1
|
Xu QY, Ren TY, Zhou YC, Xu J, Du LD, Hong DY, Zhang QR, Chu HK, Peng Z, Fan JG, Jiang L. Prevotella copri-produced 5-aminopentanoic acid promotes pediatric metabolic dysfunction-associated steatotic liver disease. Hepatobiliary Pancreat Dis Int 2025; 24:303-315. [PMID: 40057459 DOI: 10.1016/j.hbpd.2025.02.004] [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: 10/18/2024] [Accepted: 02/24/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Recent studies suggest an association between the expansion of Prevotella copri and the disease severity in children with metabolic dysfunction-associated steatotic liver disease (MASLD). We aimed to investigate the causative role and molecular mechanisms of P. copri in pediatric MASLD. METHODS C57BL/6 J mice aged 3 weeks were fed a high-fat diet (HFD) and orally administered with P. copri for 5 weeks. We assessed the key features of MASLD and the gut microbiota profile. By untargeted metabolomics on mouse fecal samples and the supernatant from P. copri culture, we identified P. copri-derived metabolite and tested its effects in vitro. RESULTS In HFD-fed mice, administration of P. copri significantly promoted liver steatosis. Genes associated with inflammation and fibrosis were significantly upregulated in the livers from the HFD + P. copri group compared with those in the livers from the HFD group. In addition, P. copri reduced gut microbial diversity, increased the proportion of Firmicutes and decreased Bacteroidota. Importantly, 5-aminopentanoic acid (5-AVA) was significantly enriched in both mouse feces from the HFD + P. copri group and the culture supernatant of P. copri. In vitro, 5-AVA aggravated palmitic acid-induced lipid accumulation in HepG2 cells and primary mouse hepatocytes. Mechanistically, P. copri-produced 5-AVA exacerbated hepatic steatosis by promoting lipogenesis and fatty acid uptake, while also reducing hepatic very-low-density lipoprotein export. CONCLUSIONS Our findings demonstrated that P. copri promotes liver steatosis in HFD-fed juvenile mice through its metabolite 5-AVA, suggesting its potential as a therapeutic target for the management of pediatric MASLD.
Collapse
Affiliation(s)
- Qing-Yang Xu
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Tian-Yi Ren
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yong-Chang Zhou
- Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| | - Juan Xu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Lan-Duoduo Du
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Dong-Yang Hong
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qian-Ren Zhang
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Hui-Kuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhong Peng
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| | - Lu Jiang
- Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
2
|
Wang K, Wang H, Sun Q, Kou X, Yang P, Zhai X, Li Y, Yang S, Wang X. Polysaccharides from Cistanche deserticola Ma prevent alcoholic fatty liver disease by regulating hepatic lipid metabolism and gut microbiota in mice. Int Immunopharmacol 2025; 156:114707. [PMID: 40288152 DOI: 10.1016/j.intimp.2025.114707] [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: 12/10/2024] [Revised: 04/08/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
Cistanche deserticola Ma polysaccharide (CDP) is an active ingredient extracted from Cistanche deserticola Ma (CD) and is believed to have hepatoprotective activity. However, the molecular mechanism of its hepatoprotective effect by intervening in alcoholic fatty liver disease (AFLD) remains unclear. In the present study, three polysaccharides were isolated and purified from CD, and the polysaccharide with the better lipid-lowering and liver-protecting activity and the highest yield, CDPS, was selected by in vitro experiments for subsequent. The efficacy of CDPS in ameliorating AFLD in mice was evaluated using hepatic lipidomics, 16S rRNA analysis and molecular biology experiments. The present study showed that CDPS significantly improved alcohol-induced weight loss, lipid accumulation, ALT, AST, inflammation and dyslipidemia, suggesting that CDPS can prevent AFLD. Lipidomics showed that CDPS improved lipid metabolism disorders by regulating glycerophospholipid metabolism, linoleic acid metabolism, and arachidonic acid metabolism. 16S rRNA analysis showed that CDPS ameliorated alcohol-induced gut microbiota disruption. In particular, CDPS supplementation reduced the abundance of Bacteroides, Parabacteroides, and Escherichia-Shigella and increased the abundance of Ruminococcaceae_UCG-010, Lachnospiraceae_NK4A136_group and Faecalibaculum abundance, and promoted the production of SCFAs. We further used Western blotting to determine the levels of proteins involved in lipogenesis and catabolism, and CDPS intervention resulted in decreased levels of lipid synthesis-associated proteins (SREBP-1c and FAS) and increased levels of lipid catabolism-associated proteins (PPARα) and p-AMPK/AMPK ratio. Our results suggest that CDPS has the potential to prevent AFLD by modulating lipid metabolism, altering the gut microbiota, increasing the content of SCFAs, activating the AMPK signaling pathway to promote fatty acid β-oxidation and limiting fatty acid biosynthesis, which provides a basis for further development of therapeutic drugs for AFLD.
Collapse
Affiliation(s)
- Kai Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Haichao Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qihui Sun
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xuefang Kou
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Pei Yang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xinyuan Zhai
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yaying Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Shangshen Yang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xiaoming Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Shandong Province Cardiovascular Disease TCM Precision Treatment Engineering Research Centre, Jinan 250355, China.
| |
Collapse
|
3
|
Bhattacharya I, Maity DK, Kumar A, Sarkar S, Bhattacharya T, Sahu A, Sreedhar R, Arumugam S. Beyond obesity: lean metabolic dysfunction-associated steatohepatitis from unveiling molecular pathogenesis to therapeutic advancement. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04257-x. [PMID: 40366398 DOI: 10.1007/s00210-025-04257-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD), now known by the name of metabolic dysfunction-associated fatty liver disease (MAFLD), with increased global incidence, has been recognized as a significant metabolic disorder. NAFLD includes a spectrum liver disease from hepatocellular fat accumulation (isolated steatosis) to an advanced form of liver injury known as nonalcoholic steatohepatitis (NASH), which refers to distinct histologic features, including hepatocellular steatosis and injury, necroinflammation, and eventually fibrosis. Nonobese or lean individuals associated with metabolic dysregulation usually demonstrated diverse risk factors compared to obese MAFLD. The presence of normal range body mass index (BMI) and excess visceral adiposity with increased cardiometabolic and renal comorbidities, along with sarcopenia, has been evidenced to be associated with lean MASH. Genetic predispositions accompanying lifestyle and environmental factors contribute to disease initiation and progression. The genetic influence in pathophysiology indicated the significant contributions of the following genes: PNPLA3, TM6SF2, APOB, LIPA, MBOAT7, and HSD17B13, and the impact of their disease-specific variants in the development of obesity-independent MASH. The epigenetic modifications exhibited differential DNA methylation patterns in the genes involved in lipid metabolism, particularly hypomethylation of PEMT. Diet-induced and genetic animal models of lean MASH, including Slc: Wistar/ST rats, PPAR-α, PTEN, and MAT1A knockout mice models, are indicated to be pivotal in the exploration of disease progression and observing the effect of therapeutic interventions. This comprehensive review comprises the molecular and genetic pathophysiology, molecular diagnostics, and therapeutic aspects of lean MASH to enunciate a diagnostic approach that combines detailed clinical phenotyping regarding genomic analysis.
