1
|
Qi Y, Du S, Li W, Qiu X, Zhou F, Bai L, Zhang B, Mi Z, Qian W, Li L, Zhao X, Li Y. Sanye tablet regulates gut microbiota and bile acid metabolism to attenuate hepatic steatosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119514. [PMID: 39971018 DOI: 10.1016/j.jep.2025.119514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sanye Tablet (SYT), a patent traditional Chinese prescription, is commonly used in treating type 2 diabetes mellitus and hyperlipidemia. Both clinical and animal studies suggest that SYT effectively regulates lipid metabolism. However, its mode of action on hepatic steatosis has yet to be fully elucidated. AIM OF STUDY This study investigates the lipid-regulating effects and underlying mechanism of SYT in high-fat diet (HFD)-induced hepatic steatosis mice. MATERIAL AND METHODS The inhibitory effects of SYT on developing hepatic steatosis were investigated in HFD-fed C57BL/6N mice. Biochemical markers, including total cholesterol (TC) and triglycerides (TG), were measured using specific kits. Hepatic histological alterations were determined by Hematoxylin and Eosin (H&E) and Oil Red O staining. Hepatic, fecal, and systemic bile acids (BAs) profiles were detected by UPLC-MS. mRNA and protein levels of BAs synthesis-related enzymes and critical nodes of farnesoid X receptor (FXR)/fibroblast growth factor 15 (FGF15)/fibroblast growth factor receptor 4 (FGFR4) signaling were detected. Fecal microbial composition was analyzed by 16S rRNA gene sequencing and the antimicrobial activity of SYT was further evaluated in vitro. RESULTS SYT alleviated HFD-induced hepatic steatosis by decreasing TG and TC levels, relieving hepatocyte ballooning, and promoting hepatic BAs synthesis. Moreover, SYT significantly increased the levels of taurine-conjugated BAs in the liver and feces, which in turn inhibited the FXR/FGF15/FGFR4 signaling. Consequently, the hepatic BAs synthesis-related enzyme expression was promoted to reduce lipid accumulation. Notably, SYT remodeled the gut microbiota composition of HFD-fed mice, especially inhibiting the growth of bile salt hydrolase (BSH)-producing bacteria, such as Lactobacillus murinus, Lactobacillus johnsonii, and Enterococcus faecalis. CONCLUSION The findings illustrated that SYT prevented hepatic steatosis by improving hepatic lipid accumulation, which is reflected in modulating the gut-liver axis. SYT corrects BAs profile, restores perturbed FXR/FGF15/FGFR4 signaling and promotes hepatic BAs synthesis, which is associated with modulation on certain BSH-producing bacteria.
Collapse
Affiliation(s)
- Yulin Qi
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Siqi Du
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wenwen Li
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xianzhe Qiu
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Fengjie Zhou
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Liding Bai
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Boli Zhang
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhuoxin Mi
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Weiqiang Qian
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lin Li
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Xin Zhao
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yuhong Li
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| |
Collapse
|
2
|
Li J, Ma Y, Cao Y, Zheng G, Ren Q, Chen C, Zhu Q, Zhou Y, Lu Y, Zhang Y, Deng C, Chen WH, Su J. Integrating microbial GWAS and single-cell transcriptomics reveals associations between host cell populations and the gut microbiome. Nat Microbiol 2025:10.1038/s41564-025-01978-w. [PMID: 40195537 DOI: 10.1038/s41564-025-01978-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025]
Abstract
Microbial genome-wide association studies (GWAS) have uncovered numerous host genetic variants associated with gut microbiota. However, links between host genetics, the gut microbiome and specific cellular contexts remain unclear. Here we use a computational framework, scBPS (single-cell Bacteria Polygenic Score), to integrate existing microbial GWAS and single-cell RNA-sequencing profiles of 24 human organs, including the liver, pancreas, lung and intestine, to identify host tissues and cell types relevant to gut microbes. Analysing 207 microbial taxa and 254 host cell types, scBPS-inferred cellular enrichments confirmed known biology such as dominant communications between gut microbes and the digestive tissue module and liver epithelial cell compartment. scBPS also identified a robust association between Collinsella and the central-veinal hepatocyte subpopulation. We experimentally validated the causal effects of Collinsella on cholesterol metabolism in mice through single-nuclei RNA sequencing on liver tissue to identify relevant cell subpopulations. Mechanistically, oral gavage of Collinsella modulated cholesterol pathway gene expression in central-veinal hepatocytes. We further validated our approach using independent microbial GWAS data, alongside single-cell and bulk transcriptomic analyses, demonstrating its robustness and reproducibility. Together, scBPS enables a systematic mapping of the host-microbe crosstalk by linking cell populations to their interacting gut microbes.
Collapse
Affiliation(s)
- Jingjing Li
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yunlong Ma
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Cao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Gongwei Zheng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qing Ren
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Cheng Chen
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qunyan Zhu
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yijun Zhou
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yu Lu
- The Second School of Clinical Medicine, Institution of Medical Artificial Intelligence, Binzhou Medical University, Yantai, China
| | - Yaru Zhang
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chunyu Deng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wei-Hua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- The Second School of Clinical Medicine, Institution of Medical Artificial Intelligence, Binzhou Medical University, Yantai, China.
- School of Biological Science, Jining Medical University, Rizhao, China.
| | - Jianzhong Su
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
3
|
Onwusereaka CO, Jalaludin J, Oluchi SE, Poh Choo VC. New generation sequencing: molecular approaches for the detection and monitoring of bioaerosols in an indoor environment: a systematic review. REVIEWS ON ENVIRONMENTAL HEALTH 2025; 40:47-62. [PMID: 38214730 DOI: 10.1515/reveh-2023-0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/14/2023] [Indexed: 01/13/2024]
Abstract
INTRODUCTION The exposure of occupants to indoor air pollutants has increased in recent decades. The aim of this review is to discuss an overview of new approaches that are used to study fungal aerosols. Thus, this motivation was to compensate the gaps caused by the use of only traditional approaches in the study of fungal exposure. CONTENT The search involved various databases such as; Science Direct, PubMed, SAGE, Springer Link, EBCOHOST, MEDLINE, CINAHL, Cochrane library, Web of Science and Wiley Online Library. It was limited to full text research articles that reported the use of non-viable method in assessing bioaerosol, written in English Language, full text publications and published from year 2015-2022. SUMMARY AND OUTLOOK A total of 15 articles met the inclusion criteria and was included in this review. The use of next-generation sequencing, which is more commonly referred to as high-throughput sequencing (HTS) or molecular methods in microbial studies is based on the detection of genetic material of organisms present in a given sample. Applying these methods to different environments permitted the identification of the microorganisms present, and a better comprehension of the environmental impacts and ecological roles of microbial communities. Based on the reviewed articles, there is evidence that dust samples harbour a high diversity of human-associated bacteria and fungi. Molecular methods such as next generation sequencing are reliable tools for identifying and tracking the bacterial and fungal diversity in dust samples using 18S metagenomics approach.
Collapse
Affiliation(s)
- Cynthia Oluchi Onwusereaka
- Department of Environmental and Occupational Health, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia Serdang, Selangor, Malaysia
| | - Juliana Jalaludin
- Department of Environmental and Occupational Health, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia Serdang, Selangor, Malaysia
| | - Sampson Emilia Oluchi
- Department of Community Health, Faculty of Medicine and Health Science, Universiti Putra Malaysia Serdang, Selangor, Malaysia
| | | |
Collapse
|
4
|
Ye L, Yao Z, Xuan Q, Liu Q, Bo T. The impact of sleeve gastrectomy on MASH development by regulating the composition of gut microbiota and metabolic homeostasis. Biochem Biophys Res Commun 2025; 752:151466. [PMID: 39938449 DOI: 10.1016/j.bbrc.2025.151466] [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: 11/22/2024] [Revised: 01/21/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
The prevalence of metabolic dysfunction-associated steatohepatitis (MASH) is increasing annually, which is a global public health issue. Although clinical trials are lacking, observational studies indicate that bariatric surgery can alleviate the progression of MASH. Here, we performed sleeve gastrectomy (SG) and Sham surgery on 8-week-old mice, and then fed a AMLN diet for 24 weeks to construct a diet-inducted MASH mice model after 4-week post-surgery recovery. Applying a multi-omics approach combining metagenomics, metabolomics, and transcriptomics, we found that SG prevents the development of hepatic steatosis, inflammation, and fibrosis in MASH mice not only by significantly altering the structure of gut microbiota including s_Akkermansia muciniphila, s_Alistiples dispar, g_Helicobacter and s_uc_Oscillospiraceae, but also by modulating the levels of serum metabolites including l-arginine and taurocholic acid (TCA). These results suggest that SG and the alteration of gut microbiota and its related serum metabolites can be served as the effective therapeutic strategies for MASH.
Collapse
Affiliation(s)
- Lingxi Ye
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhenyu Yao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Qiuhui Xuan
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Qiaoran Liu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250021, China; Key Laboratory of Metabolism and Gastrointestinal Tumor, the First Affiliated Hospital of Shandong First Medical University, China; Key Laboratory of Laparoscopic Technology, the First Affiliated Hospital of Shandong First Medical University, China.
| | - Tao Bo
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| |
Collapse
|
5
|
Schwartz LT, Ladouceur JG, Russell MM, Xie SYL, Bu S, Kerver JM, Comstock SS. The Relationship Between Fiber Intake and Gut Bacterial Diversity and Composition During the Third Trimester of Pregnancy. Nutrients 2025; 17:773. [PMID: 40077643 PMCID: PMC11901921 DOI: 10.3390/nu17050773] [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: 01/28/2025] [Revised: 02/12/2025] [Accepted: 02/15/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES High fiber (34-36 g/day) diets are recommended during pregnancy due to inverse associations with constipation and adverse pregnancy health outcomes, including pre-eclampsia and gestational diabetes. However, the mechanism for this protective effect is poorly defined. Fiber may be protective due to its impact on the composition and function of specific bacteria within the pregnancy gut microbiome. The purpose of this analysis was to investigate whether a sub-sample of cohort study participants in their third trimester met daily dietary fiber and vegetable intake recommendations and, in turn, how this impacted bacterial composition and butyrate-producing genes within the gut microbiome. METHODS Pregnant participants (n = 52) provided stool samples and survey data, which were used to calculate fiber and vegetable intake. Genomic DNA was extracted from the stool samples, followed by PCR to amplify the V4 region of the 16S rRNA gene. Amplicons were sequenced and mapped to the RDP reference. Quantitative real-time PCR was used to measure the abundance of bacterial genes for butyrate production. RESULTS Of the pregnant participants in this sample, 84.7% and 92.3% failed to meet recommendations in the Dietary Guidelines for Americans for dietary fiber and vegetable intake, respectively. All four participants who met the vegetable recommendation were a subset of those who met the fiber recommendation. The participants who met the pregnancy fiber recommendation had gut microbiotas with greater alpha diversity (Shannon and Inverse Simpson) than those who did not. However, there was no association between dietary fiber intake and the abundance of bacterial genes for butyrate production. CONCLUSIONS This research suggests that general fiber intake during pregnancy has a modest association with the gut bacterial community. These preliminary results demonstrate a need to improve fiber intake during pregnancy. Further, studies that measure the relationship between dietary intake of specific types of fiber and associations with specific gut bacterial community members and their functions are needed.
Collapse
Affiliation(s)
- Lindsay T. Schwartz
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA
| | - Jillian G. Ladouceur
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA
| | - Madeleine M. Russell
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA
| | - Shiyi Y. L. Xie
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA
| | - Sihan Bu
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA
| | - Jean M. Kerver
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
| | - Sarah S. Comstock
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
6
|
Yan Y, Zhang K, Li F, Lin L, Chen H, Zhuo LB, Xu J, Jiang Z, Zheng JS, Chen YM. The gut-liver axis links the associations between serum carotenoids and non-alcoholic fatty liver in a 7.8-year prospective study. Hepatobiliary Surg Nutr 2025; 14:16-32. [PMID: 39925899 PMCID: PMC11806141 DOI: 10.21037/hbsn-23-526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/15/2024] [Indexed: 02/11/2025]
Abstract
Background Many studies have shown that carotenoids are beneficial to non-alcoholic fatty liver disease (NAFLD). Therefore, we explored potential biomarkers of gut microbiota and fecal and serum metabolites linking the association between serum carotenoids and NAFLD in adults. Methods This 7.8-year prospective study included 2921 participants with serum carotenoids at baseline and determined NAFLD by ultrasonography (ULS-NAFLD) every 3 years. A total of 828 subjects additionally underwent magnetic resonance imaging to identify NAFLD (MRI-NAFLD). Gut microbiota was analyzed by 16S rRNA sequencing in 1,661 participants, and targeted metabolomics profiling in 893 feces and 896 serum samples was performed by ultrahigh-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) in the middle term. Results A total of 2,522 participants finished follow-up visits. Of these participants, 770, 301, 474, and 977 were categorized into NAFLD-free, improved, new-onset, and persistent NAFLD groups based on their ULS-NAFLD status changes, respectively, and 342/828 were MRI-verified NALFD. Longitudinal analyses showed an inverse association between carotenoids and NALFD risk/presence (P-trend <0.05). Multivariable-adjusted odds ratios (ORs)/hazard ratio (HR) [95% confidence intervals (CIs)] of NAFLD for quartile 4 (vs. quartile 1) of total carotenoids were 0.63 (0.50, 0.80) for incident ULS-NAFLD, 0.20 (0.15, 0.27) for persistent ULS-NAFLD, 1.53 (1.10, 2.12) for improved-NAFLD, and 0.58 (0.39, 0.87) for MRI-NAFLD. The biomarkers in the gut-liver axis significantly associated with both serum carotenoids and NAFLD included sixteen microbial genera mainly in Ruminococcaceae and Veillonellaceae family, nineteen fecal metabolites containing medium-chain fatty acids (MCFAs), bile acids, and carnitines, and sixteen serum metabolites belonging to organic acids and amino acids. The total carotenoids-related scores of significant microbial genera, fecal and serum metabolites mediated the carotenoids-NAFLD association by 8.72%, 12.30%, and 16.83% (all P<0.05) for persistent NAFLD and 9.46%, 8.74%, and 15.7% for incident-NAFLD, respectively. Conclusions Our study reveals a beneficial association of serum carotenoids and incident and persistent NAFLD. The identified gut-liver axis biomarkers provided mechanistic linkage for the epidemiological association.
Collapse
Affiliation(s)
- Yan Yan
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ke Zhang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Fanqin Li
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Lishan Lin
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Hanzu Chen
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Lai-Bao Zhuo
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jinjian Xu
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Zengliang Jiang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Ju-Sheng Zheng
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yu-Ming Chen
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
7
|
Liu Z, You C. The bile acid profile. Clin Chim Acta 2025; 565:120004. [PMID: 39419312 DOI: 10.1016/j.cca.2024.120004] [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: 07/22/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
As a large and structurally diverse family of small molecules, bile acids play a crucial role in regulating lipid, glucose, and energy metabolism. In the human body, bile acids share a similar chemical structure with many isomers, exhibit little difference in polarity, and possess various physiological activities. The types and contents of bile acids present in different diseases vary significantly. Therefore, comprehensive and accurate detection of the content of various types of bile acids in different biological samples can not only provide new insights into the pathogenesis of diseases but also facilitate the exploration of novel strategies for disease diagnosis, treatment, and prognosis. The detection of disease-induced changes in bile acid profiles has emerged as a prominent research focus in recent years. Concurrently, targeted metabolomics methods utilizing high-performance liquid chromatography-mass spectrometry (HPLC-MS) have progressively established themselves as the predominant technology for the separation and detection of bile acids. Bile acid profiles will increasingly play an important role in diagnosis and guidance in the future as the relationship between disease and changes in bile acid profiles becomes clearer. This highlights the growing diagnostic value of bile acid profiles and their potential to guide clinical decision-making. This review aims to explore the significance of bile acid profiles in clinical diagnosis from four perspectives: the synthesis and metabolism of bile acids, techniques for detecting bile acid profiles, changes in bile acid profiles associated with diseases, and the challenges and future prospects of applying bile acid profiles in clinical settings.
