzu, Malawi
| | - Aizhong Liu
- Xiangya School of Public Health, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, China
| | - Shi Wu Wen
- OMNI Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Obstetrics and Gynaecology University of Ottawa Faculty of Medicine, Ottawa, ON, Canada.,School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
| | - Miyang Luo
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Jiayou Luo
- Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
33
|
Zhuang P, Li H, Jia W, Shou Q, Zhu Y, Mao L, Wang W, Wu F, Chen X, Wan X, Wu Y, Liu X, Li Y, Zhu F, He L, Chen J, Zhang Y, Jiao J. Eicosapentaenoic and docosahexaenoic acids attenuate hyperglycemia through the microbiome-gut-organs axis in db/db mice. MICROBIOME 2021; 9:185. [PMID: 34507608 PMCID: PMC8434703 DOI: 10.1186/s40168-021-01126-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/08/2021] [Indexed: 05/04/2023]
Abstract
BACKGROUND Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have been suggested to prevent the development of metabolic disorders. However, their individual role in treating hyperglycemia and the mechanism of action regarding gut microbiome and metabolome in the context of diabetes remain unclear. RESULTS Supplementation of DHA and EPA attenuated hyperglycemia and insulin resistance without changing body weight in db/db mice while the ameliorative effect appeared to be more pronounced for EPA. DHA/EPA supplementation reduced the abundance of the lipopolysaccharide-containing Enterobacteriaceae whereas elevated the family Coriobacteriaceae negatively correlated with glutamate level, genera Barnesiella and Clostridium XlVa associated with bile acids production, beneficial Bifidobacterium and Lactobacillus, and SCFA-producing species. The gut microbiome alterations co-occurred with the shifts in the metabolome, including glutamate, bile acids, propionic/butyric acid, and lipopolysaccharide, which subsequently relieved β cell apoptosis, suppressed hepatic gluconeogenesis, and promoted GLP-1 secretion, white adipose beiging, and insulin signaling. All these changes appeared to be more evident for EPA. Furthermore, transplantation with DHA/EPA-mediated gut microbiota mimicked the ameliorative effect of DHA/EPA on glucose homeostasis in db/db mice, together with similar changes in gut metabolites. In vitro, DHA/EPA treatment directly inhibited the growth of Escherichia coli (Family Enterobacteriaceae) while promoted Coriobacterium glomerans (Family Coriobacteriaceae), demonstrating a causal effect of DHA/EPA on featured gut microbiota. CONCLUSIONS DHA and EPA dramatically attenuated hyperglycemia and insulin resistance in db/db mice, which was mediated by alterations in gut microbiome and metabolites linking gut to adipose, liver and pancreas. These findings shed light into the gut-organs axis as a promising target for restoring glucose homeostasis and also suggest a better therapeutic effect of EPA for treating diabetes. Video abstract.
Collapse
Affiliation(s)
- Pan Zhuang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Haoyu Li
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Wei Jia
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Qiyang Shou
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310005, Zhejiang, China
| | - Ya'er Zhu
- Analysis Center of Agrobiology and Environmental Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Lei Mao
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Wenqiao Wang
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Fei Wu
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Xiaoqian Chen
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Xuzhi Wan
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yuqi Wu
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Xiaohui Liu
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Yin Li
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Fanghuan Zhu
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Lilin He
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Jingnan Chen
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yu Zhang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Jingjing Jiao
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
34
|
Jia X, Xu W, Zhang L, Li X, Wang R, Wu S. Impact of Gut Microbiota and Microbiota-Related Metabolites on Hyperlipidemia. Front Cell Infect Microbiol 2021; 11:634780. [PMID: 34490132 PMCID: PMC8417472 DOI: 10.3389/fcimb.2021.634780] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Hyperlipidemia, defined as the presence of excess fat or lipids in the blood, has been considered as a high-risk factor and key indicator of many metabolic diseases. The gut microbiota has been reported playing a vital role in regulating host lipid metabolism. The pathogenic role of gut microbiota in the development of hyperlipidemia has been revealed through fecal microbiota transplantation experiment to germ-free mice. The effector mechanism of microbiota-related metabolites such as bile acids, lipopolysaccharide, and short-chain fatty acids in the regulation of hyperlipidemia has been partially unveiled. Moreover, studies on gut-microbiota-targeted hyperlipidemia interventions, including the use of prebiotics, probiotics, fecal microbiota transplantation, and natural herbal medicines, also have shown their efficacy in the treatment of hyperlipidemia. In this review, we summarize the relationship between gut microbiota and hyperlipidemia, the impact of gut microbiota and microbiota-related metabolites on the development and progression of hyperlipidemia, and the potential therapeutic management of hyperlipidemia targeted at gut microbiota.