Collapse
Affiliation(s)
- Indrajit Bhattacharya
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Deep Kumar Maity
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Amit Kumar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Sampriti Sarkar
- School of Biosciences & Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632014, India
| | - Teeshyo Bhattacharya
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Amrita Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Remya Sreedhar
- School of Pharmacy, Sister Nivedita University, DG Block, Action Area I, 1/2, Newtown, Kolkata, 700156, West Bengal, India
| | - Somasundaram Arumugam
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| |
Collapse
|
4
|
Yu H, Guo J, Li B, Ma J, Abebe BK, Mei C, Raza SHA, Cheng G, Zan L. Erucic acid promotes intramuscular fat deposition through the PPARγ-FABP4/CD36 pathway. Int J Biol Macromol 2025; 298:140121. [PMID: 39837435 DOI: 10.1016/j.ijbiomac.2025.140121] [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: 08/15/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 01/23/2025]
Abstract
The regulation of intramuscular fat (IMF) accumulation plays a crucial role in determining meat quality in the beef industry. In humans, fat deposition in skeletal muscle is closely associated with insulin resistance and obesity. However, its underlying mechanisms are not fully elucidated. We previously identified erucic acid (EA) as a key metabolite that may affect IMF deposition of beef using omics strategies. By utilizing bovine intramuscular preadipocytes in vitro, the study demonstrates a dose-dependent increase in lipid storage induced by EA, along with mRNA expression levels of transporters FABP4 and CD36. At a mechanistic level, EA triggers ERK1/2 phosphorylation and enhances the expression of PPARγ, FABP4, and CD36, thereby facilitating the formation of lipid droplets within preadipocytes. In vivo experiments conducted in mice support these findings, indicating that EA stimulates fat accumulation in skeletal muscles and enhances the levels of FABP4 and CD36 proteins. These outcomes not only enhance our comprehension of the molecular mechanisms governing IMF deposition but also provide insights into potential strategies for enhancing meat quality and addressing metabolic disorders linked to fat accumulation in skeletal muscles. The findings of the study contribute to existing scholarly knowledge and lay the groundwork for future research endeavors aimed at improving meat quality and metabolic well-being.
Collapse
Affiliation(s)
- Hengwei Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Juntao Guo
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Bingzhi Li
- Key Laboratory for Efficient Ruminant Breeding Technology of Higher Education Institutions in Shaanxi Provinc, Yangling vocational & technical college, Yangling 712100, China
| | - Jing Ma
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Belete Kuraz Abebe
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Chugang Mei
- College of Grassland Agriculture, Northwest A&F University, Yangling 712100, China; National Beef Cattle Improvement Center, Yangling 712100, China
| | - Sayed Haidar Abbas Raza
- Guangdong Provincial Key Laboratory of Food Quality and Safety / Nation-Local Joint Engineering Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou 510642, China.
| | - Gong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; National Beef Cattle Improvement Center, Yangling 712100, China.
| |
Collapse
|
5
|
Li X, Han X, Yan H, Zhu H, Wang H, Li D, Tian Y, Su Y. From gut microbiota to host genes: A dual-regulatory pathway driving body weight variation in dagu chicken (Gallus gallus domesticus). Poult Sci 2025; 104:105067. [PMID: 40239312 PMCID: PMC12032334 DOI: 10.1016/j.psj.2025.105067] [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: 01/12/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
During the growth and development of animals, there is an interaction between the gut microbiota and the host genotype. The host genotype can regulate the microbiota, and in turn, the microbiota can influence host gene expression, thereby affecting the animal's production performance. This study explored the dynamic interplay between the gut microbiota and host gene expression in body weight variation in Dagu chicken, an indigenous poultry genetic resource in China. We characterized mucosa-associated microbiota across four gastrointestinal segments (duodenum, jejunum, ileum, cecum) and ileocecal chyme microbiota in 12-week-old Dagu chickens stratified by divergent body weight phenotypes, while simultaneously quantifying region-specific intestinal epithelial transcriptional regulation. 16S rDNA sequencing was employed to identify Firmicutes as the predominant bacterial phylum, with notable differences in the abundance of specific genera (e.g., Ligilactobacillus and Lactobacillus) being observed between the high- or low-body-weight groups. Enhanced biosynthesis pathways were functionally predicted in heavier roosters, whereas reduced nutrient metabolism pathways were contrasted. A conserved functional concordance was observed between regionally predominant differential microbiota and the physiological specialization of corresponding intestinal niches. Functional analysis revealed that the high-body-weight group demonstrated superior capabilities in microbial biosynthesis, whereas the low-body-weight group exhibited enhanced microbial metabolic activity. NAA80 was identified as the common differentially expressed gene across all intestinal epithelial tissues. The Gene Ontology and KEGG pathway analyses revealed elevated nutrient absorption efficiency in the high-body-weight group, while the low-body-weight group demonstrated accelerated cellular renewal rates and shorter cycles. Correlation analysis identified significant associations between gut microbiota and host genes expression profiles, with the majority of correlations being positive. These results suggest a coordinated interaction between microbial communities and host genetic regulation, potentially driving phenotypic differences in body weight performance.
Collapse
Affiliation(s)
- Xiaohan Li
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Xueru Han
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Huan Yan
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Hongyan Zhu
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Hongcai Wang
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Desheng Li
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Yumin Tian
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Yuhong Su
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China.
| |
Collapse
|
6
|
Pei S, Yang L, Gao H, Liu Y, Lu J, Dai EH, Meng C, Feng F, Wang Y. The association between the gut microbiome and antituberculosis drug-induced liver injury. Front Pharmacol 2025; 16:1512815. [PMID: 40129950 PMCID: PMC11931021 DOI: 10.3389/fphar.2025.1512815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/10/2025] [Indexed: 03/26/2025] Open
Abstract
Background This study aimed to explore the distinct characteristics of the gut microbiota in tuberculosis (TB) patients who experienced liver injury following anti-TB treatment compared with those who did not. Method We employed a nested case-control study design, recruiting newly diagnosed pulmonary TB patients at Tangshan Infectious Disease Hospital. Participants were categorized into the Antituberculosis Drug-Induced Liver Injury (ADLI) group and the Non-ADLI group based on the occurrence of liver injury after treatment. Both groups received identical anti-TB regimens. Stool samples were collected from patients who developed liver injury within 2-3 weeks of starting treatment, alongside matched controls during the same timeframe. The samples underwent 16S rDNA sequencing, and clinical data and blood samples were also collected for further analysis. At the same time, we constructed mouse models to explore the effects of different anti-tuberculosis drugs on gut microbiota. Results Following anti-TB treatment, we observed a decrease in microbial diversity and significant structural changes in the gut microbiota of TB patients (P < 0.05). At T1, the Non_ADLI_T1 group presented relatively high levels of Phascolarctobacterium, Anaerofustis and Mailhella. In contrast, the ADLI_ T1 group presented elevated levels of Bacteroides, Veillonella, Clavibacter, Corynebacterium, Anaerococcus, Gardnerella, Peptostreptococcus and Lautropia. At T2, the ADLI_T2 group presented increased levels of Enterococcus, Faecalibacterium, unclassified_f__Burkholderiaceae, Cardiobacterium, Ruminococcus_gnavus_group and Tyzzerella_4 than did the Non_ADLI_T2 group. Additionally, the ADLI_T2 group presented decreased levels of Prevotella_9, Akkermansia, Erysipelotrichaceae_UCG-003, Rubrobacter and norank_f__Desulfovibrionaceae than did the Non_ADLI_T2 group. In animal experiments, similar changes to those in the human population were observed in the mouse model compared to the control group. Any single anti-tuberculosis drug or two-drug combination or three-drug combination can cause dysbiosis of the mouse gut microbiota. The signature genera between groups are different and related to the type of anti-tuberculosis drug. Conclusion Anti-tuberculosis treatment induces dysbiosis in the gut microbiota of TB patients. Notably, there are significant differences in microbiota characteristics between TB patients with and without liver injury at both onset and during treatment. There are some differences in the characteristics of bacterial flora in liver injury caused by different drugs.