Collapse
Affiliation(s)
- Zhenhua Liu
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Chongge You
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
| |
Collapse
|
8
|
Liu Y, Chen Z, Li C, Sun T, Luo X, Jiang B, Liu M, Wang Q, Li T, Cao J, Li Y, Chen Y, Kuai L, Xiao F, Xu H, Cui H. Associations between changes in the gut microbiota and liver cirrhosis: a systematic review and meta-analysis. BMC Gastroenterol 2025; 25:16. [PMID: 39806278 PMCID: PMC11727502 DOI: 10.1186/s12876-025-03589-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
OBJECTIVE Summaries of the relationships between the microbiota and liver cirrhosis and their conclusions are not consistent. This study describes microbial differences in patients with liver cirrhosis by performing a meta-analysis. METHODS We searched PubMed, Embase, Web of Science, and the Cochrane Library and collected related articles published before March 10, 2024. Ratio of autochthonous to non-autochthonous taxa was calculated as the cirrhosis dysbiosis ratio (CDR). Using a random-effects model, the standard mean deviation (SMD) and 95% confidence interval (CI) were calculated. We subsequently performed subgroup, sensitivity, and publication bias analyses. cirrhosis dysbiosis ratio. RESULTS A total of 53 eligible papers including 5076 participants were included. The pooled estimates revealed a moderately significant reduction in gut microbiome richness in patients with liver cirrhosis compared with controls, including the Shannon, Chao1, observed species, ACE, and PD indices, but no significant difference was observed for the Simpson index. Over 80% of the studies reported significant differences in β diversity. Families Enterobacteriaceae and Pasteurellaceae, belonging to the phylum Proteobacteria, along with the family Streptococcaceae and the genera Haemophilus, Streptococcus, and Veillonella, were significantly associated with liver cirrhosis compared to the control group. In contrast, the healthy group exhibited a higher abundance of the class Clostridia, particularly the families Lachnospiraceae and Ruminococcaceae, which are known for their diversity and role as common gut commensals. Furthermore, the class Bacilli, predominantly represented by the genus Streptococcus, was markedly enriched in the cirrhosis group. CONCLUSIONS The microbiota richness of liver cirrhosis patients was lower than that of healthy controls. Alterations in gut microbiota linked to liver cirrhosis were characterized by a decrease in Lachnospiraceae, Ruminococcaceae, and Clostridia and an enrichment of Enterobacteriaceae, Pasteurellaceae, Streptococcaceae, Bacilli, and Streptococcus.
Collapse
Affiliation(s)
- Ye Liu
- Beijing Hospital, Peking University Fifth School of Clinical Medicine, National Center of Gerontology, Beijing, China
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Ziwei Chen
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Chang Li
- Beijing Hospital, Peking University Fifth School of Clinical Medicine, National Center of Gerontology, Beijing, China
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Tianhan Sun
- Department of General Surgery, Department of Hepato-Bilio-Pancreatic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine , Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xuanmei Luo
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Boyue Jiang
- Department of General Surgery, Department of Hepato-Bilio-Pancreatic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine , Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Meilan Liu
- Department of General Surgery, Department of Hepato-Bilio-Pancreatic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine , Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qing Wang
- Department of General Surgery, Department of Hepato-Bilio-Pancreatic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine , Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tong Li
- Department of General Surgery, Department of Hepato-Bilio-Pancreatic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine , Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianfu Cao
- Department of General Surgery, Department of Hepato-Bilio-Pancreatic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine , Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yayu Li
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Chen
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Lu Kuai
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Xiao
- Beijing Hospital, Peking University Fifth School of Clinical Medicine, National Center of Gerontology, Beijing, China.
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China.
- Clinical Biobank, Beijing Hospital, No. 1 Dahua Road, Dong Dan, Beijing, 100730, China.
| | - Hongtao Xu
- Department of Laboratory Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
- Department of Laboratory Medicine, Beijing Hospital, No. 1 Dahua Road, Dong Dan, Beijing, 100730, China.
| | - Hongyuan Cui
- Department of General Surgery, Department of Hepato-Bilio-Pancreatic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine , Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- Department of General Surgery, Beijing Hospital, No. 1 Dahua Road, Dong Dan, Beijing, 100730, China.
| |
Collapse
|
9
|
Monti E, Vianello C, Leoni I, Galvani G, Lippolis A, D’Amico F, Roggiani S, Stefanelli C, Turroni S, Fornari F. Gut Microbiome Modulation in Hepatocellular Carcinoma: Preventive Role in NAFLD/NASH Progression and Potential Applications in Immunotherapy-Based Strategies. Cells 2025; 14:84. [PMID: 39851512 PMCID: PMC11764391 DOI: 10.3390/cells14020084] [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: 12/23/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a heterogeneous tumor associated with several risk factors, with non-alcoholic fatty liver disease (NAFLD) emerging as an important cause of liver tumorigenesis. Due to the obesity epidemics, the occurrence of NAFLD has significantly increased with nearly 30% prevalence worldwide. HCC often arises in the background of chronic liver disease (CLD), such as nonalcoholic steatohepatitis (NASH) and cirrhosis. Gut microbiome (GM) alterations have been linked to NAFLD progression and HCC development, with several investigations reporting a crucial role for the gut-liver axis and microbial metabolites in promoting CLD. Moreover, the GM affects liver homeostasis, energy status, and the immune microenvironment, influencing the response to immunotherapy with interesting therapeutic implications. In this review, we summarize the main changes in the GM and derived metabolites (e.g., short-chain fatty acids and bile acids) occurring in HCC patients and influencing NAFLD progression, emphasizing their potential as early diagnostic biomarkers and prognostic tools. We discuss the weight loss effects of diet-based interventions and healthy lifestyles for the treatment of NAFLD patients, highlighting their impact on the restoration of the intestinal barrier and GM structure. We also describe encouraging preclinical findings on the modulation of GM to improve liver functions in CLD, boost the antitumor immune response (e.g., probiotic supplementations or anti-hypercholesterolemic drug treatment), and ultimately delay NAFLD progression to HCC. The development of safe and effective strategies that target the gut-liver axis holds promise for liver cancer prevention and treatment, especially if personalized options will be considered.
Collapse
Affiliation(s)
- Elisa Monti
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Clara Vianello
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Ilaria Leoni
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Giuseppe Galvani
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Annalisa Lippolis
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
| | - Federica D’Amico
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.D.); (S.R.); (S.T.)
| | - Sara Roggiani
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.D.); (S.R.); (S.T.)
- Human Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Claudio Stefanelli
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.D.); (S.R.); (S.T.)
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesca Fornari
- Department for Life Quality Studies, University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (E.M.); (C.V.); (I.L.); (G.G.); (A.L.); (C.S.)
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
10
|
Xirouchakis E, Pelekanos A, Xirouchakis S, Kranidioti H, Manolakopoulos S. A Systematic Review of Microbiota in Cirrhosis: A Change Towards a More Pathogenic Predisposition. Int J Mol Sci 2025; 26:527. [PMID: 39859243 PMCID: PMC11765289 DOI: 10.3390/ijms26020527] [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: 11/25/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
The microbiome of the human intestine is a regulator of health that modulates immune response and plays an important role in metabolism. The diversity, and abundance of microbiota communities in the gut have been shown to change in cirrhosis and its complications. We aimed to review the current knowledge regarding microbiota alterations in cirrhosis, its potential differences according to etiology, and its role in the development of cirrhosis complications. A systematic search of the online bibliographic database up to July 2024 was performed. Randomized controlled trials and observational and cohort studies that included a total or at least a cohort of cirrhotic adult patients were enlisted for data extraction and analysis. A total of 73 publications were included for data extraction. Alpha diversity was found to decrease in cirrhotic patients in 30/38 (78%) of the studies, while beta diversity in 20/22 (90%) presented significant differences between healthy and cirrhotic groups. Proteobacteria significantly increased in 20/27 (74%) studies, followed by Actinobacteria and Fusobacteria, while 22/25 (88%) studies found either a reduction in cirrhotic patients or increased abundance in healthy controls for Firmicutes and Bacteroidetes. The most abundant genera in hepatic encephalopathy groups were pathobionts such as Enterococcus and Streptococcus, followed by Vellionella and Escherichia. Heterogeneity was found among studies regarding Alpha diversity in hepatocellular carcinoma (HCC) as it was decreased in three studies, indifferent in five, and increased in three studies in comparison to cirrhotic non-HCC patients. The dysbiosis of the gut microbiota is associated with cirrhosis and the development of complications such as hepatic encephalopathy and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Elias Xirouchakis
- Gastroenterology-Liver-Endoscopy Unit, 2nd Department of Internal Medicine, General Hospital of Athens “Hippocration”, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (A.P.); (H.K.); (S.M.)
- Department of Gastroenterology and Hepatology, Athens Medical, P. Faliron Hospital, 175 62 Athens, Greece;
| | - Alexandros Pelekanos
- Gastroenterology-Liver-Endoscopy Unit, 2nd Department of Internal Medicine, General Hospital of Athens “Hippocration”, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (A.P.); (H.K.); (S.M.)
| | - Spyridon Xirouchakis
- Department of Gastroenterology and Hepatology, Athens Medical, P. Faliron Hospital, 175 62 Athens, Greece;
- Medical School, European University of Cyprus, 2404 Nicosia, Cyprus
| | - Hariklia Kranidioti
- Gastroenterology-Liver-Endoscopy Unit, 2nd Department of Internal Medicine, General Hospital of Athens “Hippocration”, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (A.P.); (H.K.); (S.M.)
| | - Spilios Manolakopoulos
- Gastroenterology-Liver-Endoscopy Unit, 2nd Department of Internal Medicine, General Hospital of Athens “Hippocration”, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (A.P.); (H.K.); (S.M.)
| |
Collapse
|
11
|
Xu Z, Wang S, Liu C, Kang J, Pan Y, Zhang Z, Zhou H, Xu M, Li X, Wang H, Niu S, Liu L, Sun D, Liu X. The Role of Gut Microbiota in Male Erectile Dysfunction of Rats. World J Mens Health 2025; 43:213-227. [PMID: 38772541 PMCID: PMC11704160 DOI: 10.5534/wjmh.230337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/15/2024] [Accepted: 02/08/2024] [Indexed: 05/23/2024] Open
Abstract
PURPOSE Erectile dysfunction (ED) is a common male sexual dysfunction. Gut microbiota plays an important role in various diseases. To investigate the effects and mechanisms of intestinal flora dysregulation induced by high-fat diet (HFD) on erectile function. MATERIALS AND METHODS Male Sprague-Dawley rats aged 8 weeks were randomly divided into the normal diet (ND) and HFD groups. After 24 weeks, a measurement of erectile function was performed. We performed 16S rRNA sequencing of stool samples. Then, we established fecal microbiota transplantation (FMT) rat models by transplanting fecal microbiota from rats of ND group and HFD group to two new groups of rats respectively. After 24 weeks, erectile function of the rats was evaluated and 16S rRNA sequencing was performed, and serum samples were collected for the untargeted metabolomics detection. RESULTS The erectile function of rats and the species diversity of intestinal microbiota in the HFD group was significantly lower, and the characteristics of the intestinal microbiota community structure were also significantly different between the two groups. The erectile function of rats in the HFD-FMT group was significantly lower than that of rats in the ND-FMT group. The characteristics of the intestinal microbiota community structure were significantly different. In the HFD-FMT group, 27 metabolites were significantly different and they were mainly involved in the several inflammation-related pathways. CONCLUSIONS Intestinal microbiota disorders induced by HFD can damage the intestinal barrier of rats, change the serum metabolic profile, induce low-grade inflammation and apoptosis in the corpus cavernosum of the penis, and lead to ED.
Collapse
Affiliation(s)
- Zhunan Xu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shangren Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunxiang Liu
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiaqi Kang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yang Pan
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhexin Zhang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hang Zhou
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mingming Xu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xia Li
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Haoyu Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuai Niu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Daqing Sun
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoqiang Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
12
|
Chen Y, Liu Y, Pu J, Gui S, Wang D, Zhong X, Tao W, Chen X, Chen W, Chen X, Qiao R, Li Z, Tao X, Xie P. Treatment response of venlafaxine induced alterations of gut microbiota and metabolites in a mouse model of depression. Metab Brain Dis 2024; 39:1505-1521. [PMID: 39150654 DOI: 10.1007/s11011-024-01403-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
Antidepressants remain the first-line treatment for depression. However, the factors influencing medication response are still unclear. Accumulating evidence implicates an association between alterations in gut microbiota and antidepressant response. Therefore, the aim of this study is to investigate the role of the gut microbiota-brain axis in the treatment response of venlafaxine. After chronic social defeat stress and venlafaxine treatment, mice were divided into responders and non-responders groups. We compared the composition of gut microbiota using 16 S ribosomal RNA sequencing. Meanwhile, we quantified metabolomic alterations in serum and hippocampus, as well as hippocampal neurotransmitter levels using liquid chromatography-mass spectrometry. We found that the abundances of 29 amplicon sequence variants (ASVs) were significantly altered between the responders and non-responders groups. These ASVs belonged to 8 different families, particularly Muribaculaceae. Additionally, we identified 38 and 39 differential metabolites in serum and hippocampus between the responders and non-responders groups, respectively. Lipid, amino acid, and purine metabolisms were enriched in both serum and hippocampus. In hippocampus, the concentrations of tryptophan, phenylalanine, gamma-aminobutyric acid, glutamic acid, and glutamine were increased, while the level of succinic acid was decreased in the responders group, compared with the non-responders group. Our findings suggest that the gut microbiota may play a role in the antidepressant effect of venlafaxine by modulating metabolic processes in the central and peripheral tissues. This provides a novel microbial and metabolic framework for understanding the impact of the gut microbiota-brain axis on antidepressant response.
Collapse
Affiliation(s)
- Yue Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Yiyun Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Juncai Pu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Siwen Gui
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dongfang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaogang Zhong
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Tao
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Xiaopeng Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Weiyi Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiang Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Renjie Qiao
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Zhuocan Li
- Psychologic Medicine Science, Chongqing Medical University, Chongqing, China
| | - Xiangkun Tao
- Psychologic Medicine Science, Chongqing Medical University, Chongqing, China
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi road, Yuzhong District, Chongqing, 400016, China.
- Chongqing Institute for Brain and Intelligence, Chongqing, China.
| |
Collapse
|
13
|
Comerlato CB, Zhang X, Walker K, Mayne J, Figeys D, Brandelli A. The Influence of Protein Secretomes of Enterococcus durans on ex vivo Human Gut Microbiome. Probiotics Antimicrob Proteins 2024; 16:1954-1965. [PMID: 37589783 DOI: 10.1007/s12602-023-10136-9] [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] [Accepted: 08/07/2023] [Indexed: 08/18/2023]
Abstract
The gut microbiome plays a critical role to all animals and humans health. Methods based on ex vivo cultures are time and cost-effective solutions for rapid evaluation of probiotic effects on microbiomes. In this study, we assessed whether the protein secretome from the potential probiotic Enterococcus durans LAB18S grown on fructoligosaccharides (FOS) and galactoligosaccharides (GOS) had specific effects on ex vivo cultured intestinal microbiome obtained from a healthy individual. Metaproteomics was used to evaluate changes in microbial communities of the human intestinal microbiome. Hierarchical clustering analysis revealed 654 differentially abundant proteins from the metaproteome samples, showing that gut microbial protein expression varied on the presence of different E. durans secretomes. Increased amount of Bacteroidetes phylum was observed in treatments with secretomes from E. durans cultures on FOS, GOS and albumin, resulting in a decrease of the Firmicutes to Bacteroidetes (F/B) ratio. The most functionally abundant bacterial taxa were Roseburia, Bacteroides, Alistipes and Faecalibacterium. The results suggest that the secretome of E. durans may have favorable effects on the intestinal microbial composition, stimulating growth and different protein expression of beneficial bacteria. These findings suggest that proteins secreted by E. durans growing on FOS and GOS have different effects on the modulation of gut microbiota functional activities during cultivation.