Collapse
Affiliation(s)
- Xiaokang Jia
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Wen Xu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lei Zhang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoyan Li
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Ruirui Wang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuisheng Wu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
35
|
Jiang J, Xiong J, Ni J, Chen C, Wang K. Live Combined B. subtilis and E. faecium Alleviate Liver Inflammation, Improve Intestinal Barrier Function, and Modulate Gut Microbiota in Mice with Non-Alcoholic Fatty Liver Disease. Med Sci Monit 2021; 27:e931143. [PMID: 34482357 PMCID: PMC8428156 DOI: 10.12659/msm.931143] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a chronic, progressive liver disease with an increasing incidence rate. This study investigated the protective effects of live combined Bacillus subtilis and Enterococcus faecium (LCBE) on NAFLD, and its possible mechanisms. Material/Methods Five-week-old C57BL/6 mice were randomly divided into 3 groups: chow, HFD, and HFD+LCBE groups. The levels of serum biochemical markers, glucose tolerance, insulin, the inflammatory cytokines IL-1β, IL-6, and TNF-α, LPS, and histological staining were measured using commercial kits. qPCR was used to examine the mRNA expression levels of inflammatory cytokines in the liver. Western blotting was used to determine the protein levels of TLR4, NF-κB p65, PPAR-α, and CPT-1 in the liver, and occludin and Claudin1 in the intestine. The intestinal flora of the mice was analyzed by high-throughput sequencing of the V3–V4 region of 16S rDNA. Results LCBE significantly lowered the body weight, liver/body weight ratio, and serum glucose level, and increased the serum insulin level in NAFLD mice. In addition, LCBE treatment improved the liver function and lipid profile, decreased the levels of LPS and inflammatory cytokines, and downregulated the expression of TLR4 and NF-κB p65. Moreover, LCBE enhanced the intestinal barrier function by increasing the expression of occludin and Claudin1. Furthermore, LCBE modulated the composition of the gut microbiota by reducing the Firmicutes to Bacteroidetes ratio, and the proportion of inflammation-related and LPS-producing bacteria, thus re-arranging the structure of the gut microbiota. Conclusions LCBE protects against NAFLD by alleviating inflammation, restoring the intestinal barrier, and modulating gut microbiota composition.
Collapse
Affiliation(s)
- Jie Jiang
- Department of Gastroenterology and Hepatology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Jie Xiong
- Department of Gastroenterology and Hepatology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Jianbo Ni
- Department of Gastroenterology and Hepatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Congying Chen
- Department of Gastroenterology and Hepatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Kezhou Wang
- Department of Pathology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| |
Collapse
|
36
|
Mo J, Gao L, Zhang N, Xie J, Li D, Shan T, Fan L. Structural and quantitative alterations of gut microbiota in experimental small bowel obstruction. PLoS One 2021; 16:e0255651. [PMID: 34347831 PMCID: PMC8336877 DOI: 10.1371/journal.pone.0255651] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 07/21/2021] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE To investigate structural and quantitative alterations of gut microbiota in an experimental model of small bowel obstruction. METHOD A rat model of small bowel obstruction was established by using a polyvinyl chloride ring surgically placed surrounding the terminal ileum. The alterations of gut microbiota were studied after intestinal obstruction. Intraluminal fecal samples proximal to the obstruction were collected at different time points (24, 48 and 72 hours after obstruction) and analyzed by 16s rDNA high-throughput sequencing technology and quantitative PCR (qPCR) for target bacterial groups. Furthermore, intestinal claudin-1 mRNA expression was examined by real-time polymerase chain reaction analysis, and serum sIgA, IFABP and TFF3 levels were determined by enzyme-linked immunosorbent assay. RESULTS Small bowel obstruction led to significant bacterial overgrowth and profound alterations in gut microbiota composition and diversity. At the phylum level, the 16S rDNA sequences showed a marked decrease in the relative abundance of Firmicutes and increased abundance of Proteobacteria, Verrucomicrobia and Bacteroidetes. The qPCR analysis showed the absolute quantity of total bacteria increased significantly within 24 hours but did not change distinctly from 24 to 72 hours. Further indicators of intestinal mucosa damage and were observed as claudin-1 gene expression, sIgA and TFF3 levels decreased and IFABP level increased with prolonged obstruction. CONCLUSION Small bowel obstruction can cause significant structural and quantitative alterations of gut microbiota and induce disruption of gut mucosa barrier.