Collapse
Affiliation(s)
- Shengfei Pei
- Hebei Coordinated Innovation Center of Occupational Health and Safety, School of Public Health, North China University of Science of Technology, Tangshan, China
| | - Li Yang
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital ofShijiazhuang, Shijiazhuang, China
| | - Huixia Gao
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital ofShijiazhuang, Shijiazhuang, China
| | - Yuzhen Liu
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital ofShijiazhuang, Shijiazhuang, China
| | - Jianhua Lu
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital ofShijiazhuang, Shijiazhuang, China
| | - Er hei Dai
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital ofShijiazhuang, Shijiazhuang, China
| | - Chunyan Meng
- Hebei Coordinated Innovation Center of Occupational Health and Safety, School of Public Health, North China University of Science of Technology, Tangshan, China
| | - Fumin Feng
- Hebei Coordinated Innovation Center of Occupational Health and Safety, School of Public Health, North China University of Science of Technology, Tangshan, China
| | - Yuling Wang
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital ofShijiazhuang, Shijiazhuang, China
| |
Collapse
|
7
|
Zhu R, Xu C, Jiang S, Xia J, Wu B, Zhang S, Zhou J, Liu H, Li H, Lou J. Risk factor analysis and predictive model construction of lean MAFLD: a cross-sectional study of a health check-up population in China. Eur J Med Res 2025; 30:137. [PMID: 40001266 PMCID: PMC11863909 DOI: 10.1186/s40001-025-02373-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
AIM Cardiovascular disease morbidity and mortality rates are high in patients with metabolic dysfunction-associated fatty liver disease (MAFLD). The objective of this study was to analyze the risk factors and differences between lean MAFLD and overweight MAFLD, and establish and validate a nomogram model for predicting lean MAFLD. METHODS This retrospective cross-sectional study included 4363 participants who underwent annual health checkup at Yuyao from 2019 to 2022. The study population was stratified into three groups: non-MAFLD, lean MAFLD (defined as the presence of fatty liver changes as determined by ultrasound in individuals with a BMI < 25 kg/m2), and overweight MAFLD (BMI ≥ 25.0 kg/m2). Subsequent modeling analysis was conducted in a population that included healthy subjects with < 25 kg/m2 (n = 2104) and subjects with lean MAFLD (n = 849). The study population was randomly split (7:3 ratio) to a training vs. a validation cohort. Risk factors for lean MAFLD was identify by multivariate regression of the training cohort, and used to construct a nomogram to estimate the probability of lean MAFLD. Model performance was examined using the receiver operating characteristic (ROC) curve analysis and k-fold cross-validation (k = 5). Decision curve analysis (DCA) was applied to evaluate the clinical usefulness of the prediction model. RESULTS The multivariate regression analysis indicated that the triglycerides and glucose index (TyG) was the most significant risk factor for lean MAFLD (OR: 4.03, 95% CI 2.806-5.786). The restricted cubic spline curves (RCS) regression model demonstrated that the relationships between systolic pressure (SBP), alanine aminotransferase (ALT), serum urate (UA), total cholesterol (TCHO), triglyceride (TG), triglyceride glucose (TyG) index, high density lipoprotein cholesterol (HDLC), and MAFLD were nonlinear and the cutoff values for lean MAFLD and overweight MAFLD were different. The nomogram was constructed based on seven predictors: glycosylated hemoglobin A1c (HbA1c), serum ferritin (SF), ALT, UA, BMI, TyG index, and age. In the validation cohort, the area under the ROC curve was 0.866 (95% CI 0.842-0.891), with 83.8% sensitivity and 76.6% specificity at the optimal cutoff. The PPV and NPV was 63.3% and 90.8%, respectively. Furthermore, we used fivefold cross-validation and the average area under the ROC curve was 0.866 (Figure S3). The calibration curves for the model's predictions and the actual outcomes were in good agreement. The DCA findings demonstrated that the nomogram model was clinically useful throughout a broad threshold probability range. CONCLUSIONS Lean and overweight MAFLD exhibit distinct metabolic profiles. The nomogram model developed in this study is designed to assist clinicians in the early identification of high-risk individuals with lean MAFLD, including those with a normal BMI but at metabolic risk, as well as those with abnormal blood lipid, glucose, uric acid or transaminase levels. In addition, this model enhances screening efforts in communities and medical screening centers, ultimately ensuring more timely and effective medical services for patients.
Collapse
Affiliation(s)
- Ruya Zhu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Caicai Xu
- Chronic Liver Disease Center, The Affiliated Yangming Hospital of Ningbo University, Zhejiang, 315400, China
| | - Suwen Jiang
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jianping Xia
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Boming Wu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Sijia Zhang
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jing Zhou
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Hongliang Liu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Hongshan Li
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China.
| | - Jianjun Lou
- Chronic Liver Disease Center, The Affiliated Yangming Hospital of Ningbo University, Zhejiang, 315400, China.
| |
Collapse
|
8
|
Wang J, Huan C, Lyu Q, Tian X, Liu Y, Ji G, Yan Z. Efficacy of composite bacterial deodorant constructed with Camellia sinensis and its in-situ deodorization mechanism on pig manure. WASTE MANAGEMENT (NEW YORK, N.Y.) 2025; 192:69-81. [PMID: 39615288 DOI: 10.1016/j.wasman.2024.11.034] [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: 06/19/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 12/10/2024]
Abstract
Here, we constructed a novel bacterial deodorant (BD) composed of Delftia tsuruhatensis, Paracoccus denitrificans, Pediococcus acidilactici, and Bacillus velezensis. The BD alone removed 64.84 % of NH3, 100 % of H2S, and 63.68 % of comprehensive odor (OU) during a five-day fermentation of pig manure. The effect was enhanced by introducing Camellia sinensis in the composite bacterial deodorant (CBD) treatment, with the removal efficiency (RE) of NH3 and OU being 88.68 % and 88.14 %, respectively. In prolonged trials, maximum RE of NH3, H2S and OU RE reached 90.16 %, 92.32 % and 100 % in CBD group. Bacterial composition of manure revealed that the abundance of odor-producing microbes (Kurthia, Solibacillus, Proteiniphilum and Acholeplasma) and potential pathogens decreased after CBD application. Functional prediction and correlation analyses indicated that the process of nitrification, denitrification and S/N assimilation were facilitated, while S/N mineralization and methanogenesis processes might be inhibited. This deodorant promoted the conversion of malodorous substances into non-odorous forms, establishing an efficient odor removal system in hoggery. Therefore, the bacterial deodorant compounded with C. sinensis has been shown to be an effective method for deodorizing pig farms. This approach will advance the livestock industry toward greener practices and environmental protection, contributing positively to the development of a sustainable agro-ecosystem.