Collapse
Affiliation(s)
- Carolina Baldisserotto Comerlato
- Laboratório de Bioquímica e Microbiologia Aplicada, Instituto de Ciência e Tecnologia de Alimentos, Universidade Federal do Rio Grande do Sul, 91510-970, Porto Alegre, Brazil
| | - Xu Zhang
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Krystal Walker
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Janice Mayne
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Daniel Figeys
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| | - Adriano Brandelli
- Laboratório de Bioquímica e Microbiologia Aplicada, Instituto de Ciência e Tecnologia de Alimentos, Universidade Federal do Rio Grande do Sul, 91510-970, Porto Alegre, Brazil.
| |
Collapse
|
14
|
Scarpellini E, Scarcella M, Tack JF, Scarlata GGM, Zanetti M, Abenavoli L. Gut Microbiota and Metabolic Dysfunction-Associated Steatotic Liver Disease. Antioxidants (Basel) 2024; 13:1386. [PMID: 39594528 PMCID: PMC11591341 DOI: 10.3390/antiox13111386] [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: 09/02/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Background: The gut microbiota constitutes a complex microorganism community that harbors bacteria, viruses, fungi, protozoa, and archaea. The human gut bacterial microbiota has been extensively proven to participate in human metabolism, immunity, and nutrient absorption. Its imbalance, namely "dysbiosis", has been linked to disordered metabolism. Metabolic dysfunction-associated steatotic liver disease (MASLD) is one of the features of deranged human metabolism and is the leading cause of liver cirrhosis and hepatocellular carcinoma. Thus, there is a pathophysiological link between gut dysbiosis and MASLD. Aims and Methods: We aimed to review the literature data on the composition of the human bacterial gut microbiota and its dysbiosis in MASLD and describe the concept of the "gut-liver axis". Moreover, we reviewed the approaches for gut microbiota modulation in MASLD treatment. Results: There is consolidated evidence of particular gut dysbiosis associated with MASLD and its stages. The model explaining the relationship between gut microbiota and the liver has a bidirectional organization, explaining the physiopathology of MASLD. Oxidative stress is one of the keystones in the pathophysiology of MASLD and fibrosis generation. There is promising and consolidated evidence for the efficacy of pre- and probiotics in reversing gut dysbiosis in MASLD patients, with therapeutic effects. Few yet encouraging data on fecal microbiota transplantation (FMT) in MASLD are available in the literature. Conclusions: The gut dysbiosis characteristic of MASLD is a key target in its reversal and treatment via diet, pre/probiotics, and FMT treatment. Oxidative stress modulation remains a promising target for MASLD treatment, prevention, and reversal.
Collapse
Affiliation(s)
- Emidio Scarpellini
- Translational Research in Gastroeintestinal Disorders, Gasthuisberg University Hospital, KULeuven, Herestraat 49, 3000 Lueven, Belgium;
| | - Marialaura Scarcella
- Anesthesia, Intensive Care and Nutritional Science-Azienda Ospedaliera “Santa Maria”, Via Tristano di Joannuccio, 05100 Terni, Italy;
| | - Jan F. Tack
- Translational Research in Gastroeintestinal Disorders, Gasthuisberg University Hospital, KULeuven, Herestraat 49, 3000 Lueven, Belgium;
| | | | - Michela Zanetti
- Geriatrics Department, Nutrition and Malnutrition Unit, Azienda Sanitario-Universitaria Giuliano Isontina, Ospedale Maggiore, piazza dell’Ospitale 1, 34100 Triste, Italy;
| | - Ludovico Abenavoli
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (G.G.M.S.); (L.A.)
| |
Collapse
|
15
|
Lee YP, Chiu CC, Chang YC, Chen YH, Wu WK, Wu MS, Chuang HL. Co-exposure to different bacterial species' lipopolysaccharides with the NASH diet exacerbates NASH and liver fibrosis progression in mice. Clin Res Hepatol Gastroenterol 2024; 48:102470. [PMID: 39317267 DOI: 10.1016/j.clinre.2024.102470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 09/16/2024] [Accepted: 09/22/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND AND AIM With the obesity epidemic, nonalcoholic fatty liver disease (NAFLD) has become a public health concern, but its progression mechanism remains unclear. Experimental models mimicking human NAFLD/steatohepatitis (NASH) are crucial. This study simulates gut microbiota imbalance effects on NASH and liver fibrosis. METHODS We used different bacterial sources of lipopolysaccharide (LPS), including Escherichia coli (GEC) and Salmonella abortus equi (GSE), combined with a Gubra Amylin NASH (GAN) diet to induce NASH and liver fibrosis. RESULTS The GSE group showed significantly higher serum alanine aminotransferase, hydroxyproline, CD68-positive cells, α-smooth muscle actin, glial fibrillary acidic protein, and TNF-α, COL1A1, TGF-β, and NLRP3 expressions compared to the the GAN group. The GSE group also had higher Erysipelotrichaceae, Akkermansiaceae, and Bacteroidaceae family numbers. CONCLUSIONS The GAN diet with LPS treatment successfully induced NASH and fibrosis making this model useful for preclinical NASH drug testing.
Collapse
Affiliation(s)
- Yen-Peng Lee
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Chien-Chao Chiu
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Yung-Chi Chang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Yi-Hsun Chen
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Wei-Kai Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan.
| |
Collapse
|
16
|
Krizanac M, Štancl P, Mass-Sanchez PB, Karlić R, Moeckel D, Lammers T, Asimakopoulos A, Weiskirchen R. The influence of perilipin 5 deficiency on gut microbiome profiles in murine metabolic dysfunction-associated fatty liver disease (MAFLD) and MAFLD-hepatocellular carcinoma. Front Cell Infect Microbiol 2024; 14:1443654. [PMID: 39469452 PMCID: PMC11513398 DOI: 10.3389/fcimb.2024.1443654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/20/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction Metabolic dysfunction-associated fatty liver disease (MAFLD) has emerged as the leading cause of hepatocellular carcinoma (HCC) worldwide. Over the years, Perilipin 5 (PLIN5) has been recognized as a key regulator of both MAFLD and HCC development. In our previous studies we demonstrated that deficiency in Plin5 reduces the severity of MAFLD and HCC in mice. Interestingly, it has been established that patients with MAFLD and HCC exhibit various changes in their gut microbiome profiles. The gut microbiome itself has been shown to play a role in modulating carcinogenesis and the immune response against cancer. Methods Therefore, we conducted a study to investigate the alterations in fecal microbiome composition in wild type (WT) and Plin5-deficient (Plin5 -/-) mice models of MAFLD and MAFLD-induced HCC (MAFLD-HCC). We utilized 16S rRNA gene sequencing analysis to profile the composition of gut bacteria in fecal samples. Results Notably, we discovered that the absence of Plin5 alone is already associated with changes in gut microbiota composition. Moreover, feeding the mice a Western diet (WD) resulted in additional microbial alterations. Interestingly, Plin5 -/- animals exhibited an enrichment of the beneficial taxa Lactobacillus in both animal models. Discussion Our findings identify Plin5 as a major regulator of gut microbiota during the development of MAFLD and MAFLD-HCC.
Collapse
Affiliation(s)
- Marinela Krizanac
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Paula Štancl
- Bioinformatics Group, Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Paola Berenice Mass-Sanchez
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Rosa Karlić
- Bioinformatics Group, Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Diana Moeckel
- Institute for Experimental Molecular Imaging, RWTH Aachen, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen, Aachen, Germany
| | - Anastasia Asimakopoulos
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
17
|
Bibi S, Kerbiriou C, Uzma, Mckirdy S, Kostrytsia A, Rasheed H, Eqani SAMAS, Gerasimidis K, Nurulain SM, Ijaz UZ. Gut microbiome and function are altered for individuals living in high fluoride concentration areas in Pakistan. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116959. [PMID: 39232295 DOI: 10.1016/j.ecoenv.2024.116959] [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: 03/09/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Endemic fluorosis refers to the condition when individuals are exposed to excessive amounts of fluoride ion due to living in a region characterized by elevated levels of fluorine in the drinking water, food, and/or air. In Pakistan, a substantial proportion of the population is thereby affected, posing a public health concern. OBJECTIVES Assessing how the gut microbiota and its metabolic profiles are impacted by chronic exposure to fluoride in drinking water (that caused Dental Fluorosis) as well as to perceive how this microbiota is connected to adverse health outcomes prevailing with fluoride exposure. METHODS Drinking water (n=27) and biological samples (n=100) of blood, urine and feces were collected from 70 high fluoride exposed (with Dental Fluorosis) and 30 healthy control (without Dental Fluorosis) subjects. Water and urinary fluoride concentrations were determined. Serum/plasma biochemical testing was performed. Fecal DNA extraction, 16S rRNA analysis of microbial taxa, their predicted metabolic function and fecal short chain fatty acids (SCFAs) quantification were carried out. RESULTS The study revealed that microbiota taxonomic shifts and their metabolic characterization had been linked to certain host clinical parameters under the chronic fluoride exposure. Some sets of genera showed strong specificity to water and urine fluoride concentrations, Relative Fat Mass index and SCFAs. The SCFAs response in fluoride-exposed samples was observed to be correlated with bacterial taxa that could contribute to adverse health effects. CONCLUSIONS Microbial dysbiosis as a result of endemic fluorosis exhibits a structure that is associated with risk of metabolic deregulation and is implicated in various diseases. Our results may form the development of novel interventions and may have utility in diagnosis and monitoring.
Collapse
Affiliation(s)
- Sara Bibi
- Department of Biosciences, COMSATS University Islamabad, 45550, Pakistan; Water & Environment Research Group, University of Glasgow, Mazumdar-Shaw Advanced Research Centre, Glasgow G11 6EW, UK
| | - Caroline Kerbiriou
- School of Medicine, Dentistry & Nursing, Glasgow Royal Infirmary, Glasgow G31 2ER, UK
| | - Uzma
- Water & Environment Research Group, University of Glasgow, Mazumdar-Shaw Advanced Research Centre, Glasgow G11 6EW, UK
| | - Shona Mckirdy
- School of Medicine, Dentistry & Nursing, Glasgow Royal Infirmary, Glasgow G31 2ER, UK
| | - Anastasiia Kostrytsia
- Water & Environment Research Group, University of Glasgow, Mazumdar-Shaw Advanced Research Centre, Glasgow G11 6EW, UK
| | - Hifza Rasheed
- National Water Quality Laboratory, Pakistan Council of Research in Water Resources (PCRWR), Islamabad, Pakistan
| | | | | | | | - Umer Zeeshan Ijaz
- Water & Environment Research Group, University of Glasgow, Mazumdar-Shaw Advanced Research Centre, Glasgow G11 6EW, UK; Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 7BE, UK; National University of Ireland, University Road, Galway H91 TK33, Ireland.
| |
Collapse
|
18
|
Du Y, Wang S, Zhou T, Zhao Z. Causal Effects of Gut Microbiota and Metabolites on Chronic Obstructive Pulmonary Disease: A Bidirectional Two Sample Mendelian Randomization Study. Int J Chron Obstruct Pulmon Dis 2024; 19:2153-2167. [PMID: 39360021 PMCID: PMC11446199 DOI: 10.2147/copd.s472218] [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: 04/04/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
Background Recent evidence suggests that the gut microbiome and metabolites are intricately involved in Chronic Obstructive Pulmonary Disease (COPD) pathogenesis, yet the precise causal relationships remain unclear due to confounding factors and reverse causation. This study employs bidirectional two-sample Mendelian Randomization (MR) to clarify these connections. Methods Summary data from publicly available Genome-Wide Association Studies (GWAS) concerning the gut microbiome, metabolites, and COPD were compiled. The selection of genetic instrumental variables (Single Nucleotide Polymorphisms, or SNPs) for MR analysis was conducted meticulously, primarily utilizing the Inverse Variance Weighting (IVW) method, supplemented by MR-Egger regression and the Weighted Median (WM) approach. The evaluation of heterogeneity and horizontal pleiotropy was performed using Cochran's Q test, the MR-Egger intercept test, and the MR-PRESSO global test. Sensitivity analyses, including leave-one-out tests, were conducted to verify the robustness of our results. And the mediation effect of gut microbiota-mediated changes in metabolites on the causal relationship with COPD was analyzed. Results Our study identified nine significant gut microbiota taxa and thirteen known metabolites implicated in COPD pathogenesis. Moreover, associations between the onset of COPD and the abundance of five bacterial taxa, as well as the concentration of three known metabolites, were established. These findings consistently withstood sensitivity analyses, reinforcing their credibility. Additionally, our results revealed that gut microbiota contribute to the development of COPD by mediating changes in metabolites. Conclusion Our bidirectional Two-Sample Mendelian Randomization analysis has revealed reciprocal causal relationships between the abundance of gut microbiota and metabolite concentrations in the context of COPD. This research holds promise for identifying biomarkers for early COPD diagnosis and monitoring disease progression, thereby opening new pathways for prevention and treatment. Further investigation into the underlying mechanisms is essential to improve our understanding of COPD onset.
Collapse
Affiliation(s)
- Yongkun Du
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People's Republic of China
| | - Shuai Wang
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People's Republic of China
| | - Ting Zhou
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People's Republic of China
| | - Zhongyan Zhao
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People's Republic of China
| |
Collapse
|
19
|
Clavell-Sansalvador A, Río-López R, González-Rodríguez O, García-Gil LJ, Xifró X, Zigovski G, Ochoteco-Asensio J, Ballester M, Dalmau A, Ramayo-Caldas Y. Effect of Group Mixing and Available Space on Performance, Feeding Behavior, and Fecal Microbiota Composition during the Growth Period of Pigs. Animals (Basel) 2024; 14:2704. [PMID: 39335293 PMCID: PMC11428945 DOI: 10.3390/ani14182704] [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: 08/22/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Stress significantly affects the health, welfare, and productivity of farm animals. We performed a longitudinal study to evaluate stress's effects on pig performance, feeding behavior, and fecal microbiota composition. This study involved 64 Duroc pigs during the fattening period, divided into two experimental groups: a stress group (n = 32) and a control group (n = 32). Stressed groups had less space and were mixed twice during the experiment. We monitored body weight, feed efficiency, feeding behavior, and fecal microbiota composition. Compared to the control group, the stressed pigs exhibited reduced body weight, feed efficiency, fewer feeder visits, and longer meal durations. In the fecal microbiota, resilience was observed, with greater differences between groups when sampling was closer to the stressful stimulus. Stressed pigs showed an increase in opportunistic bacteria, such as Streptococcus, Treponema and members of the Erysipelotrichaceae family, while control pigs had more butyrate- and propionate-producing genera like Anaerobutyricum, Coprococcus and HUN007. Our findings confirm that prolonged stress negatively impacts porcine welfare, behavior, and performance, and alters their gut microbiota. Specific microorganisms identified could serve as non-invasive biomarkers for stress, potentially informing both animal welfare and similar gut-brain axis mechanisms relevant to human research.
Collapse
Affiliation(s)
- Adrià Clavell-Sansalvador
- Animal Breeding and Genetics Program, Institute of Agrifood Research and Technology (IRTA), Torre Marimon, 08140 Caldes de Montbui, Barcelona, Spain; (O.G.-R.); (M.B.)