Collapse
MESH Headings
- Animals
- Bacteroidetes/genetics
- Claudin-1/genetics
- DNA, Bacterial/genetics
- DNA, Bacterial/isolation & purification
- DNA, Ribosomal/genetics
- DNA, Ribosomal/isolation & purification
- Disease Models, Animal
- Feces/microbiology
- Firmicutes/genetics
- Gastrointestinal Microbiome/genetics
- Gene Expression
- Ileum/microbiology
- Ileum/pathology
- Immunoglobulin A, Secretory/blood
- Immunoglobulin A, Secretory/metabolism
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/microbiology
- Intestinal Obstruction/blood
- Intestinal Obstruction/microbiology
- Male
- Phylogeny
- Proteobacteria/genetics
- RNA, Ribosomal, 16S/genetics
- Rats
- Rats, Wistar
- Verrucomicrobia/genetics
Collapse
Affiliation(s)
- Jiali Mo
- Graduate school of Tianjin Medical University, Tianjin, China
| | - Lei Gao
- Graduate school of Tianjin Medical University, Tianjin, China
| | - Nan Zhang
- Department of Gastrointestinal Surgery, Nankai Hospital, Tianjin, China
| | - Jiliang Xie
- Department of Gastrointestinal Surgery, Nankai Hospital, Tianjin, China
| | - Donghua Li
- Department of Pharmacology, Tianjin Nankai Hospital, Tianjin, China
| | - Tao Shan
- Department of Gastrointestinal Surgery, Nankai Hospital, Tianjin, China
| | - Liuyang Fan
- Graduate school of Tianjin Medical University, Tianjin, China
| |
Collapse
|
37
|
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
|
38
|
Zhang Y, Zhao M, Jiang X, Qiao Q, Liu T, Zhao C, Wang M. Comprehensive Analysis of Fecal Microbiome and Metabolomics in Hepatic Fibrosis Rats Reveal Hepatoprotective Effects of Yinchen Wuling Powder From the Host-Microbial Metabolic Axis. Front Pharmacol 2021; 12:713197. [PMID: 34385924 PMCID: PMC8353151 DOI: 10.3389/fphar.2021.713197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatic fibrosis (HF) is a typical consequence in the development of multiple chronic liver diseases, which is intimately related to the composition and metabolic status of gut microbiota. A myriad of evidence has indicated that traditional Chinese medicine can treat HF by regulating gut microbiota. Yinchen Wuling powder (YCWLP) is a famous traditional Chinese medicine prescription, which has been used to relieve liver diseases for thousands of years. YCWLP has demonstrated protective function on HF, but its effect on the alterations of gut microbiota is still unclear, and its explicit therapeutic mechanism also needs to be further elucidated. In this study, 16S rRNA gene sequencing and fecal metabolomics analysis were combined to investigate the influence of YCWLP on gut microbiota in HF rats and the interactions between gut microbiota and host metabolism. The results showed that YCWLP treatment significantly improved the disorder of multiple organ indices, HF-related cytokines and plasma LPS induced by HF. Masson's trichrome stainings also showed that YCWLP treatment could significantly alleviate the severity of HF in rats. Additionally, YCWLP could reverse the significant changes in the abundance of certain genera closely related to HF phenotype, including Barnesiella [Ruminococcus] and Christensenella. Meanwhile, YCWLP significantly increased the abundance of Bifidobacterium, Coprococcus and Anaerostipes, which are closely related to butyrate production. Metabolomics and Spearman's correlation analysis showed that YCWLP could regulate the disorder of arginine biosynthesis, sphingolipid metabolism and alanine, aspartate and glutamate metabolism in HF rats, and these regulations were intimately related to Barnesiella, [Ruminococcus], Christensenella, Coprococcus and Anaerostipes. By explaining the biological significance of the above results, we concluded that YCWLP might ameliorate HF by regulating the imbalance of gut microbiota, increasing the abundance of butyrate-producing bacteria to reduce ammonia production, promote ammonia degradation, and regulate pro-inflammatory cytokines and immune function.