Collapse
Affiliation(s)
- Jialing Wang
- CAS Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenchen Huan
- CAS Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
| | - Qingyang Lyu
- CAS Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
| | - Xueping Tian
- CAS Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
| | - Yang Liu
- CAS Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gaosheng Ji
- CAS Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
| | - Zhiying Yan
- CAS Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
9
|
Lin Z, Li T, Huang F, Wu M, Zhu L, Zhou Y, Ming Y, Lu Z, Peng W, Gao F, Zhu Y. Comparison of diet and exercise on cardiometabolic factors in young adults with overweight/obesity: multiomics analysis and gut microbiota prediction, a randomized controlled trial. MedComm (Beijing) 2025; 6:e70044. [PMID: 39802638 PMCID: PMC11725046 DOI: 10.1002/mco2.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/13/2024] [Accepted: 11/22/2024] [Indexed: 01/16/2025] Open
Abstract
The optimal strategy for improving cardiometabolic factors (CMFs) in young obese individuals through diet and exercise remains unclear, as do the potential mechanisms. We conducted an 8-week randomized controlled trial to compare the effects of different interventions in youth with overweight/obesity. Gut microbes and serum metabolites were examined to identify regulating mechanisms. A total of 129 undergraduates were randomly assigned to fiber-rich (FR) diet, rope-skipping (RS), combined FR-RS and control groups. The results showed that single interventions were as effective as combined interventions in improving weight, waist circumference, body fat, and lipid profile compared with control group. Notably, the FR group further reduced low-density lipoprotein (LDL-C) and uric acid (UA) (all p < 0.05). Mediation analysis revealed four gut microbiota-metabolite-host axes in improving CMFs. Additionally, we used machine learning algorithms to further predict individual responses based on baseline gut microbiota composition, with specific microbial genera guiding targeted intervention selection. In conclusion, FR diet and/or RS were effective in improving CMFs, with the FR diet particular effectiveness in reducing LDL-C and UA levels. These benefits may drive by gut microbiome-metabolite-host interactions. Moreover, the predictability of gut microbiota composition supports making targeted decisions in selecting interventions. Trial Registration: NCT04834687.
Collapse
Affiliation(s)
- Zongyu Lin
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Tianze Li
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Fenglian Huang
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Miao Wu
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Lewei Zhu
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Yueqin Zhou
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Ying‐An Ming
- Department of Physical EducationSun Yat‐sen UniversityGuangzhouChina
| | - Zhijun Lu
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Wei Peng
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Fei Gao
- Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | - Yanna Zhu
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of FoodNutrition and HealthGuangzhouChina
| |
Collapse
|
10
|
Jiang H, Wang H, Guo Y, Zhu Y, Dai H, Liang C, Gao J. Characterization of the hepatic flora and metabolome in nonalcoholic fatty liver disease. Front Microbiol 2024; 15:1528258. [PMID: 39760085 PMCID: PMC11697427 DOI: 10.3389/fmicb.2024.1528258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Background/aim The purpose of this study was to examine the hepatic bacterial composition and metabolome characteristics of patients with NAFLD using 16S rDNA sequencing and metabolomics. The results of the study revealed substantial differences in hepatic bacterial composition and metabolites between the NAFLD group and the control group. These differences were used to identify potential biomarkers that could be employed to diagnose NAFLD. Subjects/methods Liver tissues from 13 patients in the NAFLD group and 12 patients in the control group were collected for microbiota examination. Results The bacterial DNA profiles of the liver were significantly different between NAFLD patients and controls. NAFLD patients exhibited an enrichment of Enterobacterales, Mycobacteriales, Pseudomonadales, Flavobacteriales and Xanthomonadales, Sphingomonadales, Lysobact, which was characterised by a lack of erales. At the genus level, the abundance values of Escherichia-Shigella, Rhodococcus, and Chryseobacterium in the NAFLD group were significantly elevated, while the abundance values of Stenotrophomonas, Lawsonella and Sphingobium were significantly reduced. A total of 402 distinct metabolites were identified between the two groups, with 78 metabolites that were up-regulated and 14 metabolites that were down-regulated. The enrichment of metabolic pathways indicated that linoleic acid metabolism was the most significant contributor to the metabolic differences, and lipid metabolism was substantially differentiated. The hepatic metabolite levels were substantially correlated with the changes in hepatic microflora, as demonstrated by the correlation analysis. Conclusion Differences in pathogenesis and host physiological function of NAFLD may be attributed to the hepatic flora and metabolomic characteristics. In the future, this presents new opportunities for the investigation of prospective diagnostic and therapeutic targets for NAFLD.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Gastroenterology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Hui Wang
- Department of Gastroenterology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Yangfan Guo
- Central Laboratory of Yan’an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Yankun Zhu
- Department of Surgery, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Hui Dai
- Department of Oncology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Chenchen Liang
- Department of Gastroenterology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Jianpeng Gao
- Shenzhen Ruipuxun Academy for Stem Cell & Regenerative Medicine, Shenzhen, China
- Department of Gastroenterology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
11
|
Wei J, Luo J, Yang F, Dai W, Huang Z, Yan Y, Luo M. Comparative genomic and metabolomic analysis reveals the potential of a newly isolated Enterococcus faecium B6 involved in lipogenic effects. Gene 2024; 927:148668. [PMID: 38852695 DOI: 10.1016/j.gene.2024.148668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/01/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Evidence has indicated that Enterococcus plays a vital role in non-alcoholic fatty liver disease (NAFLD) development. However, the microbial genetic basis and metabolic potential in the disease are yet unknown. We previously isolated a bacteria Enterococcus faecium B6 (E. faecium B6) from children with NAFLD for the first time. Here, we aim to systematically investigate the potential of strain B6 in lipogenic effects. The lipogenic effects of strain B6 were explored in vitro and in vivo. The genomic and functional characterizations were investigated by whole-genome sequencing and comparative genomic analysis. Moreover, the metabolite profiles were unraveled by an untargeted metabolomic analysis. We demonstrated that strain B6 could effectively induce lipogenic effects in the liver of mice. Strain B6 contained a circular chromosome and two circular plasmids and posed various functions. Compared to the other two probiotic strains of E. faecium, strain B6 exhibited unique functions in pathways of ABC transporters, phosphotransferase system, and amino sugar and nucleotide sugar metabolism. Moreover, strain B6 produced several metabolites, mainly enriched in the protein digestion and absorption pathway. The unique potential of strain B6 in lipogenic effects was probably associated with glycolysis, fatty acid synthesis, and glutamine and choline transport. This study pioneeringly revealed the metabolic characteristics and specific detrimental traits of strain B6. The findings provided new insights into the underlying mechanisms of E. faecium in lipogenic effects, and laid essential foundations for further understanding of E. faecium-related disease.