- Animal Welfare Subprogram, Institute of Agrifood Research and Technology (IRTA), 17121 Monells, Girona, Spain; (R.R.-L.); (J.O.-A.); (A.D.)
| | - Raquel Río-López
- Animal Welfare Subprogram, Institute of Agrifood Research and Technology (IRTA), 17121 Monells, Girona, Spain; (R.R.-L.); (J.O.-A.); (A.D.)
| | - Olga González-Rodríguez
- Animal Breeding and Genetics Program, Institute of Agrifood Research and Technology (IRTA), Torre Marimon, 08140 Caldes de Montbui, Barcelona, Spain; (O.G.-R.); (M.B.)
| | - L. Jesús García-Gil
- Digestive Diseases and Microbiota Group, Biomedical Research Institute of Girona (IDIBGI), 17190 Girona, Girona, Spain;
- New Therapeutic Targets Lab Research Group, Medical Sciences Departament, Faculty of Medicine, Universitat de Girona, 17071 Girona, Girona, Spain;
| | - Xavier Xifró
- New Therapeutic Targets Lab Research Group, Medical Sciences Departament, Faculty of Medicine, Universitat de Girona, 17071 Girona, Girona, Spain;
| | - Gustavo Zigovski
- School of Medicine and Life Sciences, Graduate Program in Animal Science, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Paraná, Brazil;
| | - Juan Ochoteco-Asensio
- Animal Welfare Subprogram, Institute of Agrifood Research and Technology (IRTA), 17121 Monells, Girona, Spain; (R.R.-L.); (J.O.-A.); (A.D.)
| | - Maria Ballester
- Animal Breeding and Genetics Program, Institute of Agrifood Research and Technology (IRTA), Torre Marimon, 08140 Caldes de Montbui, Barcelona, Spain; (O.G.-R.); (M.B.)
| | - Antoni Dalmau
- Animal Welfare Subprogram, Institute of Agrifood Research and Technology (IRTA), 17121 Monells, Girona, Spain; (R.R.-L.); (J.O.-A.); (A.D.)
| | - Yuliaxis Ramayo-Caldas
- Animal Breeding and Genetics Program, Institute of Agrifood Research and Technology (IRTA), Torre Marimon, 08140 Caldes de Montbui, Barcelona, Spain; (O.G.-R.); (M.B.)
| |
Collapse
|
20
|
Panyod S, Wu WK, Hsieh YC, Tseng YJ, Peng SY, Chen RA, Huang HS, Chen YH, Shen TCD, Ho CT, Liu CJ, Chuang HL, Huang CC, Wu MS, Sheen LY. Ginger essential oil prevents NASH progression by blocking the NLRP3 inflammasome and remodeling the gut microbiota-LPS-TLR4 pathway in mice. Nutr Diabetes 2024; 14:65. [PMID: 39152116 PMCID: PMC11329514 DOI: 10.1038/s41387-024-00306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Diet and gut microbiota contribute to non-alcoholic steatohepatitis (NASH) progression. High-fat diets (HFDs) change gut microbiota compositions, induce gut dysbiosis, and intestinal barrier leakage, which facilitates portal influx of pathogen-associated molecular patterns including lipopolysaccharides (LPS) to the liver and triggers inflammation in NASH. Current therapeutic drugs for NASH have adverse side effects; however, several foods and herbs that exhibit hepatoprotection could be an alternative method to prevent NASH. METHODS We investigated ginger essential oil (GEO) against palm oil-containing HFDs in LPS-injected murine NASH model. RESULTS GEO reduced plasma alanine aminotransferase levels and hepatic pro-inflammatory cytokine levels; and increased antioxidant catalase, glutathione reductase, and glutathione levels to prevent NASH. GEO alleviated hepatic inflammation through mediated NLR family pyrin domain-containing 3 (NLRP3) inflammasome and LPS/Toll-like receptor four (TLR4) signaling pathways. GEO further increased beneficial bacterial abundance and reduced NASH-associated bacterial abundance. CONCLUSION This study demonstrated that GEO prevents NASH progression which is probably associated with the alterations of gut microbiota and inhibition of the LPS/TLR4/NF-κB pathway. Hence, GEO may offer a promising application as a dietary supplement for the prevention of NASH.
Collapse
Affiliation(s)
- Suraphan Panyod
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan, ROC
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Wei-Kai Wu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan, ROC
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC
- Bachelor Program of Biotechnology and Food Nutrition, National Taiwan University, Taipei, Taiwan, ROC
| | - Ya-Chi Hsieh
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Yea-Jing Tseng
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Sin-Yi Peng
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Rou-An Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Huai-Syuan Huang
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Yi-Hsun Chen
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Ting-Chin David Shen
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Chun-Jen Liu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC
| | - Chi-Chang Huang
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan City, Taiwan, ROC
| | - Ming-Shiang Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC.
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC.
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC.
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan, ROC.
- National Center for Food Safety Education and Research, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
21
|
Akhlaghi E, Salari E, Mansouri M, Shafiei M, Kalantar-Neyestanaki D, Aghassi H, Fasihi Harandi M. Identification and comparison of intestinal microbial diversity in patients at different stages of hepatic cystic echinococcosis. Sci Rep 2024; 14:18912. [PMID: 39143364 PMCID: PMC11324937 DOI: 10.1038/s41598-024-70005-5] [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: 04/22/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024] Open
Abstract
There is a significant focus on the role of the host microbiome in different outcomes of human parasitic diseases, including cystic echinococcosis (CE). This study was conducted to identify the intestinal microbiome of patients with CE at different stages of hydatid cyst compared to healthy individuals. Stool samples from CE patients as well as healthy individuals were collected. The samples were divided into three groups representing various stages of hepatic hydatid cyst: active (CE1 and CE2), transitional (CE3), and inactive (CE4 and CE5). One family member from each group was selected to serve as a control. The gut microbiome of patients with different stages of hydatid cysts was investigated using metagenomic next-generation amplicon sequencing of the V3-V4 region of the 16S rRNA gene. In this study, we identified 4862 Operational Taxonomic Units from three stages of hydatid cysts in CE patients and healthy individuals with a combined frequency of 2,955,291. The most abundant genera observed in all the subjects were Blautia, Agathobacter, Faecalibacterium, Bacteroides, Bifidobacterium, and Prevotella. The highest microbial frequency was related to inactive forms of CE, and the lowest frequency was observed in the group with active forms. However, the lowest OTU diversity was found in patients with inactive cysts compared with those with active and transitional cyst stages. The genus Agatobacter had the highest OTU frequency. Pseudomonas, Gemella, and Ligilactobacillus showed significant differences among the patients with different stages of hydatid cysts. Additionally, Anaerostipes and Candidatus showed significantly different reads in CE patients compared to healthy individuals. Our findings indicate that several bacterial genera can play a role in the fate of hydatid cysts in patients at different stages of the disease.
Collapse
Affiliation(s)
- Elham Akhlaghi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Salari
- Department of Plant Protection, Faculty of Plant Production, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Mehdi Mansouri
- Department of Agricultural Biotechnology, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mohammad Shafiei
- Research Center for Hydatid Disease in Iran, Department of Surgery, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Davood Kalantar-Neyestanaki
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Medical Microbiology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Aghassi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Fasihi Harandi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
22
|
Guo L, Zhou J, Xie F, Lang Q, Xu Y, Chen L, Xue Z, Mao Y, Wang R. The profile of oral microbiome in Chinese elderly population associated with aging and systemic health status. BMC Oral Health 2024; 24:895. [PMID: 39103866 PMCID: PMC11299356 DOI: 10.1186/s12903-024-04676-x] [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: 03/04/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024] Open
Abstract
OBJECTIVE The health of oral cavity is considered as an important indicator of aging. Oral microbiota is highly associated with the oral health, while the variation of oral microbiome in elderly population and characteristic microbes associated with aging remain unclear. SUBJECTS AND METHODS In this study, 130 elderly subjects were recruited and divided into 3 groups according to their age: Stage I group (65 ≤ years < 70), Stage II group (70 ≤ years < 75), and Stage III group (75 ≤ years < 80). Their physiological indices were analyzed with using Illumina MiSeq platform and the oral microbiome was determined by high-throughput sequencing. RESULTS Along with aging, the level of fasting blood glucose, systolic pressure and monocytes are significantly increased. No significant difference was detected on the whole structure of the oral microbiome among groups. While using Metastats and Spearman's correlation analysis, specific bacteria were identified as potential age- or health index-related bacterial genera including Fusobacterium, Parvimonas, Porphyromonas, Aminobacter, Collinsella, Clostridium and Acinetobacter. CONCLUSION Our study revealed that the composition structure of salivary microbiota in elderly population was relatively stable while specific bacteria were correlated with age and health status, which is promising to be served as health indicators of the elderly after further exploration.
Collapse
Affiliation(s)
- Liqiang Guo
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Zhou
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feng Xie
- Beicai Community Health Service Center of Pudong New District, No. 271 Lianyuan Road, Shanghai, China
| | - Qing Lang
- Beicai Community Health Service Center of Pudong New District, No. 271 Lianyuan Road, Shanghai, China
| | - Yuesong Xu
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Luping Chen
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhengsheng Xue
- China Mengniu Dairy Company LimitedGlobal R&D Innovation Center, Inner Mongolia Mengniu Dairy (Group) Co. Ltd., Hohhot, 011500, China
| | - Yuejian Mao
- China Mengniu Dairy Company LimitedGlobal R&D Innovation Center, Inner Mongolia Mengniu Dairy (Group) Co. Ltd., Hohhot, 011500, China.
| | - Ruirui Wang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
23
|
Belobrajdic DP, Osborne S, Conlon M, Brook H, Addepalli R, Muhlhausler BS. Assessing the Protein Quality, In Vitro Intestinal Iron Absorption and Human Faecal Microbiota Impacts of Plant-Based Mince. Nutrients 2024; 16:2339. [PMID: 39064781 PMCID: PMC11279466 DOI: 10.3390/nu16142339] [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: 06/12/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The nutritional quality of plant-based meat analogues compared to traditional meat products has been questioned in recent commentary, particularly in relation to protein quality and micronutrient bioavailability. However, the attributes of specific products within this category are unclear. We therefore undertook a comprehensive assessment of the compositional and functional attributes of v2food® (Sydney, Australia) plant-based mince, including an assessment of the effects of reformulation, including the addition of amino acids, ascorbic acid, and different forms of elemental iron. The protein digestibility and protein quality of v2food® plant-based mince were comparable to beef mince in the standardized INFOGEST system, and favourable effects on microbiota composition and short-chain fatty acid (SCFA) production were demonstrated in an in vitro digestion system. The use of ferrous sulphate as an iron source improved in vitro intestinal iron absorption by ~50% in comparison to other forms of iron (p < 0.05), although levels were ~3-fold lower than beef mince, even in the presence of ascorbic acid. In conclusion, the current study identified some favourable nutritional attributes of plant-based v2food® mince, specifically microbiota and SCFA changes, as well as other areas where further reformulation could be considered to further enhance the bioavailability of key nutrients. Further studies to assess the effect of plant-based meat analogues on health measures in vivo will be important to improve knowledge in this area.
Collapse
Affiliation(s)
- Damien P. Belobrajdic
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Adelaide, SA 5000, Australia; (D.P.B.); (M.C.); (H.B.)
- College of Medicine and Public Health, Health Flinders University, Bedford Park, SA 5042, Australia
| | - Simone Osborne
- Agriculture and Food, Commonwealth Scientific and Industrial Research Organisation, Brisbane, QLD 4067, Australia; (S.O.); (R.A.)
| | - Michael Conlon
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Adelaide, SA 5000, Australia; (D.P.B.); (M.C.); (H.B.)
| | - Henri Brook
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Adelaide, SA 5000, Australia; (D.P.B.); (M.C.); (H.B.)
| | - Rama Addepalli
- Agriculture and Food, Commonwealth Scientific and Industrial Research Organisation, Brisbane, QLD 4067, Australia; (S.O.); (R.A.)
| | - Beverly S. Muhlhausler
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Adelaide, SA 5000, Australia; (D.P.B.); (M.C.); (H.B.)
- School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, SA 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| |
Collapse
|
24
|
Chen LP, Zhang LF, Liu S, Hua H, Zhang L, Liu BC, Wang RR. Ling-Gui-Zhu-Gan decoction ameliorates nonalcoholic fatty liver disease via modulating the gut microbiota. Microbiol Spectr 2024; 12:e0197923. [PMID: 38647315 PMCID: PMC11237417 DOI: 10.1128/spectrum.01979-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 02/27/2024] [Indexed: 04/25/2024] Open
Abstract
Numerous studies have supported that nonalcoholic fatty liver disease (NAFLD) is highly associated with gut microbiota dysbiosis. Ling-Gui-Zhu-Gan decoction (LG) has been clinically used to treat NAFLD, but the underlying mechanism remains unknown. This study investigated the therapeutic effect and mechanisms of LG in mice with NAFLD induced by a high-fat diet (HD). An HD-induced NAFLD mice model was established to evaluate the efficacy of LG followed by biochemical and histopathological analysis. Metagenomics, metabolomics, and transcriptomics were used to explore the structure and metabolism of the gut microbiota. LG significantly improved hepatic function and decreased lipid droplet accumulation in HD-induced NAFLD mice. LG reversed the structure of the gut microbiota that is damaged by HD and improved intestinal barrier function. Meanwhile, the LG group showed a lower total blood bile acids (BAs) concentration, a shifted BAs composition, and a higher fecal short-chain fatty acids (SCFAs) concentration. Furthermore, LG could regulate the hepatic expression of genes associated with the primary BAs biosynthesis pathway and peroxisome proliferator-activated receptor (PPAR) signaling pathway. Our study suggested that LG could ameliorate NAFLD by altering the structure and metabolism of gut microbiota, while BAs and SCFAs are considered possible mediating substances. IMPORTANCE Until now, there has still been no study on the gut microbiota and metabolomics of Ling-Gui-Zhu-Gan decoction (LG) in nonalcoholic fatty liver disease (NAFLD) mouse models. Our study is the first to report on the reshaping of the structure and metabolism of the gut microbiota by LG, as well as explore the potential mechanism underlying the improvement of NAFLD. Specifically, our study demonstrates the potential of gut microbial-derived short-chain fatty acids (SCFAs) and blood bile acids (BAs) as mediators of LG therapy for NAFLD in animal models. Based on the results of transcriptomics, we further verified that LG attenuates NAFLD by restoring the metabolic disorder of BAs via the up-regulation of Fgf15/FXR in the ileum and down-regulation of CYP7A1/FXR in the liver. LG also reduces lipogenesis in NAFLD mice by mediating the peroxisome proliferator-activated receptor (PPAR) signaling pathway, which then contributes to reducing hepatic inflammation and improving intestinal barrier function to treat NAFLD.
Collapse
Affiliation(s)
- Lu-ping Chen
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lin-fang Zhang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Oxford Suzhou Centre for Advanced Research, Suzhou Industrial Park, Jiangsu, China
| | - Shuang Liu
- Shanxi Institute for Function Food, Shanxi Agricultural University, Taiyuan, Shanxi, China
| | - Hua Hua
- Sichuan Institute for Translational Chinese Medicine, Chengdu, China
- Sichuan Academy of Chinese Medical Sciences, Chengdu, China
| | - Lei Zhang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bao-cheng Liu
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui-rui Wang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
25
|
Song X, Wang Z, Xia Y, Chen Z, Wang G, Yang Y, Zhu B, Ai L, Xu H, Wang C. A Cross Talking between the Gut Microbiota and Metabolites of Participants in a Confined Environment. Nutrients 2024; 16:1761. [PMID: 38892694 PMCID: PMC11175105 DOI: 10.3390/nu16111761] [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: 04/27/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Certain workplaces, like deep-sea voyages, subject workers to chronic psychological stress and circadian rhythm disorders due to confined environments and frequent shifts. In this study, participants lived in a strictly controlled confined environment, and we analyzed the effects of a confined environment on gut microbiota and metabolites. The results showed that living in confined environments can significantly alter both the gut microbiota and the gut metabolome, particularly affecting lipid metabolism pathways like glycerophospholipid metabolism. There was a significant reduction in the abundance of Faecalibacterium and Bacteroides, while Blautia, Bifidobacterium, and Collinsella showed significant increases. An association analysis revealed a strong correlation between changes in the gut microbiota and the metabolome. Four upregulated lipid metabolites may serve as biomarkers for damage induced by confined environments, and certain gut microbiota alterations, such as those involving Faecalibacterium and Bacteroides, could be potential psychobiotics or therapeutic targets for enhancing mental health in a confined environment.