Collapse
Affiliation(s)
- Yumeng Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Min Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xue Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Qiaoyu Qiao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Tingting Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Chunjie Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Miao Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
39
|
Tong T, Niu X, Li Q, Ling Y, Li Z, Liu J, Zhang M, Bai Z, Xia R, Wu Z, Liu X. The Effect of Lactobacillus plantarum BW2013 on The Gut Microbiota in Mice Analyzed by 16S rRNA Amplicon Sequencing. Pol J Microbiol 2021; 70:235-243. [PMID: 34349813 PMCID: PMC8326986 DOI: 10.33073/pjm-2021-022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022] Open
Abstract
Lactobacillus plantarum BW2013 was isolated from the fermented Chinese cabbage. This study aimed to test the effect of this strain on the gut microbiota in BALB/c mice by 16S rRNA amplicon sequencing. The mice were randomly allocated to the control group and three treatment groups of L. plantarum BW2013 (a low-dose group of 108 CFU/ml, a medium-dose group of 109 CFU/ml, and a high-dose group of 1010 CFU/ml). The weight of mice was recorded once a week, and the fecal samples were collected for 16S rRNA amplicon sequencing after 28 days of continuous treatment. Compared with the control group, the body weight gain in the treatment groups was not significant. The 16S rRNA amplicon sequencing analysis showed that both the Chao1 and ACE indexes increased slightly in the medium-dose group compared to the control group, but the difference was not significant. Based on PCoA results, there was no significant difference in β diversity between the treatment groups. Compared to the control group, the abundance of Bacteroidetes increased in the low-dose group. The abundance of Firmicutes increased in the medium-dose group. At the genus level, the abundance of Alloprevotella increased in the low-dose group compared to the control group. The increased abundance of Ruminococcaceae and decreased abundance of Candidatus_Saccharimonas was observed in the medium-dose group. Additionally, the abundance of Bacteroides increased, and Alistipes and Candidatus_Saccharimonas decreased in the high-dose group. These results indicated that L. plantarum BW2013 could ameliorate gut microbiota composition, but its effects vary with the dose.
Collapse
Affiliation(s)
- Tong Tong
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Xiaohui Niu
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Qian Li
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Yuxi Ling
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Zuming Li
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Jia Liu
- Internal Trade Food Science and Technology (Beijing) Co., Ltd, Beijing, China
| | - Michael Zhang
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Zhihui Bai
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Ran Xia
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Zhichao Wu
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Xiu Liu
- China National Research Institute of Food and Fermentation Industies Co., Ltd, Beijing, China
| |
Collapse
|
40
|
Pang B, Jin H, Liao N, Li J, Jiang C, Shao D, Shi J. Lactobacillus rhamnosus from human breast milk ameliorates ulcerative colitis in mice via gut microbiota modulation. Food Funct 2021; 12:5171-5186. [PMID: 33977948 DOI: 10.1039/d0fo03479g] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Gut microbiota imbalance is one of the major causes of ulcerative colitis (UC). L. rhamnosus SHA113 (LRS), a strain isolated from healthy human milk, influences the regulation of gut flora. This study aims to determine whether this strain can ameliorate UC by modulating gut microbiota. Mouse models of UC were established using C57BL/6Cnc mice with intragastric administration of 3.0% (w/v) dextran sodium sulfate (DSS). LRS was used to treat the mouse models of UC with 109 cfu mL-1 cell suspension via intragastric administration. To verify the effect of gut microbiota on UC, fecal microbiota collected from the mice after the treatment with LRS were also used to treat the UC mouse models (FMT). The severity of UC was evaluated based on body weight, colon length, disease activity index (DAI), and hematoxylin-eosin staining. The microbial composition was analyzed by 16S rRNA sequencing. The mRNA expression levels of cytokines, mucins, tight junction proteins, and antimicrobial peptides in the gastrointestinal tract were detected by quantitative real-time polymerase chain reaction. The short-chain fatty acid (SCFAs) in the cecal contents of all mice were quantitatively detected by gas chromatography and mass spectrometry. Both LRS and FMT exerted excellent therapeutic effects on UC, as evidenced by the reduction in body weight loss, colon length, and colon structural integrity, as well as the increase in the DAI (disease activity index). LRS and FMT treatments showed similar effects: (1) an increase of total SCFA production in the cecal contents and the abundance of gut microbial diversity and flora composition; (2) decreases in two genera (Parabacteroides and Escherichia/Shigella) related to the DAI and the enhancement of SCFAs and IL-10 positively related genera in the gut microbiota (Bilophila, Roseburia, Akkermansia, and Bifidobacterium); (3) downregulation of the expression of tumor necrosis factor-α, interleukin IL-6, and IL-1β, and upregulation of the expression of the anti-inflammatory cytokine IL-10; and (4) upregulation of the expression of mucins (Muc1-4) and tight junction protein ZO-1. Overall, L. rhamnosus SHA113 relieves UC via the regulation of gut microbiota: increases in SCFA-producing genera and decreases in UC-related genera. In addition, a single strain is sufficient to induce a significant change in the gut microbiota and exert therapeutic effects on UC.