Collapse
Affiliation(s)
- Jia Wei
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Jiayou Luo
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Fei Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang 421001, Hunan, China
| | - Wen Dai
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Zhihang Huang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Yulin Yan
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Miyang Luo
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China.
| |
Collapse
|
12
|
Yuan L, Liu C, Li B, Wang S, Sun J, Mao X. Multi-omics analysis reveals that agaro-oligosaccharides with different degrees of polymerization alleviate colitis in mice by regulating intestinal flora and arginine synthesis. Food Funct 2024; 15:10628-10643. [PMID: 39310981 DOI: 10.1039/d4fo03650f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Inflammatory bowel disease (IBD) is a common chronic disease with a complex etiology, characterized by body weight loss, intestinal barrier damage, and an imbalance of intestinal flora, posing a significant threat to people's health. In this work, we studied whether safer natural active agaro-oligosaccharides (AOSs) benefit mice with IBD and elucidated their underlying mechanisms. The findings indicated that oral administration of agarobiose (A2), agarotriose (A3), and agarotetraose (A4) contributed to alleviating body weight loss and colon shortening, as well as enhancing IL-10 levels while reducing IL-6, IL-1β, and TNF-α. AOSs improved colon disruption, reduced the number of goblet cells caused by DSS, and enhanced the expression of Muc2, ZO-1, and occludin-1 to repair the intestinal barrier. It is noteworthy that A3 demonstrated superior outcomes in the evaluated AOSs relative to A2 and A4. This was evidenced by an increase in Bacteroidota and reduced Firmicutes at the phylum level, which corrected DSS-induced intestinal dysbiosis and significantly restored disrupted metabolic pathways, including amino acid and lipid metabolism. The differential metabolites between the AOS treatment groups and the model group were mainly enriched in arginine synthesis with co-regulated critical substances N-acetyl-L-citrulline and N2-acetylornithine, which alleviated colitis. This evidence offers a fresh perspective on the potential application of AOSs as functional foods to improve intestinal inflammation and metabolism.
Collapse
Affiliation(s)
- Long Yuan
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, PR China.
- Qingdao Key Laboratory of Food Biotechnology, Qingdao, 266404, PR China
| | - Chunhui Liu
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, PR China.
- Qingdao Key Laboratory of Food Biotechnology, Qingdao, 266404, PR China
| | - Bolun Li
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, PR China.
- Qingdao Key Laboratory of Food Biotechnology, Qingdao, 266404, PR China
| | - Sai Wang
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, PR China.
- Qingdao Key Laboratory of Food Biotechnology, Qingdao, 266404, PR China
| | - Jianan Sun
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, PR China.
- Qingdao Key Laboratory of Food Biotechnology, Qingdao, 266404, PR China
- Sanya Ocean Research Institute, Ocean University of China, Sanya, 572025, PR China
| | - Xiangzhao Mao
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, PR China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, PR China
- Qingdao Key Laboratory of Food Biotechnology, Qingdao, 266404, PR China
- Sanya Ocean Research Institute, Ocean University of China, Sanya, 572025, PR China
| |
Collapse
|
13
|
Li J, Shen N, He W, Pan Y, Wu J, Zhao R, Mo X, Li Y. Gut microbiome impact on childhood allergic rhinitis and house dust mite IgE responses. Pediatr Res 2024:10.1038/s41390-024-03645-y. [PMID: 39433961 DOI: 10.1038/s41390-024-03645-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND The correlation between the gut microbiota and airway inflammation in childhood allergic rhinitis (AR), particularly concerning allergen exposure, remains insufficiently explored. This study aimed to link gut microbiota changes with house dust mite (HDM)-specific IgE responses in pediatric AR. METHODS Using metagenomic shotgun sequencing, we compared the fecal microbiota of 60 children with HDM-AR to 48 healthy controls (HC), analyzing the link to IgE reactions. We examined the effects of oral Escherichia (E.) fergusonii treatment in mice sensitized with ovalbumin and HDM on allergic symptoms, mucosal cell infiltration, Th1/Th2/Tregs balance in the spleen, serum cytokine levels, and E. fergusonii presence in feces. RESULTS Children with HDM-AR have a less diverse gut microbiome and lower levels of E. fergusonii compared to controls, with a negative correlation between E. fergusonii abundance and HDM-specific IgE levels. In mice sensitized with OVA and HDM, oral administration of E. fergusonii improved allergic symptoms, reduced nasal eosinophils/mast cells infiltration and adjusted Th cell populations towards a non-allergic profile in splenic lymphocytes with exception of IFN-γ change in serum. CONCLUSION These findings underline the potential of targeting gut microbiota, particularly E. fergusonii, in managing childhood HDM-AR, suggesting a promising approach for future interventions. IMPACT The composition and distribution of gut microbiota in children with HDM-AR are significant changed. The abundance of Escherichia genus is decreased in HDM-AR children. HDM-specific IgE levels are strongly negatively associated with E. fergusonii abundance. Oral administration of E. fergusonii effectively suppresses allergic responses in murine model. These findings offer novel insights into the diagnosis and treatment of HDM-AR, which suggested that E. fergusonii holds promise as a potential therapeutic avenue for managing HDM-AR.
Collapse
Affiliation(s)
- Junyang Li
- Department of Otolaryngology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nan Shen
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Infectious Diseases, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjun He
- Department of Otolaryngology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Pan
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Wu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruike Zhao
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xi Mo
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Youjin Li
- Department of Otolaryngology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Department of Otolaryngology, Hainan Branch, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China.
| |
Collapse
|
14
|
Wang L, Wang W, Jiang G, Ke Z, Luo R, Tian W. Autophagy Improves Inflammatory Response in Sepsis Accompanied by Changes in Gut Microbiota. Mediators Inflamm 2024; 2024:9550301. [PMID: 39465181 PMCID: PMC11511597 DOI: 10.1155/2024/9550301] [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: 08/30/2023] [Revised: 07/25/2024] [Accepted: 09/13/2024] [Indexed: 10/29/2024] Open
Abstract
Background: Sepsis is defined as a life-threatening disease. Autophagy and the microbiome are increasingly connected with sepsis. The aim of this study was to investigate the protective effect of autophagy and the possible mechanisms. Methods: The septic rat model was established by cecal ligation perforation (CLP). Rapamycin (Rap), 3-methyladenine (3-MA), and chloroquine (CQ) were administered to interfere autophagy. Western blot (WB) was used to detect the expression of key proteins in autophagy. Hematoxylin and eosin (H&E) staining and enzyme-linked immunosorbent assays (ELISAs) were used to identify the effect of autophagy on various organs. 16S ribosomal RNA gene sequencing was used to analyze the changes of the gut microbiota. Results: Rap significantly upregulated the expression of key autophagy proteins, and 3-MA reduced the relative expression compared to the CLP group. The autophagic flux showed a corresponding trend. Interestingly, the autophagy inducer significantly decreased the mortality and the lipopolysaccharide (LPS) level in serum compared with the CLP group. Autophagy activation significantly improves the inflammatory response in sepsis. Histopathological sections showed that CLP destroyed the tight junctions between ileal epithelial cells, while autophagy induction reversed the damage. The sequencing results showed that autophagy activation increased the alpha diversity and alterted the composition and structure of gut microbiota. The abundance of Proteobacteria was markedly decreased in the Rap group, whereas Bacteroidetes was notably increased compared with the CLP group. Additionally, the protective effect of autophagy further changed the biomarkers in the microbial community. The top 35 functions in each sample were analyzed to obtain 18 genes including RNA synthesis, ATP binding and transport, chromosome assignment, osmotic polysaccharide transport, transcytosis, and methylation. Conclusion: Autophagy is able to improve inflammation and may directly or indirectly regulate the microbiota of septic rats. Autophagy may be an important target for future clinical interventions in the treatment of sepsis.