Collapse
Affiliation(s)
- Xin Song
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (X.S.); (Y.X.); (G.W.); (Y.Y.); (L.A.)
| | - Ziying Wang
- Naval Medical Center, Naval Medical University, Shanghai 200433, China;
| | - Yongjun Xia
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (X.S.); (Y.X.); (G.W.); (Y.Y.); (L.A.)
| | - Zheng Chen
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (Z.C.); (B.Z.)
| | - Guangqiang Wang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (X.S.); (Y.X.); (G.W.); (Y.Y.); (L.A.)
| | - Yijin Yang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (X.S.); (Y.X.); (G.W.); (Y.Y.); (L.A.)
| | - Beiwei Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (Z.C.); (B.Z.)
| | - Lianzhong Ai
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (X.S.); (Y.X.); (G.W.); (Y.Y.); (L.A.)
| | - Haodan Xu
- Naval Medical Center, Naval Medical University, Shanghai 200433, China;
| | - Chuan Wang
- Naval Medical Center, Naval Medical University, Shanghai 200433, China;
| |
Collapse
|
26
|
Lu K, Zhou Y, He L, Li Y, Shahzad M, Li D. Coprococcus protects against high-fat diet-induced nonalcoholic fatty liver disease in mice. J Appl Microbiol 2024; 135:lxae125. [PMID: 38830802 DOI: 10.1093/jambio/lxae125] [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: 01/30/2024] [Revised: 04/21/2024] [Accepted: 06/01/2024] [Indexed: 06/05/2024]
Abstract
AIMS The incidence of nonalcoholic fatty liver disease (NAFLD) is increasing annually, leading to substantial medical and health burdens. Numerous studies have demonstrated the potential effectiveness of intestinal probiotics as a treatment strategy for NAFLD. Therefore, the objective of this study is to identify a probiotic for the treatment of NAFLD. METHODS AND RESULTS In this study, blood and fecal samples were collected from 41 healthy volunteers and 44 patients diagnosed with NAFLD. Analysis of the 16S rDNA sequencing data and quantitative real-time PCR (RT-qPCR) revealed a significant reduction in the abundance of Coprococcus in NAFLD patients. Subsequent animal experiments demonstrated that Coprococcus was able to effectively reverse liver lipid accumulation, inflammation, and fibrosis induced by a high-fat diet (HFD) in mice. CONCLUSIONS This study provides the first in vivo evidence that Coprococcus is a beneficial bacterium capable of preventing NAFLD and has the same probiotic effect in mice as Lactobacillus GG (LGG), a positive control. Therefore, Coprococcus has the potential to serve as a probiotic for the prevention and treatment of NAFLD in humans.
Collapse
Affiliation(s)
- Kaikai Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, Shaan Xi 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, Shaan Xi 710061, China
| | - Yimeng Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, Shaan Xi 710061, China
- Department of Planned Immunization, Xi'an Center for Disease Control and Prevention, No. 599 Xiying Road, Yanta District, Xi'an 710054 Shaanxi, China
| | - Lei He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, Shaan Xi 710061, China
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center of China, Beijing 100034, China
| | - Ya Li
- Department of Clinical Laboratory, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, Shaanxi 710004, P.R. China
| | - Muhammad Shahzad
- Department of Pharmacology, University of Health Sciences, Lahore 54600, Pakistan
| | - Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, Shaan Xi 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, Shaan Xi 710061, China
| |
Collapse
|
27
|
Prins FM, Collij V, Groot HE, Björk JR, Swarte JC, Andreu-Sánchez S, Jansen BH, Fu J, Harmsen HJM, Zhernakova A, Lipsic E, van der Harst P, Weersma RK, Gacesa R. The gut microbiome across the cardiovascular risk spectrum. Eur J Prev Cardiol 2024; 31:935-944. [PMID: 38060843 DOI: 10.1093/eurjpc/zwad377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 06/04/2024]
Abstract
AIMS Despite treatment advancements, cardiovascular disease remains a leading cause of death worldwide. Identifying new targets is crucial for enhancing preventive and therapeutic strategies. The gut microbiome has been associated with coronary artery disease (CAD), however our understanding of specific changes during CAD development remains limited. We aimed to investigate microbiome changes in participants without clinically manifest CAD with different cardiovascular risk levels and in patients with ST-elevation myocardial infarction (STEMI). METHODS AND RESULTS In this cross-sectional study, we characterized the gut microbiome using metagenomics of 411 faecal samples from individuals with low (n = 130), intermediate (n = 130), and high (n = 125) cardiovascular risk based on the Framingham score, and STEMI patients (n = 26). We analysed diversity, and differential abundance of species and functional pathways while accounting for confounders including medication and technical covariates. Collinsella stercoris, Flavonifractor plautii, and Ruthenibacterium lactatiformans showed increased abundances with cardiovascular risk, while Streptococcus thermophilus was negatively associated. Differential abundance analysis revealed eight species and 49 predicted metabolic pathways that were differently abundant among the groups. In the gut microbiome of STEMI patients, there was a depletion of pathways linked to vitamin, lipid, and amino acid biosynthesis. CONCLUSION We identified four microbial species showing a gradual trend in abundance from low-risk individuals to those with STEMI, and observed differential abundant species and pathways in STEMI patients compared to those without clinically manifest CAD. Further investigation is warranted to gain deeper understanding of their precise role in CAD progression and potential implications, with the ultimate goal of identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Femke M Prins
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Valerie Collij
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Hilde E Groot
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Johannes R Björk
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - J Casper Swarte
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Sergio Andreu-Sánchez
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Bernadien H Jansen
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Jingyuan Fu
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Hermie J M Harmsen
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection prevention, Groningen, The Netherlands
| | - Alexandra Zhernakova
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Erik Lipsic
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rinse K Weersma
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Ranko Gacesa
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| |
Collapse
|
28
|
Mohr AE, Sweazea KL, Bowes DA, Jasbi P, Whisner CM, Sears DD, Krajmalnik-Brown R, Jin Y, Gu H, Klein-Seetharaman J, Arciero KM, Gumpricht E, Arciero PJ. Gut microbiome remodeling and metabolomic profile improves in response to protein pacing with intermittent fasting versus continuous caloric restriction. Nat Commun 2024; 15:4155. [PMID: 38806467 PMCID: PMC11133430 DOI: 10.1038/s41467-024-48355-5] [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: 09/13/2023] [Accepted: 04/26/2024] [Indexed: 05/30/2024] Open
Abstract
The gut microbiome (GM) modulates body weight/composition and gastrointestinal functioning; therefore, approaches targeting resident gut microbes have attracted considerable interest. Intermittent fasting (IF) and protein pacing (P) regimens are effective in facilitating weight loss (WL) and enhancing body composition. However, the interrelationships between IF- and P-induced WL and the GM are unknown. The current randomized controlled study describes distinct fecal microbial and plasma metabolomic signatures between combined IF-P (n = 21) versus a heart-healthy, calorie-restricted (CR, n = 20) diet matched for overall energy intake in free-living human participants (women = 27; men = 14) with overweight/obesity for 8 weeks. Gut symptomatology improves and abundance of Christensenellaceae microbes and circulating cytokines and amino acid metabolites favoring fat oxidation increase with IF-P (p < 0.05), whereas metabolites associated with a longevity-related metabolic pathway increase with CR (p < 0.05). Differences indicate GM and metabolomic factors play a role in WL maintenance and body composition. This novel work provides insight into the GM and metabolomic profile of participants following an IF-P or CR diet and highlights important differences in microbial assembly associated with WL and body composition responsiveness. These data may inform future GM-focused precision nutrition recommendations using larger sample sizes of longer duration. Trial registration, March 6, 2020 (ClinicalTrials.gov as NCT04327141), based on a previous randomized intervention trial.
Collapse
Affiliation(s)
- Alex E Mohr
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - Karen L Sweazea
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
- Center for Evolution and Medicine, College of Liberal Arts and Sciences, Arizona State University, Tempe, AZ, USA
| | - Devin A Bowes
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - Paniz Jasbi
- School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
- Systems Precision Engineering and Advanced Research (SPEAR), Theriome Inc., Phoenix, AZ, USA
| | - Corrie M Whisner
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - Dorothy D Sears
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - Yan Jin
- Center of Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Center of Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Judith Klein-Seetharaman
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
| | - Karen M Arciero
- Human Nutrition and Metabolism Laboratory, Department of Health and Human Physiological Sciences, Skidmore College, Saratoga Springs, NY, USA
| | | | - Paul J Arciero
- Human Nutrition and Metabolism Laboratory, Department of Health and Human Physiological Sciences, Skidmore College, Saratoga Springs, NY, USA.
- School of Health and Rehabilitation Sciences, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Mussabay K, Kozhakhmetov S, Dusmagambetov M, Mynzhanova A, Nurgaziyev M, Jarmukhanov Z, Vinogradova E, Dusmagambetova A, Daulbaeva A, Chulenbayeva L, Tauekelova A, Bekbossynova M, Kushugulova A. Gut Microbiome and Cytokine Profiles in Post-COVID Syndrome. Viruses 2024; 16:722. [PMID: 38793604 PMCID: PMC11126011 DOI: 10.3390/v16050722] [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: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 05/26/2024] Open
Abstract
Recent studies highlight the crucial role of the gut microbiome in post-infectious complications, especially in patients recovering from severe COVID-19. Our research aimed to explore the connection between gut microbiome changes and the cytokine profile of patients with post-COVID syndrome. Using 16S rRNA amplicon sequencing, we analyzed the composition of the gut microbiome in 60 COVID-19 patients over the course of one year. We also measured the levels of serum cytokines and chemokines using the Milliplex system. Our results showed that severe SARS-CoV-2 infection cases, especially those complicated by pneumonia, induce a pro-inflammatory microbial milieu with heightened presence of Bacteroides, Faecalibacterium, and Prevotella_9. Furthermore, we found that post-COVID syndrome is characterized by a cross-correlation of various cytokines and chemokines MDC, IL-1b, Fractalkine, TNFa, FGF-2, EGF, IL-1RA, IFN-a2, IL-10, sCD40L, IL-8, Eotaxin, IL-12p40, and MIP-1b as well as a shift in the gut microbiome towards a pro-inflammatory profile. At the functional level, our analysis revealed associations with post-COVID-19 in homolactic fermentation, pentose phosphate, NAD salvage, and flavin biosynthesis. These findings highlight the intricate interplay between the gut microbiota, their metabolites, and systemic cytokines in shaping post-COVID symptoms. Unraveling the gut microbiome's role in post-infectious complications opens avenues for new treatments for those patients with prolonged symptoms.
Collapse
Affiliation(s)
- Karakoz Mussabay
- Department of Microbiology and Virology Named after Sh.I.Sarbasova, Astana Medical University, Astana 010000, Kazakhstan; (M.D.); (A.D.)
| | - Samat Kozhakhmetov
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (S.K.); (M.N.); (Z.J.); (E.V.); (L.C.)
| | - Marat Dusmagambetov
- Department of Microbiology and Virology Named after Sh.I.Sarbasova, Astana Medical University, Astana 010000, Kazakhstan; (M.D.); (A.D.)
| | - Aitolkyn Mynzhanova
- Department of Pediatric Infectious Diseases, Astana Medical University, Astana 010000, Kazakhstan; (A.M.); (A.D.)
| | - Madiyar Nurgaziyev
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (S.K.); (M.N.); (Z.J.); (E.V.); (L.C.)
| | - Zharkyn Jarmukhanov
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (S.K.); (M.N.); (Z.J.); (E.V.); (L.C.)
| | - Elizaveta Vinogradova
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (S.K.); (M.N.); (Z.J.); (E.V.); (L.C.)
| | - Aigul Dusmagambetova
- Department of Microbiology and Virology Named after Sh.I.Sarbasova, Astana Medical University, Astana 010000, Kazakhstan; (M.D.); (A.D.)
| | - Aiganym Daulbaeva
- Department of Pediatric Infectious Diseases, Astana Medical University, Astana 010000, Kazakhstan; (A.M.); (A.D.)
| | - Laura Chulenbayeva
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (S.K.); (M.N.); (Z.J.); (E.V.); (L.C.)
| | - Ainur Tauekelova
- National Research Cardiac Surgery Center, Astana 010000, Kazakhstan; (A.T.); (M.B.)
| | | | - Almagul Kushugulova
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (S.K.); (M.N.); (Z.J.); (E.V.); (L.C.)
| |
Collapse
|
30
|
Schwenger KJP, Sharma D, Ghorbani Y, Xu W, Lou W, Comelli EM, Fischer SE, Jackson TD, Okrainec A, Allard JP. Links between gut microbiome, metabolome, clinical variables and non-alcoholic fatty liver disease severity in bariatric patients. Liver Int 2024; 44:1176-1188. [PMID: 38353022 DOI: 10.1111/liv.15864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/25/2024] [Accepted: 01/28/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND AND AIMS Bacterial species and microbial pathways along with metabolites and clinical parameters may interact to contribute to non-alcoholic fatty liver disease (NAFLD) and disease severity. We used integrated machine learning models and a cross-validation approach to assess this interaction in bariatric patients. METHODS 113 patients undergoing bariatric surgery had clinical and biochemical parameters, blood and stool metabolite measurements as well as faecal shotgun metagenome sequencing to profile the intestinal microbiome. Liver histology was classified as normal liver obese (NLO; n = 30), simple steatosis (SS; n = 41) or non-alcoholic steatohepatitis (NASH; n = 42); fibrosis was graded F0 to F4. RESULTS We found that those with NASH versus NLO had an increase in potentially harmful E. coli, a reduction of potentially beneficial Alistipes putredinis and an increase in ALT and AST. There was higher serum glucose, faecal 3-(3-hydroxyphenyl)-3-hydroxypropionic acid and faecal cholic acid and lower serum glycerophospholipids. In NAFLD, those with severe fibrosis (F3-F4) versus F0 had lower abundance of anti-inflammatory species (Eubacterium ventriosum, Alistipes finegoldii and Bacteroides dorei) and higher AST, serum glucose, faecal acylcarnitines, serum isoleucine and homocysteine as well as lower serum glycerophospholipids. Pathways involved with amino acid biosynthesis and degradation were significantly more represented in those with NASH compared to NLO, with severe fibrosis having an overall stronger significant association with Superpathway of menaquinol-10 biosynthesis and Peptidoglycan biosynthesis IV. CONCLUSIONS In bariatric patients, NASH and severe fibrosis were associated with specific bacterial species, metabolic pathways and metabolites that may contribute to NAFLD pathogenesis and disease severity.
Collapse
Affiliation(s)
| | - Divya Sharma
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yasaman Ghorbani
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Wei Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Wendy Lou
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Elena M Comelli
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Sandra E Fischer
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Timothy D Jackson
- Division of General Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Allan Okrainec
- Division of General Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Johane P Allard
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Yang K, Zeng J, Wu H, Liu H, Ding Z, Liang W, Wu L, Lin Z, Huang W, Xu J, Dong F. Nonalcoholic Fatty Liver Disease: Changes in Gut Microbiota and Blood Lipids. J Clin Transl Hepatol 2024; 12:333-345. [PMID: 38638378 PMCID: PMC11022063 DOI: 10.14218/jcth.2023.00199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 10/10/2023] [Accepted: 11/29/2023] [Indexed: 04/20/2024] Open
Abstract
Background and Aims The global prevalence of nonalcoholic fatty liver disease (NAFLD) is 25%. This study aimed to explore differences in the gut microbial community and blood lipids between normal livers and those affected by NAFLD using 16S ribosomal deoxyribonucleic acid sequencing. Methods Gut microbiome profiles of 40 NAFLD and 20 non-NAFLD controls were analyzed. Information about four blood lipids and 13 other clinical features was collected. Patients were divided into three groups by ultrasound and FibroScan, those with a normal liver, mild FL (FL1), and moderate-to-severe FL (FL2). FL1 and FL2 patients were divided into two groups, those with either hyperlipidemia or non-hyperlipidemia based on their blood lipids. Potential keystone species within the groups were identified using univariate analysis and a specificity-occupancy plot. Significant difference in biochemical parameters ion NAFLD patients and healthy individuals were identified by detrended correspondence analysis and canonical correspondence analysis. Results Decreased gut bacterial diversity was found in patients with NAFLD. Firmicutes/Bacteroidetes decreased as NAFLD progressed. Faecalibacterium and Ruminococcus 2 were the most representative fatty-related bacteria. Glutamate pyruvic transaminase, aspartate aminotransferase, and white blood cell count were selected as the most significant biochemical indexes. Calculation of areas under the curve identified two microbiomes combined with the three biochemical indexes that identified normal liver and FL2 very well but performed poorly in diagnosing FL1. Conclusions Faecalibacterium and Ruminococcus 2, combined with glutamate pyruvic transaminase, aspartate aminotransferase, and white blood cell count distinguished NAFLD. We speculate that regulating the health of gut microbiota may release NAFLD, in addition to providing new targets for clinicians to treat NAFLD.