Collapse
Affiliation(s)
- Bing Pang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Han Jin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Ning Liao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Junjun Li
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Chunmei Jiang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Dongyan Shao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| | - Junling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China. NingLiao
| |
Collapse
|
41
|
Tang C, Kong L, Shan M, Lu Z, Lu Y. Protective and ameliorating effects of probiotics against diet-induced obesity: A review. Food Res Int 2021; 147:110490. [PMID: 34399486 DOI: 10.1016/j.foodres.2021.110490] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 05/05/2021] [Accepted: 05/23/2021] [Indexed: 02/07/2023]
Abstract
Diet-induced obesity is one of the major public health concerns all over the world, and obesity also contributes to the development of other chronic diseases such as non-alcoholic fatty acid liver disease, type 2 diabetes mellitus and cardiovascular diseases. Evidence shows that the pathogenesis of obesity and obesity-associated chronic diseases are closely related to dysregulation of lipid metabolism, glucose metabolism and cholesterol metabolism, and oxidative stress, endoplasmic reticulum stress, abnormal gut microbiome and chronic low-grade inflammation. Recently, in view of potential effects on lipid metabolism, glucose metabolism, cholesterol metabolism and intestinal microbiome, as well as anti-oxidative and anti-inflammatory activities, natural probiotics, including live and dead probiotics, and probiotic components and metabolites, have attracted increasing attention and are considered as novel strategies for preventing and ameliorating obesity and obesity-related chronic diseases. Specifically, this review is presented on the anti-obesity effects of probiotics and underlying molecular mechanisms, which will provide a theoretical basis of anti-obesity probiotics for the development of functional foods.
Collapse
Affiliation(s)
- Chao Tang
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Liangyu Kong
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Mengyuan Shan
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhaoxin Lu
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yingjian Lu
- College of Food Science & Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China.
| |
Collapse
|
42
|
Cunha LF, Ongaratto MA, Endres M, Barschak AG. Modelling hypercholesterolaemia in rats using high cholesterol diet. Int J Exp Pathol 2021; 102:74-79. [PMID: 33710712 PMCID: PMC7981591 DOI: 10.1111/iep.12387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/02/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022] Open
Abstract
Hypercholesterolaemia is a complex condition with multiple causes, including both lifestyle and genetic aspects. It is also a risk factor for cardiovascular diseases (CVDs), which are responsible for 172 million deaths/year. Although the reasons for hypercholesterolaemia are known, there are many critical questions that remain to be answered so that new therapeutics can be developed. In order to elucidate the pathobiology of this condition, animal models can mimic the pathology of human hypercholesterolaemia. One example of an animal model is induced by the hypercholesterolaemic diet in Wistar rats. The present review first summarizes the current understanding of the metabolic profile involved in hypercholesterolaemia in humans. Next it comments about the lack of consensus as to which hypercholesterolaemia induction protocol should be used. The present work aimed to review experimental studies that induced hypercholesterolaemia in Wistar rats it was not intended to judge the "best" model, since they all achieved the goal of inducing an increase in serum cholesterol.