Collapse
Affiliation(s)
- La Wang
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - WenJia Wang
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | | | - ZunLi Ke
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - RuiXi Luo
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - WeiYi Tian
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
15
|
Su J, Dai Y, Wu X, Zhou X, Fang X, Ge X, Zhao L. Maslinic acid alleviates alcoholic liver injury in mice and regulates intestinal microbiota via the gut-liver axis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:7928-7938. [PMID: 38837352 DOI: 10.1002/jsfa.13624] [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: 12/05/2023] [Revised: 04/01/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Maslinic acid (MA), a pentacyclic triterpene acid, is widely distributed in natural plants and mainly found in the fruit and leaves of olives and hawthorn. MA has been reported as having many health-promoting functions, such as anticancer, anti-inflammation and neuroprotective activities. According to previous study, hawthorn extract has certain hepatoprotective effects. However, the detailed mechanism is still unclear, especially the effect of MA on gut microbiota. RESULTS Our study reveals that MA effectively counteracts alcohol-induced liver injury and oxidative stress. It mitigates alcohol-induced intestinal barrier damage, reverses increased permeability and reduces translocation of lipopolysaccharide (LPS). This prevents LPS/Toll-like receptor 4 activation, leading to decreased TNF-α and IL-1β production. Furthermore, MA rebalances gut microbiota by reversing harmful bacterial abundance and enhancing beneficial bacteria post-alcohol consumption. CONCLUSION MA, through modulation of gut microbiota, alleviates alcohol-induced liver injury via the gut-liver axis. These findings support the potential use of MA as a functional food ingredient for preventing or treating alcoholic liver disease. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jingwen Su
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, China
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
| | - Yuan Dai
- Jiangsu Yanghe Distillery Co. Ltd, Suqian, China
| | - Xianyao Wu
- Jinling High School Hexi Campus International Department, Nanjing, China
| | - Xinhu Zhou
- Jiangsu Yanghe Distillery Co. Ltd, Suqian, China
| | - Xianying Fang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, China
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
- Jinpu Research institute, Nanjing Forestry University, Nanjing, China
| | - Xiangyang Ge
- Jiangsu Yanghe Distillery Co. Ltd, Suqian, China
| | - Linguo Zhao
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, China
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
- Jinpu Research institute, Nanjing Forestry University, Nanjing, China
| |
Collapse
|
16
|
Xu R, Wu J, Pan J, Zhang S, Yang Y, Zhang L, Zhou W, Wu N, Hu D, Ji G, Dang Y. Gan-jiang-ling-zhu decoction improves steatohepatitis by regulating gut microbiota-mediated 12-tridecenoic acid inhibition. Front Pharmacol 2024; 15:1444561. [PMID: 39246653 PMCID: PMC11377346 DOI: 10.3389/fphar.2024.1444561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/02/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction: Gan-jiang-ling-zhu (GJLZ) decoction is a classical traditional Chinese medicine prescription. Through invigorating yang, activating qi and dissipating dampness, GJLZ decoction is widely applied for the treatment of chronic digestive disease, including nonalcoholic fatty liver disease. However, efficacy and mechanism of GJLZ decoction behind nonalcoholic steatohepatitis (NASH) treatment remains unelucidated. Methods: NASH was induced in mice, followed by treatment with GJLZ decoction. Various methods including hematoxylin-eosin, oil red O staining, and triglyceride analysis were employed to evaluate the treatment effects of GJLZ decoction on NASH. Gut microbiota, metabolomics, cell viability assays, immunofluorescence and Western blotting were performed to unveil the mechanism behind GJLZ decoction. Results: GJLZ decoction treatment significantly improved hepatic steatosis in mice with NASH. It led to remodeling of gut flora and metabolite structures, including the 12-tridecenoic acid level. 12-Tridecenoic acid aggravated hepatic steatosis by promoting acetyl-coenzyme A carboxylase alpha (ACC) expression and inhibiting carnitine palmitoyltransferase 1A (CPT1A) expression. GJLZ decoction treatment reduced the 12-tridecenoic acid level, inhibited ACC activity and promoted CPT1A expression. Conclusion: Our results demonstrated that 12-tridecenoic acid aggravated hepatic steatosis by affecting the ACC-CPT1A axis and GJLZ decoction treatment effectively reduced the 12-tridecenoic acid level and improved steatosis.
Collapse
Affiliation(s)
- Ruohui Xu
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Traditional Chinese Medicine, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiaxuan Wu
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Jiashu Pan
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Shengan Zhang
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Yunuo Yang
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Li Zhang
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Na Wu
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Hu
- Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Yanqi Dang
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| |
Collapse
|
17
|
Pan Z, Khatry MA, Yu ML, Choudhury A, Sebastiani G, Alqahtani SA, Eslam M. MAFLD: an ideal framework for understanding disease phenotype in individuals of normal weight. Ther Adv Endocrinol Metab 2024; 15:20420188241252543. [PMID: 38808010 PMCID: PMC11131400 DOI: 10.1177/20420188241252543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/10/2024] [Indexed: 05/30/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated fatty liver disease (MAFLD) is significant, impacting almost one-third of the global population. MAFLD constitutes a primary cause of end-stage liver disease, liver cancer and the need for liver transplantation. Moreover, it has a strong association with increased mortality rates due to various extrahepatic complications, notably cardiometabolic diseases. While MAFLD is typically correlated with obesity, not all individuals with obesity develop the disease and a significant percentage of MAFLD occurs in patients without obesity, termed lean MAFLD. The clinical features, progression and underlying physiological mechanisms of patients with lean MAFLD remain inadequately characterized. The present review aims to provide a comprehensive summary of current knowledge on lean MAFLD and offer a perspective on defining MAFLD in individuals with normal weight. Key to this process is the concept of metabolic health and flexibility, which links states of dysmetabolism to the development of lean MAFLD. This perspective offers a more nuanced understanding of MAFLD and its underlying mechanisms and highlights the importance of considering the broader metabolic context in which the disease occurs. It also bridges the knowledge gap and offers insights that can inform clinical practice.
Collapse
Affiliation(s)
- Ziyan Pan
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | - Maryam Al Khatry
- Department of Gastroenterology, Obaidullah Hospital, Emirates Health Services, Ministry of Health, Ras Al Khaimah, United Arab Emirates
| | - Ming-Lung Yu
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ashok Choudhury
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Giada Sebastiani
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, QC, Canada
| | - Saleh A. Alqahtani
- Organ Transplant Center of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD, USA
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, 176 Hawkesbury Road, Westmead 2145, NSW, Australia
| |
Collapse
|
18
|
Mao ZH, Gao ZX, Pan SK, Liu DW, Liu ZS, Wu P. Ferroptosis: a potential bridge linking gut microbiota and chronic kidney disease. Cell Death Discov 2024; 10:234. [PMID: 38750055 PMCID: PMC11096411 DOI: 10.1038/s41420-024-02000-8] [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: 03/16/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Ferroptosis is a novel form of lipid peroxidation-driven, iron-dependent programmed cell death. Various metabolic pathways, including those involved in lipid and iron metabolism, contribute to ferroptosis regulation. The gut microbiota not only supplies nutrients and energy to the host, but also plays a crucial role in immune modulation and metabolic balance. In this review, we explore the metabolic pathways associated with ferroptosis and the impact of the gut microbiota on host metabolism. We subsequently summarize recent studies on the influence and regulation of ferroptosis by the gut microbiota and discuss potential mechanisms through which the gut microbiota affects ferroptosis. Additionally, we conduct a bibliometric analysis of the relationship between the gut microbiota and ferroptosis in the context of chronic kidney disease. This analysis can provide new insights into the current research status and future of ferroptosis and the gut microbiota.