Collapse
Affiliation(s)
| | | | - Huaiyu Wu
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen,Guangdong, China
| | - Huiyu Liu
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen,Guangdong, China
| | - Zhimin Ding
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen,Guangdong, China
| | - Weiyu Liang
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen,Guangdong, China
| | - Linghu Wu
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen,Guangdong, China
| | - Ziwei Lin
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen,Guangdong, China
| | - Wenhui Huang
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen,Guangdong, China
| | - Jinfeng Xu
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen,Guangdong, China
| | - Fajin Dong
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen,Guangdong, China
| |
Collapse
|
32
|
Vázquez-Cuesta S, Lozano García N, Rodríguez-Fernández S, Fernández-Avila AI, Bermejo J, Fernández-Avilés F, Muñoz P, Bouza E, Reigadas E. Impact of the Mediterranean Diet on the Gut Microbiome of a Well-Defined Cohort of Healthy Individuals. Nutrients 2024; 16:793. [PMID: 38542704 PMCID: PMC10974552 DOI: 10.3390/nu16060793] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 01/04/2025] Open
Abstract
A comprehensive understanding of gut microbiota in a clearly defined group of healthy individuals is essential when making meaningful comparisons with various diseases. The Mediterranean diet (MD), renowned for its potential health benefits, and the influence of adherence thereto on gut microbiota have become a focus of research. Our aim was to elucidate the impact of adherence to the MD on gut microbiota composition in a well-defined cohort. In this prospective study, healthy volunteers completed a questionnaire to provide demographic data, medical history, and dietary intake. Adherence was evaluated using the Med-DQI. The V4 region of the 16S rRNA gene was sequenced. Analysis of sequencing data and statistical analysis were performed using MOTHUR software and R. The study included 60 patients (51.7% females). Adherence correlated with alpha diversity, and higher values were recorded in good adherers. Good adherers had a higher abundance of Paraprevotella and Bacteroides (p < 0.001). Alpha diversity correlated inversely with fat intake and positively with non-starch polysaccharides (NSPs). Evenness correlated inversely with red meat intake and positively with NSPs. Predicted functional analysis highlighted metabolic pathway differences based on adherence to the MD. In conclusion, our study adds useful information on the relationship between the MD and the gut microbiome.
Collapse
Affiliation(s)
- Silvia Vázquez-Cuesta
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Biochemistry and Molecular Biology Department, School of Biology, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| | - Nuria Lozano García
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
| | - Sara Rodríguez-Fernández
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
| | - Ana I. Fernández-Avila
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Javier Bermejo
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Francisco Fernández-Avilés
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES CB06/06/0058), 28029 Madrid, Spain
| | - Emilio Bouza
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES CB06/06/0058), 28029 Madrid, Spain
| | - Elena Reigadas
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| |
Collapse
|
33
|
Sheng D, Li P, Xiao Z, Li X, Liu J, Xiao B, Liu W, Zhou L. Identification of bidirectional causal links between gut microbiota and narcolepsy type 1 using Mendelian randomization. Sleep 2024; 47:zsae004. [PMID: 38174762 DOI: 10.1093/sleep/zsae004] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/16/2023] [Indexed: 01/05/2024] Open
Abstract
STUDY OBJECTIVES Narcolepsy type 1 (NT1), characterized by cataplexy and orexin deficiency, is a rare and frequently debilitating neurological disorder. It has been noted to have connections with the gut microbiota, yet the exact causal relationships remain unclear. METHODS We conducted a comprehensive bidirectional Mendelian randomization (MR) study to rigorously investigate the causal links between the gut microbiota and NT1, utilizing genetic datasets from the MiBioGen consortium and FinnGen consortium, respectively. The inverse-variance weighted (IVW) method was employed to obtain the primary MR estimates, supplemented by several alternative methods as well as sensitivity analyses including Cochran's Q, MR-Egger, MR pleiotropy residual sum and outlier, leave-one-out, and genetic colocalization. RESULTS Our findings indicated that an increased relative abundance of five genera including Blautia (p = 4.47E-5), Collinsella (p = 0.036), Gordonibacter (p = 0.047), Hungatella (p = 0.015), and Lachnospiraceae UCG010 (p = 0.027) may be associated with a decreased risk of NT1. Conversely, an increased relative abundance of class Betaproteobacteria (p = 0.032), genus Alloprevotella (p = 0.009), and genus Ruminiclostridium6 (p = 0.029) may potentially heighten the risk of NT1. The onset of NT1 may lead to a decrease in the relative abundance of genus Eubacterium eligens group (p = 0.022), while a increase in the family Family XI (p = 0.009), genus Hungatella (p = 0.005), genus Prevotella (p = 0.013), and unknown genus id.2001 (p = 0.019). These findings remained robust under all sensitivity analyses. CONCLUSIONS Our results offer robust evidence for the bidirectional causal links between particular gut microbial taxa and NT1, underscoring the significance of the microbiota-gut-brain axis in the pathological process of NT1.
Collapse
Affiliation(s)
- Dandan Sheng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peihong Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Xiao
- Department of Pathology, First Hospital of Changsha, Changsha, Hunan, China
| | - Xinru Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weiping Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luo Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
34
|
Long Q, Luo F, Li B, Li Z, Guo Z, Chen Z, Wu W, Hu M. Gut microbiota and metabolic biomarkers in metabolic dysfunction-associated steatotic liver disease. Hepatol Commun 2024; 8:e0310. [PMID: 38407327 PMCID: PMC10898672 DOI: 10.1097/hc9.0000000000000310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/05/2023] [Indexed: 02/27/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), a replacement of the nomenclature employed for NAFLD, is the most prevalent chronic liver disease worldwide. Despite its high global prevalence, NAFLD is often under-recognized due to the absence of reliable noninvasive biomarkers for diagnosis and staging. Growing evidence suggests that the gut microbiome plays a significant role in the occurrence and progression of NAFLD by causing immune dysregulation and metabolic alterations due to gut dysbiosis. The rapid advancement of sequencing tools and metabolomics has enabled the identification of alterations in microbiome signatures and gut microbiota-derived metabolite profiles in numerous clinical studies related to NAFLD. Overall, these studies have shown a decrease in α-diversity and changes in gut microbiota abundance, characterized by increased levels of Escherichia and Prevotella, and decreased levels of Akkermansia muciniphila and Faecalibacterium in patients with NAFLD. Furthermore, bile acids, short-chain fatty acids, trimethylamine N-oxide, and tryptophan metabolites are believed to be closely associated with the onset and progression of NAFLD. In this review, we provide novel insights into the vital role of gut microbiome in the pathogenesis of NAFLD. Specifically, we summarize the major classes of gut microbiota and metabolic biomarkers in NAFLD, thereby highlighting the links between specific bacterial species and certain gut microbiota-derived metabolites in patients with NAFLD.
Collapse
|
35
|
Zhu L, Cao F, Hu Z, Zhou Y, Guo T, Yan S, Xie Q, Xia X, Yuan H, Li G, Luo F, Lin Q. Cyanidin-3-O-Glucoside Alleviates Alcoholic Liver Injury via Modulating Gut Microbiota and Metabolites in Mice. Nutrients 2024; 16:694. [PMID: 38474822 DOI: 10.3390/nu16050694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Alcoholic liver disease (ALD) is primarily caused by long-term excessive alcohol consumption. Cyanidin-3-O-glucoside (C3G) is a widely occurring natural anthocyanin with multiple biological activities. This study aims to investigate the effects of C3G isolated from black rice on ALD and explore the potential mechanism. C57BL/6J mice (male) were fed with standard diet (CON) and Lieber-DeCarli liquid-fed (Eth) or supplemented with a 100 mg/kg/d C3G Diet (Eth-C3G), respectively. Our results showed that C3G could effectively ameliorate the pathological structure and liver function, and also inhibited the accumulation of liver lipids. C3G supplementation could partially alleviate the injury of intestinal barrier in the alcohol-induced mice. C3G supplementation could increase the abundance of Norank_f_Muribaculaceae, meanwhile, the abundances of Bacteroides, Blautia, Collinsella, Escherichia-Shigella, Enterococcus, Prevotella, [Ruminococcus]_gnavus_group, Methylobacterium-Methylorubrum, Romboutsia, Streptococcus, Bilophila, were decreased. Spearman's correlation analysis showed that 12 distinct genera were correlated with blood lipid levels. Non-targeted metabolic analyses of cecal contents showed that C3G supplementation could affect the composition of intestinal metabolites, particularly bile acids. In conclusion, C3G can attenuate alcohol-induced liver injury by modulating the gut microbiota and metabolites, suggesting its potential as a functional food ingredient against alcoholic liver disease.
Collapse
Affiliation(s)
- Lingfeng Zhu
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
- Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Fuliang Cao
- College of Forestry, Nanjing Forestry University, Nanjing 210037, China
| | - Zuomin Hu
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Yaping Zhou
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Tianyi Guo
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Sisi Yan
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Qiutao Xie
- Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Xinxin Xia
- Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Hongyan Yuan
- Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Gaoyang Li
- Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Feijun Luo
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Qinlu Lin
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| |
Collapse
|
36
|
Purohit A, Kandiyal B, Kumar S, Pragasam AK, Kamboj P, Talukdar D, Verma J, Sharma V, Sarkar S, Mahajan D, Yadav R, Ahmed R, Nanda R, Dikshit M, Banerjee SK, Shalimar, Das B. Collinsella aerofaciens linked with increased ethanol production and liver inflammation contribute to the pathophysiology of NAFLD. iScience 2024; 27:108764. [PMID: 38313048 PMCID: PMC10837629 DOI: 10.1016/j.isci.2023.108764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/21/2023] [Accepted: 12/15/2023] [Indexed: 02/06/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an emerging global health problem and a potential risk factor for metabolic diseases. The bidirectional interactions between liver and gut made dysbiotic gut microbiome one of the key risk factors for NAFLD. In this study, we reported an increased abundance of Collinsella aerofaciens in the gut of obese and NASH patients living in India. We isolated C. aerofaciens from the fecal samples of biopsy-proven NASH patients and observed that their genome is enriched with carbohydrate metabolism, fatty acid biosynthesis, and pro-inflammatory functions and have the potency to increase ethanol level in blood. An animal study indicated that mice supplemented with C. aerofaciens had increased levels of circulatory ethanol, high levels of hepatic hydroxyproline, triglyceride, and inflammation in the liver. The present findings indicate that perturbation in the gut microbiome composition is a key risk factor for NAFLD.
Collapse
Affiliation(s)
- Ayushi Purohit
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| | - Bharti Kandiyal
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| | - Shakti Kumar
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| | - Agila Kumari Pragasam
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| | - Parul Kamboj
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| | - Daizee Talukdar
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| | - Jyoti Verma
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| | - Vipin Sharma
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| | - Soumalya Sarkar
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| | - Dinesh Mahajan
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| | - Rajni Yadav
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Riya Ahmed
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Ranjan Nanda
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Madhu Dikshit
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| | - Sanjay K. Banerjee
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER-Guwahati), Changsari, Guwahati, Assam 781101, India
| | - Shalimar
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Bhabatosh Das
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121004, India
| |
Collapse
|
37
|
Liu S, Imad S, Hussain S, Xiao S, Yu X, Cao H. Sex, health status and habitat alter the community composition and assembly processes of symbiotic bacteria in captive frogs. BMC Microbiol 2024; 24:34. [PMID: 38262927 PMCID: PMC10804495 DOI: 10.1186/s12866-023-03150-y] [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: 06/28/2023] [Accepted: 12/11/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Frogs are critical economic animals essential to agricultural ecosystem equilibrium. However, Meningitis-like Infectious Disease (MID) often affects them in agricultural settings. While frog-associated microbiota contribute to elemental cycling and immunity, the effects of frog sex and health on gut bacteria remain understudied, and the relationship between frog habitat and soil microbes is unclear. We aimed to determine how frog sex, health status and habitat influence symbiotic bacteria and community assembly mechanism to provide guidance for sustainable frog farming and conservation. RESULTS We employed 16S rRNA sequencing to investigate gut microbiota differences in relation to frog sex and health status. We also compared symbiotic communities in frog-aggregation, native and soybean soil on the farm. Results showed that gut bacterial β-diversity and taxonomy were markedly influenced by frog sex and health. Healthy frogs had more robust gut bacterial metabolism than frogs infected with MID. Cooccurrence network analysis revealed that healthy female frogs had more complex microbial network structure than males; however, diseased males showed the greatest network complexity. The assembly mechanism of gut bacteria in male frogs was dominated by deterministic processes, whereas in female frogs it was dominated by stochastic processes. Among symbiotic bacteria in frog habitat soils, deterministic processes predominantly shaped the community assembly of soybean soil. In particular, soybean soil was enriched in pathogens and nitrogen functions, whereas frog-aggregation soil was markedly increased in sulphur respiration and hydrocarbon degradation. CONCLUSION Our study reveals that sex mainly alters the interaction network and assembly mechanism of frog intestinal bacteria; MID infection significantly inhibits the metabolic functions of intestinal bacteria. Furthermore, diverse frog habitat soils could shape more symbiotic bacteria to benefit frog farming. Our findings provide new horizons for symbiotic bacteria among frogs, which could contribute to sustainable agriculture and ecological balance.