Collapse
Affiliation(s)
- Luiza Ferracini Cunha
- Programa de Pós‐Graduação em Ciências da SaúdeUniversidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreBrazil
| | | | - Marcelo Endres
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreBrazil
| | - Alethea Gatto Barschak
- Programa de Pós‐Graduação em Ciências da SaúdeUniversidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreBrazil
| |
Collapse
|
43
|
Yu H, Fang C, Li P, Wu M, Shen S. The relevance of DHA with modulating of host-gut microbiome signatures alterations and repairing of lipids metabolism shifts. Eur J Pharmacol 2021; 895:173885. [PMID: 33482183 DOI: 10.1016/j.ejphar.2021.173885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022]
Abstract
Huge of previous reports recommended that gut microbiome have a crucial role in the human health and its change was profound impact for the metabolic improvements associated with lipids metabolism. In order to explore the relevance of a direct dysbiosis effect of gut microbiome on lipids metabolism shifts and repaired position of DHA, we built the animal model for the study with gut microbiome dysbiosis administrated by i.g. with CRO and intervened by DHA in the present work. Gut microbiome was analyzed by high throughput sequencing and bioinformatics analyses of bacteria. The composition of fatty acids and short chain fatty acids (SCFAs) were determined by gas chromatography. Blood lipids and bile acids were assayed by kit and UPLC-MS/MS, respectively. The expressions of enzymes of long chain fatty acid metabolism were analyzed by qRT-PCR. The results showed that gut microbiome dysbiosis caused lipid metabolism abnormal, and DHA was able to repair the lipids metabolism shifts resulted from gut microbiome dysbiosis. DHA could modulate host-gut microbiome signatures, improve concentrations of SCFAs, regulate fatty acids metabolism but modify bile acid profiles. In conclusion, we considered that DHA repaired lipid metabolism by modulating gut microbiome and regulating fatty acids metabolism pathway.
Collapse
Affiliation(s)
- Haining Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China.
| | - Chengjie Fang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Peng Li
- Department of Geratoloy, The Third People's Hospital of Hangzhou, Hangzhou, China
| | - Manman Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | | |
Collapse
|
44
|
Abstract
The substantial burden of colorectal cancer and its increasing trend in young adults highlight the importance of dietary and lifestyle modifications for improved cancer prevention and survivorship. In this chapter, we review the cutting-edge evidence for the interplay between diet/lifestyle and the gut microbiota in the incidence and prognosis of colorectal cancer. We focus on factors for which there are data supporting their importance for the gut microbiota and colorectal cancer, including excess body weight, fiber, red and processed meat, and coffee. We discuss the potential precision nutrition approaches for modifying and exploiting the gut microbiota for improved cancer prevention and survivorship.
Collapse
Affiliation(s)
- Kai Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
45
|
|
46
|
Zhang JM, Liu XY, Gu W, Xu HY, Jiao HC, Zhao JP, Wang XJ, Li HF, Lin H. Different effects of probiotics and antibiotics on the composition of microbiota, SCFAs concentrations and FFAR2/3 mRNA expression in broiler chickens. J Appl Microbiol 2021; 131:913-924. [PMID: 33263216 DOI: 10.1111/jam.14953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/26/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022]
Abstract
AIMS The aims of this study were to investigate the effects of probiotics and antibiotics on microbial composition, short chain fatty acids (SCFAs) concentration and free fatty acid receptor 2/3 (FFAR2/3) expression in boiler chickens. METHODS AND RESULTS A total of 150 1-day-old male broilers were randomly allocated into three groups, control (CON) group, probiotics (PB) group and antibiotics (ATB) group. Results indicated that PB improved the average body weight from 1 to 21 days and feed intake from 21 to 42 days (P < 0·05), while ATB improved the feed efficiency from 1 to 42 days (P < 0·05). Based on 16s rRNA sequencing, PB treatment increased the amount of kingdom bacteria, and the relative abundance of the main bacteria including acetate and butyrate producing bacteria of phylum Firmicutes, family Ruminococcaceae and genus Faecalibacterium. ATB treatment also increased the relative abundance of phylum Firmicutes, family Ruminococcaceae and Lachnospiraceae, however, it introduced some pathogenic bacteria, such as bacteria of family Rikenellaceae and Enterobacteriaceae. Gas chromatography and mass spectrometry (GC-MS) assay revealed that PB increased acetate and butyrate concentrations at both 21 and 42 days, and propionate at 42 days in the colorectum. Moreover qRT-PCR analysis showed PB treatment significantly activated the FFAR2/3 mRNA expressions. On the contrast, ATB treatment lowered the colorectal propionate at 21 days, and decreased acetate, propionate and butyrate concentrations at 42 days, accompanied with decreased FFAR2/3 mRNA expressions. CONCLUSIONS Compared to the CON birds, an enriched SCFAs producing bacteria with higher SCFAs contents and activated FFAR2/3 expressions are prominent features of PB birds. However, antibiotics treatment plays the reverse effect compared to PB treatment. SIGNIFICANCE AND IMPACT OF THE STUDY This study brings a significant idea that less SCFAs concentration may be another reason why the antibiotics inhibit the immune system development and immunity of the body.