Collapse
Affiliation(s)
- Zi-Hui Mao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
- Henan Province Research Center for Kidney Disease, Zhengzhou, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
| | - Zhong-Xiuzi Gao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
- Henan Province Research Center for Kidney Disease, Zhengzhou, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
| | - Shao-Kang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
- Henan Province Research Center for Kidney Disease, Zhengzhou, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
| | - Dong-Wei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
- Henan Province Research Center for Kidney Disease, Zhengzhou, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
| | - Zhang-Suo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.
- Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, PR China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China.
| | - Peng Wu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.
- Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, PR China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China.
| |
Collapse
|
19
|
Li Z, Ma N, Gong X, Shi W, Meng X, Yan J, Zhao Z, Li J. Effects of herbal dregs supplementation of Salvia miltiorrhiza and Isatidis Radix residues improved production performance and gut microbiota abundance in late-phase laying hens. Front Vet Sci 2024; 11:1381226. [PMID: 38764854 PMCID: PMC11100463 DOI: 10.3389/fvets.2024.1381226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 03/26/2024] [Indexed: 05/21/2024] Open
Abstract
The present study was designed to evaluate the effect of a mixture of Chinese medicinal residues (CMRs) consisting of Salvia miltiorrhiza residues (SMR) and Isatidis Radix residues (IRR) on productive performance, egg quality, serum lipid and hormone levels, liver and blood antioxidant capacity, oviduct inflammation levels, and gut microbiota in the late-laying stage. A total of 288 fifty-four-week-old BaShang long-tailed hens were divided into four groups. The feed trial period was 8 weeks. The control group was fed the basic diet as a CCMR group, supplemented with 3, 4, and 6% for the experimental groups LCMR, MCMR, and HCMR. The egg production rate of the MCMR group was 8.1% higher than that of the CCMR group (p < 0.05). Serum triglyceride (TG) levels of hens of the CMR-supplemented group were significantly decreased than those of the CCMR group (p < 0.05). The group supplemented with different levels of CMR had significantly higher serum HDL-C levels compared with the control group (p < 0.05). Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) levels were remarkably increased for the LCMR and MCMR groups and significantly decreased for the HCMR group compared to CCMR (p < 0.05). Serum and liver glutathione peroxidase (GSH-PX) activities were significantly increased, and malondialdehyde (MDA) levels were significantly decreased in the MCMR group compared to the CCMR group (p < 0.05). The expression levels of tubal inflammatory factor markers (IL-4, IL-1β, TNF-α) in the MCMR and HCMR groups were consistent with the pathological findings of the sections. As for cecal microbiota, supplementation with CMR affected the alpha diversity of the cecum microbiome at the genus level. The Shannon index was significantly higher in the MCMR group than in the CCMR and HCMR groups (p < 0.05). Supplementation with different levels of CMR mainly regulated the ratio of intestinal Firmicutes to Bacteroidetes and the abundance of phyla such as Proteobacteria. In addition, CMR supplementation at different levels in the diet enriched lipid-metabolizing bacteria, such as Bacteroides and Ruminococcus_gnavus_group. Furthermore, according to linear discriminant analysis (LDA) effect size (LEfSe) analysis, the MCMR group showed an increase in the number of short-chain fatty acid-producing bacteria Romboutsia and fiber-degrading specialized bacteria Monoglobus. Therefore, supplementation of appropriate amounts of CMR to the diet of laying hens enhanced reproductive hormone levels, hepatic antioxidant capacity, and lipid metabolism, alleviated the levels of oviductal inflammatory factors, and modulated the abundance structure of bacterial flora to improve the late-laying performance and egg quality. The results of the current study showed that CMR is a beneficial feed supplement for chickens when added in moderation.
Collapse
Affiliation(s)
- Zhaonian Li
- Institute of Animal Husbandry and Veterinary Medicine of Hebei Province, Baoding, China
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Ning Ma
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Xincheng Gong
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Wanyu Shi
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Xianhua Meng
- Hebei General Station of Animal Husbandry, Shijiazhuang, China
| | - Jingjiao Yan
- Animal Husbandry Technology Promotion Institution of Zhangjiakou, Zhangjiakou, China
| | - Zhiqiang Zhao
- Institute of Animal Husbandry and Veterinary Medicine of Hebei Province, Baoding, China
| | - Jiefeng Li
- Institute of Animal Husbandry and Veterinary Medicine of Hebei Province, Baoding, China
| |
Collapse
|
20
|
Zhou X, Hao R, Zhu X, Tan X, Li D. Isoleucine‐restricted diets improve high‐fat diet‐induced nonalcoholic fatty liver disease via regulating insulin resistance and gut microbiota. FOOD FRONTIERS 2024; 5:893-906. [DOI: 10.1002/fft2.379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
AbstractObesity, caused by a long‐term high‐fat diet, is a risk factor for insulin resistance and nonalcoholic fatty liver disease (NAFLD). The gut microbiota plays an important role in NAFLD development by producing lipopolysaccharides (LPS). Isoleucine (ILE), as one of the branched‐chain amino acids (BCAAs), has a negative effect on glucose and lipid metabolism in obese mice. Therefore, we speculated that isoleucine‐restricted diets could prevent high‐fat diet‐induced insulin resistance and NAFLD. For this purpose, 30 C57BL/6J mice received a control check diet (CK), a high‐fat diet (HFD), and a isoleucine‐restricted diet (IR) for 12 weeks, respectively. The current study revealed that IR diets reversed HFD‐induced weight gain, increased fasting glucose levels, and lipid metabolism disorder. Furthermore, IR diets attenuated HFD‐induced hepatic inflammation by regulating the LPS/TLR4/NF‐κB signaling pathway. Moreover, hepatic insulin resistance and gluconeogenesis disorder were significantly improved by IR diets. In addition, IR diets reshaped HFD‐induced gut microbiota imbalance, reflected in decreasing the proportion of Proteobacteria phylum and LPS contents. Taken together, our studies support that restricting isoleucine in high‐fat diets was a novel means of preventing obesity‐induced NAFLD and insulin resistance.