Collapse
Affiliation(s)
- Senlin Liu
- College of Life Sciences/Key Laboratory of Agricultural Environmental Microbiology, Ministry of Agriculture and Rural Affair, Nanjing Agricultural University, 6 Tongwei Road, Nanjing, Jiangsu, 210095, People's Republic of China
- School of Life Sciences, University of Warwick, Gibbet Hill Road, Coventry, UK
| | - Sewar Imad
- College of Life Sciences/Key Laboratory of Agricultural Environmental Microbiology, Ministry of Agriculture and Rural Affair, Nanjing Agricultural University, 6 Tongwei Road, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Sarfraz Hussain
- College of Life Sciences/Key Laboratory of Agricultural Environmental Microbiology, Ministry of Agriculture and Rural Affair, Nanjing Agricultural University, 6 Tongwei Road, Nanjing, Jiangsu, 210095, People's Republic of China
| | | | - Xiaowei Yu
- College of Life Sciences/Key Laboratory of Agricultural Environmental Microbiology, Ministry of Agriculture and Rural Affair, Nanjing Agricultural University, 6 Tongwei Road, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Hui Cao
- College of Life Sciences/Key Laboratory of Agricultural Environmental Microbiology, Ministry of Agriculture and Rural Affair, Nanjing Agricultural University, 6 Tongwei Road, Nanjing, Jiangsu, 210095, People's Republic of China.
| |
Collapse
|
38
|
Su X, Chen S, Liu J, Feng Y, Han E, Hao X, Liao M, Cai J, Zhang S, Niu J, He S, Huang S, Lo K, Zeng F. Composition of gut microbiota and non-alcoholic fatty liver disease: A systematic review and meta-analysis. Obes Rev 2024; 25:e13646. [PMID: 37813400 DOI: 10.1111/obr.13646] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 08/03/2023] [Accepted: 09/09/2023] [Indexed: 10/11/2023]
Abstract
The present systematic review and meta-analysis aimed to summarize the associations between gut microbiota composition and non-alcoholic fatty liver disease. To compare the differences between individuals with or without NAFLD, the standardized mean difference and 95% confidence interval were computed for each α-diversity index and relative abundance of gut microbes. The β-diversity indices were summarized in a qualitative manner. A total of 54 studies with 8894 participants were included. Overall, patients with NAFLD had moderate reduction in α-diversity indices including Shannon (SMD = -0.36, 95% CI = [-0.53, -0.19], p < 0.001) and Chao 1 (SMD = -0.42, 95% CI = [-0.68, -0.17], p = 0.001), but no significant differences were found for Simpson, observed species, phylogenetic diversity, richness, abundance-based coverage estimator, and evenness (p ranged from 0.081 to 0.953). Over 75% of the included studies reported significant differences in β-diversity. Although there was substantial interstudy heterogeneity, especially for analyses at the phylum, class, and family levels, the majority of the included studies showed alterations in the depletion of anti-inflammatory microbes (i.e., Ruminococcaceae and Coprococcus) and the enrichment of proinflammatory microbes (i.e., Fusobacterium and Escherichia) in patients with NAFLD. Perturbations in gut microbiota were associated with NAFLD, commonly reflected by a reduction in beneficial species and an increase in the pathogenic species.
Collapse
Affiliation(s)
- Xin Su
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Shiyun Chen
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jiazi Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yonghui Feng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Eerdun Han
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiaolei Hao
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Minqi Liao
- Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Epidemiology, Neuherberg, PR, Germany
| | - Jun Cai
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Shiwen Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jianxiang Niu
- General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Shihua He
- Department of Infectious Disease, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Shaofen Huang
- Shenzhen Qianhai Shekou Free Zone Hospital, Shenzhen, China
| | - Kenneth Lo
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Research Institute for Future Food, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Fangfang Zeng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
39
|
Banerjee G, Papri SR, Satapathy SK, Banerjee P. Akkermansia muciniphila - A Potential Next-generation Probiotic for Non-alcoholic Fatty Liver Disease. Curr Pharm Biotechnol 2024; 25:426-433. [PMID: 37724669 DOI: 10.2174/1389201025666230915103052] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/20/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a spectrum of liver conditions, and its growing prevalence is a serious concern worldwide, especially in Western countries. Researchers have pointed out several genetic mutations associated with NAFLD; however, the imbalance of the gut microbial community also plays a critical role in the progression of NAFLD. Due to the lack of approved medicine, probiotics gain special attention in controlling metabolic disorders like NAFLD. Among these probiotics, Akkermansia muciniphila (a member of natural gut microflora) is considered one of the most efficient and important bacterium in maintaining gut health, energy homeostasis, and lipid metabolism. In this perspective, we discussed the probable molecular mechanism of A. muciniphila in controlling the progression of NAFLD and restoring liver health. The therapeutic potential of A. muciniphila in NAFLD has been tested primarily on animal models, and thus, more randomized human trials should be conducted to prove its efficacy.
Collapse
Affiliation(s)
- Goutam Banerjee
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Suraya R Papri
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Sanjaya K Satapathy
- 2Department of Medicine, Northwell Health Center for Liver Disease & Transplantation, North Shore, University Hospital/Northwell Health, 400 Community Drive, Manhasset, NY 11030, USA
| | - Pratik Banerjee
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| |
Collapse
|
40
|
Zhao C, Li H, Gao C, Tian H, Guo Y, Liu G, Li Y, Liu D, Sun B. Moringa oleifera leaf polysaccharide regulates fecal microbiota and colonic transcriptome in calves. Int J Biol Macromol 2023; 253:127108. [PMID: 37776927 DOI: 10.1016/j.ijbiomac.2023.127108] [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: 04/19/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
This study investigated the effects of Moringa oleifera polysaccharide on growth performance indicators, serum biochemical indicators, immune organ indicators, colonic morphology, colonic microbiomics and colonic transcriptomics in newborn calves. 21 newborn calves were randomly divided into three groups of 7 calves per treatment group: control group (no Moringa oleifera polysaccharide addition); low-dose group (Moringa oleifera polysaccharide 0.5 g/kg); and high-dose group (Moringa oleifera polysaccharide 1 g/kg). This trial used gavage to feed MOP to calves. The test lasted 8 weeks. Calves were humanely electroshocked on the last day of the trial and slaughtered afterwards. Thymus, spleen, blood and colonic contents were collected for further testing. The results of this trial showed that MOP significantly increased the body weight of newborn calves and reduced the rate of calf diarrhea, thus promoting calf growth. Fecal scores showed a linear decrease with the addition of MOP. In terms of serum biochemistry, feeding MOP significantly increased serum ALB levels in a linear fashion. In terms of serum antioxidants, feeding MOP linearly increased CAT and T-AOC levels and decreased MDA concentrations, and in terms of serum immunity, feeding MOP linearly increased IgA, IgG, and IgM levels. At the same time, MOP regulated the abundance of Firmicutes and Bacteroidetes in the intestinal tract of calves, which reduced the occurrence of diarrhea. In addition, moringa polysaccharide could regulate genes related to inflammatory signaling pathways such as MAPK signaling pathway, TGF-beta signaling pathway, PI3K-Akt signaling pathway and TNF signaling pathway in calves' intestine to reduce the occurrence of intestinal inflammation. In conclusion, MOP can be used as a novel ruminant additive for the prevention of enteritis in calves.
Collapse
Affiliation(s)
- Chao Zhao
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hangfan Li
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Chongya Gao
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hanchen Tian
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yongqing Guo
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Guangbin Liu
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yaokun Li
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Dewu Liu
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Baoli Sun
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
41
|
Long L, Zhao X, Chen J, Wang Z, Tang Y, Huang J, Yin Y. Piglet growth performance improved by dietary supplementation of porous or nano particles of zinc oxide may be related to the gut microbiota. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 15:159-172. [PMID: 38023375 PMCID: PMC10679868 DOI: 10.1016/j.aninu.2023.08.011] [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: 12/15/2022] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 12/01/2023]
Abstract
Previous studies on porous or nano particles zinc oxide (ZnO) in the piglets have mainly focused on growth performance and intestinal inflammation, but have scarcely explored the efficacy on gut microbiota. In addition, the efficacy of nano particles ZnO, which is related to its product quality, remains undefined. This study aimed to determine the efficacy of dietary 500 mg/kg porous or nano particles ZnO on the growth performance and gut microbiota of the weaned piglets. A total of 128 weaned piglets were randomly assigned to the dietary groups: NC (basal diet), PC (basal diet + 3,000 mg/kg conventional ZnO), 500HiZ (basal diet + 500 mg/kg porous particles ZnO), and 500ZNP (basal diet + 500 mg/kg nano particles ZnO). Compared with the NC diet group, both 500HiZ and 500ZNP increased (P < 0.05) average daily feed intake (1 to 28 d) and average daily gain (1 to 28 d), and the 500ZNP tended to decrease feed to gain ratio (F:G ratio, 1 to 28 d) (P = 0.09). Both 500HiZ and 500ZNP decreased crypt depth of the ileum and increased claudin-2 in the duodenum and zonula occludens-1 in the ileum (P < 0.05). Moreover, both 500HiZ and 500ZNP decreased IL-1β and tumor necrosis factor-α (TNF-α) in the jejunum and decreased TNF-α and IL-6 in the ileum (P < 0.05). Both 500HiZ and 500ZNP increased microbial β-diversity index in the ileum and microbial α-diversity indices in the colon of piglets (P < 0.05). The probiotic genera Coprococcus (500ZNP) and Blautia (500HiZ) were positively correlated with the F:G ratio (1 to 28 d) in colon of piglets (P < 0.05). In addition, 500HiZ promoted mitochondrial fusion protein 1 (MFN1) and zinc transporter-1 (ZnT-1) in the jejunum (P < 0.05), whilst 500ZNP decreased MFN1 in the jejunum and ZnT-1 in the ileum (P < 0.05). In summary, both 500HiZ and 500ZNP improved the growth performance of piglets, which is likely via the genera Blautia and Coprococcus, respectively. Both 500HiZ and 500ZNP improved barrier function and inflammation of the intestine, and 500HiZ achieved better efficacy than 500ZNP on intestine mitochondrial functions.
Collapse
Affiliation(s)
- Lina Long
- School of Life Science and Engineering, Foshan University, Foshan 528231, China
| | - Xichen Zhao
- Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jie Chen
- Foshan Guangmuxing Feed Co., Ltd, Foshan 528000, China
| | - Zixi Wang
- School of Life Science and Engineering, Foshan University, Foshan 528231, China
| | - Yanfang Tang
- School of Life Science and Engineering, Foshan University, Foshan 528231, China
| | - Jian Huang
- School of Life Science and Engineering, Foshan University, Foshan 528231, China
| | - Yulong Yin
- Key Laboratory for Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha 410125, China
| |
Collapse
|
42
|
Munjoma PT, Chandiwana P, Wyss J, Mazhandu AJ, Jordi SBU, Gutsire R, Katsidzira L, Yilmaz B, Misselwitz B, Duri K. Immune activation and inflammation in lactating women on combination antiretroviral therapy: role of gut dysfunction and gut microbiota imbalance. Front Immunol 2023; 14:1280262. [PMID: 38045684 PMCID: PMC10693333 DOI: 10.3389/fimmu.2023.1280262] [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: 08/21/2023] [Accepted: 10/17/2023] [Indexed: 12/05/2023] Open
Abstract
Introduction Combination antiretroviral therapy (cART) effectively controls HIV; however, chronic low-level viremia and gut microbiota dysbiosis remain significant drivers of gut and systemic inflammation. In this study, we explored the relationship between gut microbiota composition, intestinal inflammation, microbial translocation, and systemic inflammation in women on cART in Sub-Saharan Africa. Methods We conducted a study in HIV-infected and HIV-uninfected lactating women followed up at 6 weeks and 6 months postpartum in Harare, Zimbabwe. We used 16S ribosomal Ribonucleic Acid (rRNA) sequencing and MesoScale Discovery V-Plex assays to examine the gut microbiome and to quantify plasma inflammatory biomarkers, respectively. In addition, we measured fecal calprotectin, plasma lipopolysaccharide-binding protein (LBP), and soluble cluster of differentiation 14 (sCD14) by enzyme-linked immunosorbent assay to assess gut inflammation, microbial translocation, and monocyte/macrophage activation. Results A group of 77 lactating women were studied, of which 35% were HIV-infected. Fecal calprotectin levels were similar by HIV status at both follow-up time points. In the HIV-infected group at 6 weeks postpartum, fecal calprotectin was elevated: median (interquartile range) [158.1 µg/g (75.3-230.2)] in women who had CD4+ T-lymphocyte counts <350 cells/µL compared with those with ≥350 cells/µL [21.1 µg/g (0-58.4)], p = 0.032. Plasma sCD14 levels were significantly higher in the HIV-infected group at both 6 weeks and 6 months postpartum, p < 0.001. Plasma LBP levels were similar, but higher levels were observed in HIV-infected women with elevated fecal calprotectin. We found significant correlations between fecal calprotectin, LBP, and sCD14 with proinflammatory cytokines. Gut microbial alpha diversity was not affected by HIV status and was not affected by use of antibiotic prophylaxis. HIV significantly affected microbial beta diversity, and significant differences in microbial composition were noted. The genera Slackia and Collinsella were relatively more abundant in the HIV-infected group, whereas a lower relative abundance of Clostriduim sensu_stricto_1 was observed. Our study also found correlations between gut microbial taxa abundance and systemic inflammatory biomarkers. Discussion and conclusion HIV-infected lactating women had increased immune activation and increased microbial translocation associated with increased gut inflammation. We identified correlations between the gut inflammation and microbial composition, microbial translocation, and systemic inflammation. The interplay of these parameters might affect the health of this vulnerable population.
Collapse
Affiliation(s)
- Privilege Tendai Munjoma
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Panashe Chandiwana
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Jacqueline Wyss
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research, Maurice Müller Laboratories, University of Bern, Bern, Switzerland
| | - Arthur John Mazhandu
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Sebastian Bruno Ulrich Jordi
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research, Maurice Müller Laboratories, University of Bern, Bern, Switzerland
| | - Rutendo Gutsire
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Leolin Katsidzira
- Department of Internal Medicine, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research, Maurice Müller Laboratories, University of Bern, Bern, Switzerland
| | - Benjamin Misselwitz
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research, Maurice Müller Laboratories, University of Bern, Bern, Switzerland
| | - Kerina Duri
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| |
Collapse
|
43
|
Aparicio A, Sun Z, Gold DR, Litonjua AA, Weiss ST, Lee-Sarwar K, Liu YY. Genotype-microbiome-metabolome associations in early childhood, and their link to BMI and childhood obesity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.13.23298467. [PMID: 38014043 PMCID: PMC10680902 DOI: 10.1101/2023.11.13.23298467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The influence of genotype on defining the human gut microbiome has been extensively studied, but definite conclusions have not yet been found. To fill this knowledge gap, we leverage data from children enrolled in the Vitamin D Antenatal Asthma Reduction Trial (VDAART) from 6 months to 8 years old. We focus on a pool of 12 genes previously found to be associated with the gut microbiome in independent studies, establishing a Bonferroni corrected significance level of p-value < 2.29 × 10 -6 . We identified significant associations between SNPs in the FHIT gene (known to be associated with obesity and type 2 diabetes) and obesity-related microbiome features, and the children's BMI through their childhood. Based on these associations, we defined a set of SNPs of interest and a set of taxa of interest. Taking a multi-omics approach, we integrated plasma metabolome data into our analysis and found simultaneous associations among children's BMI, the SNPs of interest, and the taxa of interest, involving amino acids, lipids, nucleotides, and xenobiotics. Using our association results, we constructed a quadripartite graph where each disjoint node set represents SNPs in the FHIT gene, microbial taxa, plasma metabolites, or BMI measurements. Network analysis led to the discovery of patterns that identify several genetic variants, microbial taxa and metabolites as new potential markers for obesity, type 2 diabetes, or insulin resistance risk.
Collapse
|
44
|
Alkader DAA, Asadi N, Solangi U, Singh R, Rasuli SF, Farooq MJ, Raheela FNU, Waseem R, Gilani SM, Abbas K, Ahmed M, Tanoh DB, Shah HH, Dulal A, Hussain MS, Talpur AS. Exploring the role of gut microbiota in autoimmune thyroid disorders: a systematic review and meta-analysis. Front Endocrinol (Lausanne) 2023; 14:1238146. [PMID: 37964972 PMCID: PMC10641821 DOI: 10.3389/fendo.2023.1238146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/15/2023] [Indexed: 11/16/2023] Open
Abstract
Background Autoimmune thyroid diseases (AITDs) are characterized by unique immune responses against thyroid antigens and persist over time. The most common types of AITDs are Graves' disease (GD) and Hashimoto's thyroiditis (HT). There is mounting evidence that changes in the microbiota may play a role in the onset and development of AITDs. Objective The purpose of this comprehensive literature study was to answer the following query: Is there a difference in microbiota in those who have AITDs? Methods According to the standards set out by the PRISMA statement, 16 studies met the requirements for inclusion after being screened for eligibility. Results The Simpson index was the only diversity measure shown to be considerably lower in patients with GD compared to healthy participants, whereas all other indices were found to be significantly greater in patients with HT. The latter group, however, showed a greater relative abundance of Bacteroidetes and Actinobacteria at the phylum level, and consequently of Prevotella and Bifidobacterium at the genus level. The strongest positive and negative relationships were seen for thyroid peroxidase antibodies and bacterial load. Conclusion Overall, both GD and HT patients showed significant changes in the gut microbiota's diversity and composition. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42023432455.