Collapse
Affiliation(s)
- J-M Zhang
- College of Animal Science and Veterinary Medicine, Shandong Key Lab for Animal Biotechnology and Disease Control, Shandong Agricultural University, Tai'an, China.,Biological Research Institute, Shandong Key Laboratory of Animal Microecological agents, Shandong Baolai-leelai Bioengineering Co., Ltd, Tai'an, China
| | - X-Y Liu
- Shandong Institute of Scientific and Technical Information, Ji Nan, China
| | - W Gu
- Biological Research Institute, Shandong Key Laboratory of Animal Microecological agents, Shandong Baolai-leelai Bioengineering Co., Ltd, Tai'an, China
| | - H-Y Xu
- Biological Research Institute, Shandong Key Laboratory of Animal Microecological agents, Shandong Baolai-leelai Bioengineering Co., Ltd, Tai'an, China
| | - H-C Jiao
- College of Animal Science and Veterinary Medicine, Shandong Key Lab for Animal Biotechnology and Disease Control, Shandong Agricultural University, Tai'an, China
| | - J-P Zhao
- College of Animal Science and Veterinary Medicine, Shandong Key Lab for Animal Biotechnology and Disease Control, Shandong Agricultural University, Tai'an, China
| | - X-J Wang
- College of Animal Science and Veterinary Medicine, Shandong Key Lab for Animal Biotechnology and Disease Control, Shandong Agricultural University, Tai'an, China
| | - H-F Li
- College of Life Sciences, Shandong Agricultural University, Tai'an, China
| | - H Lin
- College of Animal Science and Veterinary Medicine, Shandong Key Lab for Animal Biotechnology and Disease Control, Shandong Agricultural University, Tai'an, China
| |
Collapse
|
47
|
Yu Y, Lu J, Sun L, Lyu X, Chang XY, Mi X, Hu MG, Wu C, Chen X. Akkermansia muciniphila: A potential novel mechanism of nuciferine to improve hyperlipidemia. Biomed Pharmacother 2021; 133:111014. [PMID: 33246225 DOI: 10.1016/j.biopha.2020.111014] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Intestinal microbiota is a novel drug target of metabolic diseases, especially for those with poor oral bioavailability. Nuciferine, with poor bioavailability, has an anti-hyperlipidemic effect at low dosages. PURPOSE In the present study, we aimed to explore the role of intestinal microbiota in the anti-hyperlipidemic function of nuciferine and identify the key bacterial targets that might confer the therapeutic actions. METHODS The contribution of gut microbes in the anti-hyperlipidemic effect of nuciferine was evaluated by conventional and antibiotic-established pseudo-sterile mice. Whole-metagenome shotgun sequencing was used to characterize the changes in microbial communities by various agents. RESULTS Nuciferine exhibited potent anti-hyperlipidemic and liver steatosis-alleviating effects at the doses of 7.5-30 mg/kg. The beneficial effects of nuciferine were substantially abolished when combined with antibiotics. Metagenomic analysis showed that nuciferine significantly shifted the microbial structure, and the enrichment of Akkermansia muciniphila was closely related to the therapeutic effect of nuciferine. CONCLUSIONS Our results revealed that gut microbiota played an essential role in the anti-hyperlipidemic effect of nuciferine, and enrichment of Akkermansia muciniphila represented a key mechanism through which nuciferine exerted its therapeutic effects.