Collapse
Affiliation(s)
| | | | - Uzma Solangi
- Department of Medicine, Liaquat University of Medical & Health Sciences, Jamshoro, Pakistan
| | - Ransherjit Singh
- Department of Medicine, Civil Hospital Karachi, Karachi, Pakistan
| | | | - Muhammad Jawad Farooq
- Department of Medicine, Liaquat University of Medical & Health Sciences, Jamshoro, Pakistan
| | - F. N. U. Raheela
- Department of Medicine, University of Toledo, Toledo, OH, United States
| | - Radeyah Waseem
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Syed Mujahid Gilani
- Department of Medicine, Shaheed Zulfiqar Ali Bhutto Medical University (SZABMU), Islamabad, Pakistan
| | - Kiran Abbas
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Moiz Ahmed
- Department of Medicine, National Institute of Cardiovascular Diseases, Karachi, Pakistan
| | - Desmond Boakye Tanoh
- Department of Medicine, Insight Hospital and Medical Center Chicago, Chicago, IL, United States
| | - Hussain Haider Shah
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Ayusha Dulal
- Department of Human Physiology, Nepalese Army Institute of Health Science, Kathmandu, Nepal
| | | | - Abdul Subhan Talpur
- Department of Medicine, Liaquat University of Medical & Health Sciences, Jamshoro, Pakistan
| |
Collapse
|
45
|
Korobeinikova AV, Zlobovskaya OA, Sheptulina AF, Ashniev GA, Bobrova MM, Yafarova AA, Akasheva DU, Kabieva SS, Bakoev SY, Zagaynova AV, Lukashina MV, Abramov IA, Pokrovskaya MS, Doludin YV, Tolkacheva LR, Kurnosov AS, Zyatenkova EV, Lavrenova EA, Efimova IA, Glazunova EV, Kiselev AR, Shipulin GA, Kontsevaya AV, Keskinov AA, Yudin VS, Makarov VV, Drapkina OM, Yudin SM. Gut Microbiota Patterns in Patients with Non-Alcoholic Fatty Liver Disease: A Comprehensive Assessment Using Three Analysis Methods. Int J Mol Sci 2023; 24:15272. [PMID: 37894951 PMCID: PMC10607775 DOI: 10.3390/ijms242015272] [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: 08/17/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 10/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered the most common chronic liver disease worldwide, affecting nearly 25% of the global adult population. Increasing evidence suggests that functional and compositional changes in the gut microbiota may contribute to the development and promote the progression of NAFLD. 16S rRNA gene next-generation sequencing is widely used to determine specific features of the NAFLD microbiome, but a complex system such as the gut microbiota requires a comprehensive approach. We used three different approaches: MALDI-TOF-MS of bacterial cultures, qPCR, and 16S NGS sequencing, as well as a wide variety of statistical methods to assess the differences in gut microbiota composition between NAFLD patients without significant fibrosis and the control group. The listed methods showed enrichment in Collinsella sp. and Oscillospiraceae for the control samples and enrichment in Lachnospiraceae (and in particular Dorea sp.) and Veillonellaceae in NAFLD. The families, Bifidobacteriaceae, Lactobacillaceae, and Enterococcaceae (particularly Enterococcus faecium and Enterococcus faecalis), were also found to be important taxa for NAFLD microbiome evaluation. Considering individual method observations, an increase in Candida krusei and a decrease in Bacteroides uniformis for NAFLD patients were detected using MALDI-TOF-MS. An increase in Gracilibacteraceae, Chitinophagaceae, Pirellulaceae, Erysipelatoclostridiaceae, Muribaculaceae, and Comamonadaceae, and a decrease in Acidaminococcaceae in NAFLD were observed with 16S NGS, and enrichment in Fusobacterium nucleatum was shown using qPCR analysis. These findings confirm that NAFLD is associated with changes in gut microbiota composition. Further investigations are required to determine the cause-and-effect relationships and the impact of microbiota-derived compounds on the development and progression of NAFLD.
Collapse
Affiliation(s)
- Anna V. Korobeinikova
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Olga A. Zlobovskaya
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Anna F. Sheptulina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - German A. Ashniev
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Maria M. Bobrova
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Adel A. Yafarova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Dariga U. Akasheva
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Shuanat Sh. Kabieva
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Siroj Yu. Bakoev
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Anjelica V. Zagaynova
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Maria V. Lukashina
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Ivan A. Abramov
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Mariya S. Pokrovskaya
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Yurii V. Doludin
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Larisa R. Tolkacheva
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Alexander S. Kurnosov
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Elena V. Zyatenkova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Evgeniya A. Lavrenova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Irina A. Efimova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Evgeniya V. Glazunova
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Anton R. Kiselev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - German A. Shipulin
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Anna V. Kontsevaya
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Anton A. Keskinov
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Vladimir S. Yudin
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Valentin V. Makarov
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Oxana M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Sergey M. Yudin
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| |
Collapse
|
46
|
Shi X, Li Z, Lin W, Shi W, Hu R, Chen G, Li X, Li X, Zhang S. Altered Intestinal Microbial Flora and Metabolism in Patients with Idiopathic Membranous Nephropathy. Am J Nephrol 2023; 54:451-470. [PMID: 37793354 DOI: 10.1159/000533537] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/31/2023] [Indexed: 10/06/2023]
Abstract
INTRODUCTION Dysbiosis of the intestinal microbiome and related metabolites have been observed in chronic kidney disease, yet their roles in idiopathic membranous nephropathy (IMN) are poorly understood. METHODS In this study, we describe the variation of intestinal bacteria and fecal metabolites in patients with IMN in Chinese population. Stool samples were collected from 41 IMN patients at the beginning of diagnosis confirmation and 41 gender- and age-matched healthy control (HC). Microbial communities are investigated by sequencing of 16S rRNA genes and functional profiles predicted using Tax4Fun, and the correlation between intestinal bacteria and IMN clinical characteristics is also analyzed. Untargeted metabolomic analysis is performed to explore the relationship between colon's microbiota and fecal metabolites. RESULTS IMN gastrointestinal microbiota demonstrates lower richness and diversity compared to HC, and exhibits a marked taxonomic and inferred functional dysbiosis when compared to HC. Some genera are closely related to the clinical parameters, such as Citrobacter and Akkermansia. Twenty characteristic microbial biomarkers are selected to establish a disease prediction model with a diagnostic accuracy of 93.53%. Fecal metabolomics shows that tryptophan metabolism is reduced in IMN patients but uremic toxin accumulation in feces is not noticeable. Fecal microbiota transplantation demonstrates that gut dysbiosis impairs gut permeability in microbiota-depleted mice and induces NOD-like receptor activation in the kidneys. CONCLUSIONS Clarifying the changes in intestinal microbiota in IMN patients will help further know the pathogenesis of this disease, and microbiota-targeted biomarkers will provide a potentially powerful tool for diagnosing and treating IMN.
Collapse
Affiliation(s)
- Xiaohu Shi
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhaojun Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weifeng Lin
- Department of Nephropathy, Peking University Third Hospital, Beijing, China
| | - Wenying Shi
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, College of Pharmacy, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Rongrong Hu
- Department of Nephropathy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Gang Chen
- Department of Nephropathy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuemei Li
- Department of Nephropathy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuewang Li
- Department of Nephropathy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Sen Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
47
|
Chhimwal J, Anand P, Mehta P, Swarnkar MK, Patial V, Pandey R, Padwad Y. Metagenomic signatures reveal the key role of phloretin in amelioration of gut dysbiosis attributed to metabolic dysfunction-associated fatty liver disease by time-dependent modulation of gut microbiome. Front Microbiol 2023; 14:1210517. [PMID: 37744933 PMCID: PMC10516607 DOI: 10.3389/fmicb.2023.1210517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
The importance of gut-liver axis in the pathophysiology of metabolic dysfunction-associated fatty liver disease (MAFLD) is being investigated more closely in recent times. However, the inevitable changes in gut microbiota during progression of the disease merits closer look. The present work intends to assess the time-dependent gut dysbiosis in MAFLD, its implications in disease progression and role of plant-derived prebiotics in its attenuation. Male C57BL/6J mice were given western diet (WD) for up to 16 weeks and phloretin was administered orally. The fecal samples of mice were collected every fourth week for 16 weeks. The animals were sacrificed at the end of the study and biochemical and histological analyses were performed. Further, 16S rRNA amplicon sequencing analysis was performed to investigate longitudinal modification of gut microbiome at different time points. Findings of our study corroborate that phloretin alleviated the metabolic changes and mitigated circulating inflammatory cytokines levels. Phloretin treatment resists WD induced changes in microbial diversity of mice and decreased endotoxin content. Prolonged exposure of WD changed dynamics of gut microbiota abundance and distribution. Increased abundance of pathogenic taxa like Desulfovibrionaceae, Peptostreptococcus, Clostridium, and Terrisporobacter was noted. Phloretin treatment not only reversed this dysbiosis but also modulated taxonomic signatures of beneficial microbes like Ruminococcus, Lactobacillus, and Alloprevotella. Therefore, the potential of phloretin to restore gut eubiosis could be utilized as an intervention strategy for the prevention of MAFLD and related metabolic disorders.
Collapse
Affiliation(s)
- Jyoti Chhimwal
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Prince Anand
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Priyanka Mehta
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Mohit Kumar Swarnkar
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajesh Pandey
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Yogendra Padwad
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
48
|
Shi X, Zhang Y, Shi Y, Zhang Q, Duan H, Liu J, Yang B, Zhang Y. Analysis of the alleviating effect of modified Huangqi Chifeng decoction on rats with focal segmental glomerulosclerosis based on gut microbiota and fecal metabolomics. J Appl Microbiol 2023; 134:lxad205. [PMID: 37675978 DOI: 10.1093/jambio/lxad205] [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: 08/20/2023] [Accepted: 09/06/2023] [Indexed: 09/08/2023]
Abstract
AIMS To investigate the reno-protective effects of modified Huangqi Chifeng decoction (MHCD) on focal segmental glomerulosclerosis (FSGS) rats, and the underlying mechanisms of systemic regulation of gut microbiota and metabolite profiles. METHODS AND RESULTS A rat FSGS model was established via unilateral nephrectomy plus doxorubicin injections. Rats were divided into sham, FSGS, and MHCD groups from which urine, blood, and histological tests were conducted. Fecal microbiotas were identified via 16S rRNA gene sequencing. Fecal metabolomics allowed for metabolic pathways analysis. Biochemical indices and pathological examination revealed that MHCD treatment improved the symptoms of FSGS, and corrected dysbiosis of gut microbiota, enriched the abundance of Bifidobacterium, Odoribacter, Christensella, Oscillospira, and reduced that of harmful bacteria such as Collinsella and Coprobacterilus at the genus level. Fecal metabolomic profiles revealed 152 different metabolites between the FSGS and sham groups, which are mainly enriched in signaling pathways like arachidonic acid, serotonergic synapse, and oxytocin. Besides, 93 differential metabolites between MHCD and FSGS groups were identified, which are mainly enriched in signaling pathways like steroid hormone biosynthesis, prostate cancer, and linoleic acid metabolism. Spearman's correlation analysis showed a correlation between differential fecal metabolites and enriched gut microbiota or serum biochemical parameters. CONCLUSIONS MHCD may exert a reno-protective effect by regulating the gut microbiome and metabolite profiles in FSGS rats.
Collapse
Affiliation(s)
- Xiujie Shi
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yifan Zhang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Graduate School of Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yue Shi
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Qi Zhang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Hangyu Duan
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Jing Liu
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Graduate School of Beijing University of Chinese Medicine, Beijing 100029, China
| | - Bin Yang
- Department of Pathology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yu Zhang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Xin-Huangpu Joint Innovation Institute of Chinese Medicine, Guangzhou 510535, China
| |
Collapse
|
49
|
Kei N, Wong VWS, Lauw S, You L, Cheung PCK. Utilization of Food-Derived β-Glucans to Prevent and Treat Non-Alcoholic Fatty Liver Disease (NAFLD). Foods 2023; 12:3279. [PMID: 37685211 PMCID: PMC10486587 DOI: 10.3390/foods12173279] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease nowadays. Currently, there is no officially approved drug to treat NAFLD. In view of the increasing global prevalence of NAFLD and an absence of treatments, the development of effective treatments is of utmost importance. β-glucan, a natural bioactive polysaccharide, has demonstrated hepatoprotective effects in NAFLD prevention and treatment. This review solely focuses on gathering the published preclinical animal studies that demonstrated the anti-liver injury, anti-steatotic, anti-inflammatory, anti-fibrotic, and antioxidant activities of β-glucan. The impact of β-glucan on gut microbiota and its metabolites including short-chain fatty acids and bile acids as the underlying mechanism for its bioactive beneficial effect on NAFLD is also explored. Given the limited knowledge of β-glucan on anti-fibrotic activity, bile acid metabolism, and gut microbiota function, additional relevant research is highly encouraged to lay a solid foundation for the use of food-derived β-glucan as a functional food for NAFLD. It is envisaged that further investigation of food-derived β-glucan in human clinical studies should be carried out for its wider utilization.
Collapse
Affiliation(s)
- Nelson Kei
- Food and Nutritional Sciences Program, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; (N.K.); (S.L.)
| | - Vincent Wai Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China;
| | - Susana Lauw
- Food and Nutritional Sciences Program, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; (N.K.); (S.L.)
| | - Lijun You
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China;
| | - Peter Chi Keung Cheung
- Food and Nutritional Sciences Program, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; (N.K.); (S.L.)
| |
Collapse
|
50
|
Revel-Muroz A, Akulinin M, Shilova P, Tyakht A, Klimenko N. Stability of human gut microbiome: Comparison of ecological modelling and observational approaches. Comput Struct Biotechnol J 2023; 21:4456-4468. [PMID: 37745638 PMCID: PMC10511340 DOI: 10.1016/j.csbj.2023.08.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/27/2023] [Accepted: 08/27/2023] [Indexed: 09/26/2023] Open
Abstract
The gut microbiome plays a pivotal role in the human body, and perturbations in its composition have been linked to various disorders. Stability is an essential property of a healthy human gut microbiome, which allows it to maintain its functional richness under the external influences. This property has been explored through two distinct methodologies - mathematical modelling based on ecological principles and statistical analysis drawn from observations in interventional studies. Here we conducted a meta-analysis aimed to compare the two approaches utilising the data from 9 interventional and time series studies encompassing 3512 gut microbiome profiles obtained via 16S rRNA gene sequencing. By employing the previously published compositional Lotka-Volterra method, we modelled the dynamics of the microbial community and evaluated ecological stability measures. These measures were compared to those based on observed microbiome changes. There was a substantial correlation between the outcomes of the two approaches. Particularly, local stability assessed within the ecological paradigm was positively correlated with observational stability measures accounting for the compositional nature of microbiome data. Additionally, we were able to reproduce the previously reported inverse relationship between the community's robustness to microorganism loss and local stability, attributed to the distinct impacts of coefficient characterising the network decomposition on these two stability assessments. Our findings demonstrate harmonisation between the ecological and observational approaches to microbiome analysis, advancing the understanding of healthy gut microbiome concept. This paves the way to develop efficient microbiome-targeting interventions for disease prevention and treatment.
Collapse
Affiliation(s)
- Anastasia Revel-Muroz
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Mikhail Akulinin
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Institutskiy per. 9, Dolgoprudny, Moscow Region, Russia
| | - Polina Shilova
- Department of Biology, Moscow State University, 1–12 Leninskie Gory, Moscow, Russia
| | - Alexander Tyakht
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Atlas Biomed Group - Knomx LLC, Interchange House, Office 1.58, 81–85 Station Road, Croydon CR0 2AJ, United Kingdom
| | - Natalia Klimenko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Atlas Biomed Group - Knomx LLC, Interchange House, Office 1.58, 81–85 Station Road, Croydon CR0 2AJ, United Kingdom
| |
Collapse
|