Collapse
Affiliation(s)
- Yue Yu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China
| | - Juan Lu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China
| | - Le Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China
| | - Xinkai Lyu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China
| | - Xin-Yue Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China
| | - Xiao Mi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China
| | - Mei-Geng Hu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China
| | - Chongming Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China.
| | - Xi Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China.
| |
Collapse
|
48
|
Li C, Cui L, Wang X, Yan Z, Wang S, Zheng Y. Using intestinal flora to distinguish non-alcoholic steatohepatitis from non-alcoholic fatty liver. J Int Med Res 2020; 48:300060520978122. [PMID: 33327816 PMCID: PMC7747123 DOI: 10.1177/0300060520978122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Objective To explore specific flora in mouse models of non-alcoholic steatohepatitis
(NASH) to improve NASH diagnostic protocols. Methods Sixty mice were divided into normal diet (ND, 20 mice) and
high-fat/high-sugar diet (HFSD) groups (40 mice). After 8 weeks of feeding,
10 mice in the ND group and 20 mice in the HFSD group were sacrificed to
create the short-term ND and non-alcoholic fatty liver (NAFL) groups,
respectively. After 16 weeks of feeding, the remaining mice were sacrificed
to create the long-term ND and NASH groups, respectively. We then examined
fecal flora, serum biochemical indices, and lipopolysaccharide and tumor
necrosis factor-α levels and analyzed liver tissue. Results The relative abundance of Lactobacillus,
Desulfovibrio, Ruminiclostridium 9,
and Turicibacter differed between NASH and NAFL mice, and
the areas under the receiver operating characteristic curve of the four
genera for diagnosing NASH were 0.705, 0.734, 0.737, and 0.937. The
non-alcoholic fatty liver disease activity score was positively correlated
with the relative abundance of Desulfovibrio (r = 0.353),
Ruminiclostridium 9 (r = 0.431), and
Turicibacter (r = 0.688). Conclusions The relative abundance of Lactobacillus,
Desulfovibrio, Ruminiclostridium, and
Turicibacter may help distinguish NASH from NAFL.
Collapse
Affiliation(s)
- Chao Li
- Medical School of Chinese PLA, Beijing, China.,Department of Gastroenterology, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Lihong Cui
- Medical School of Chinese PLA, Beijing, China.,Department of Gastroenterology, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaohui Wang
- Department of Gastroenterology, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhihui Yan
- Department of Gastroenterology, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shaoxin Wang
- Department of Gastroenterology, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yan Zheng
- Department of Gastroenterology, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
49
|
Jingjing E, Lili M, Zichao C, Rongze M, Qiaoling Z, Ruiyin S, Zongbai H, Junguo W. Effects of buffer salts on the freeze-drying survival rate of Lactobacillus plantarum LIP-1 based on transcriptome and proteome analyses. Food Chem 2020; 326:126849. [DOI: 10.1016/j.foodchem.2020.126849] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/16/2020] [Accepted: 04/17/2020] [Indexed: 10/24/2022]
|
50
|
Gut microbial metabolism of dietary fibre protects against high energy feeding induced ovarian follicular atresia in a pig model. Br J Nutr 2020; 125:38-49. [PMID: 32600501 DOI: 10.1017/s0007114520002378] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To investigate the effects of dietary fibre on follicular atresia in pigs fed a high-fat diet, we fed thirty-two prepubescent gilts a basal diet (CON) or a CON diet supplemented with 300 g/d dietary fibre (fibre), 240 g/d soya oil (SO) or both (fibre + SO). At the 19th day of the 4th oestrus cycle, gilts fed the SO diet showed 112 % more atretic follicles and greater expression of the apoptotic markers, Bax and caspase-3, and these effects were reversed by the fibre diet. The abundance of SCFA-producing microbes was decreased by the SO diet, but this effect was reversed by fibre treatment. Concentrations of serotonin and melatonin in the serum and follicular fluid were increased by the fibre diet. Overall, dietary fibre protected against high fat feeding-induced follicular atresia at least partly via gut microbiota-related serotonin-melatonin synthesis. These results provide insight into preventing negative effects on fertility in humans consuming a high-energy diet.
Collapse
|