Collapse
Affiliation(s)
- Xing Zhou
- Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, College of Food Science and Engineering Shandong Agricultural University Taian People's Republic of China
| | - Rili Hao
- Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, College of Food Science and Engineering Shandong Agricultural University Taian People's Republic of China
| | - Xiangyang Zhu
- Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, College of Food Science and Engineering Shandong Agricultural University Taian People's Republic of China
| | - Xintong Tan
- Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, College of Food Science and Engineering Shandong Agricultural University Taian People's Republic of China
| | - Dapeng Li
- Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, College of Food Science and Engineering Shandong Agricultural University Taian People's Republic of China
| |
Collapse
|
21
|
Ding S, Cheng Y, Azad MAK, Zhu Q, Huang P, Kong X. Development of small intestinal barrier function and underlying mechanism in Chinese indigenous and Duroc piglets during suckling and weaning periods. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 16:429-442. [PMID: 38406666 PMCID: PMC10885791 DOI: 10.1016/j.aninu.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/18/2023] [Accepted: 09/30/2023] [Indexed: 02/27/2024]
Abstract
This study explored the developmental changes in small intestinal barrier function and the potential regulatory roles of intestinal microbiota and metabolites in different breeds of piglets during suckling and weaning periods. Taoyuan black (TB), Xiangcun black (XB), and Duroc (DR) piglets (10 litters per breed; half male and half female) were selected for sampling to evaluate the intestinal barrier-related indexes and intestinal microbiota and metabolites at 1, 10, 21 (weaned), and 24 (3 d after weaning) d old. The results showed that weaning led to severe shedding of small intestinal microvilli and sparse microvilli arrangement. D-lactate level in the ileum of TB and XB piglets during suckling and weaning periods was lower (P < 0.01) than that of DR piglets, as well as the ileal diamine oxidase level at 1 d old. The expression level of mucin 1 was higher (P < 0.05) in the ileum of TB and XB piglets than that of DR piglets, and it was the highest in the ileum of TB piglets at 21 d old. The expression levels of mucin 2 and mucin 13 were higher (P < 0.10) in TB and XB piglets than those of DR piglets at 21 d old, whereas mucin 2 and mucin 13 in the ileum of TB and XB piglets were higher (P < 0.05) than those of DR piglets at 24 d old. TB and XB piglets had a lower relative abundance of Escherichia_Shigella at 21 and 24 d old, but they had higher Streptococcus at 1 and 24 d old than DR piglets (P < 0.01). Differential metabolites between the three breeds of piglets were mainly related to oxidative phosphorylation, steroid biosynthesis, and bile acid synthesis. Collectively, these findings suggest that different pig breeds present differences in the development of the small intestinal barrier function. Compared with DR piglets, TB and XB piglets had higher intestinal permeability during the suckling period and a stronger intestinal mechanical barrier after weaning. Moreover, intestinal microbiota and metabolites are the key factors for developing small intestinal barrier functions in different breeds of piglets.
Collapse
Affiliation(s)
- Sujuan Ding
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yating Cheng
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Md Abul Kalam Azad
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Zhu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pan Huang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Xiangfeng Kong
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
22
|
Wei J, Luo J, Yang F, Dai W, Pan X, Luo M. Identification of commensal gut bacterial strains with lipogenic effects contributing to NAFLD in children. iScience 2024; 27:108861. [PMID: 38313052 PMCID: PMC10835367 DOI: 10.1016/j.isci.2024.108861] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/07/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
Gut microbiota is known to have a significant impact on nonalcoholic fatty liver disease (NAFLD), particularly in children with obesity. However, the specific functions of microbiota at the strain level in this population have not been fully elucidated. In this study, we successfully isolated and identified several commensal gut bacterial strains that were dominant in children with obesity and NAFLD. Among these, four novel isolates were found to have significant lipogenic effects in vitro. These strains exhibited a potential link to hepatocyte steatosis by regulating the expression of genes involved in lipid metabolism and inflammation. Moreover, a larger cohort analysis confirmed that these identified bacterial strains were enriched in the NAFLD group. The integrated analysis of these strains effectively distinguished NASH from NAFL. These four strains might serve as potential biomarkers in children with NAFLD. These findings provided new insights into the exploration of therapeutic targets for NAFLD.
Collapse
Affiliation(s)
- Jia Wei
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Jiayou Luo
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Fei Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang 421001, Hunan, China
| | - Wen Dai
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Xiongfeng Pan
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| | - Miyang Luo
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, Hunan, China
| |
Collapse
|
23
|
Gao Q, Chen N, Li B, Zu M, Ma Y, Xu H, Zhu Z, Reis RL, Kundu SC, Xiao B. Natural lipid nanoparticles extracted from Morus nigra L. leaves for targeted treatment of hepatocellular carcinoma via the oral route. J Nanobiotechnology 2024; 22:4. [PMID: 38169394 PMCID: PMC10763359 DOI: 10.1186/s12951-023-02286-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
The clinical application of conventional medications for hepatocellular carcinoma treatment has been severely restricted by their adverse effects and unsatisfactory therapeutic effectiveness. Inspired by the concept of 'medicine food homology', we extracted and purified natural exosome-like lipid nanoparticles (LNPs) from black mulberry (Morus nigra L.) leaves. The obtained MLNPs possessed a desirable hydrodynamic particle size (162.1 nm), a uniform size distribution (polydispersity index = 0.025), and a negative surface charge (-26.6 mv). These natural LNPs were rich in glycolipids, functional proteins, and active small molecules (e.g., rutin and quercetin 3-O-glucoside). In vitro experiments revealed that MLNPs were preferentially internalized by liver tumor cell lines via galactose receptor-mediated endocytosis, increased intracellular oxidative stress, and triggered mitochondrial damage, resulting in suppressing the viability, migration, and invasion of these cells. Importantly, in vivo investigations suggested that oral MLNPs entered into the circulatory system mainly through the jejunum and colon, and they exhibited negligible adverse effects and superior anti-liver tumor outcomes through direct tumor killing and intestinal microbiota modulation. These findings collectively demonstrate the potential of MLNPs as a natural, safe, and robust nanomedicine for oral treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Qiang Gao
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing, 400715, China
| | - Nanxi Chen
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing, 400715, China
| | - Baoyi Li
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing, 400715, China
| | - Menghang Zu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing, 400715, China
| | - Ya Ma
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing, 400715, China
| | - Haiting Xu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing, 400715, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Zhenhua Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, AvePark, Braga, Guimarães, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, AvePark, Braga, Guimarães, Portugal
| | - Bo Xiao
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing, 400715, China.
| |
Collapse
|
24
|
Wei J, Luo J, Yang F, Feng X, Zeng M, Dai W, Pan X, Yang Y, Li Y, Duan Y, Xiao X, Ye P, Yao Z, Liu Y, Huang Z, Zhang J, Zhong Y, Xu N, Luo M. Cultivated Enterococcus faecium B6 from children with obesity promotes nonalcoholic fatty liver disease by the bioactive metabolite tyramine. Gut Microbes 2024; 16:2351620. [PMID: 38738766 PMCID: PMC11093035 DOI: 10.1080/19490976.2024.2351620] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/01/2024] [Indexed: 05/14/2024] Open
Abstract
Gut microbiota plays an essential role in nonalcoholic fatty liver disease (NAFLD). However, the contribution of individual bacterial strains and their metabolites to childhood NAFLD pathogenesis remains poorly understood. Herein, the critical bacteria in children with obesity accompanied by NAFLD were identified by microbiome analysis. Bacteria abundant in the NAFLD group were systematically assessed for their lipogenic effects. The underlying mechanisms and microbial-derived metabolites in NAFLD pathogenesis were investigated using multi-omics and LC-MS/MS analysis. The roles of the crucial metabolite in NAFLD were validated in vitro and in vivo as well as in an additional cohort. The results showed that Enterococcus spp. was enriched in children with obesity and NAFLD. The patient-derived Enterococcus faecium B6 (E. faecium B6) significantly contributed to NAFLD symptoms in mice. E. faecium B6 produced a crucial bioactive metabolite, tyramine, which probably activated PPAR-γ, leading to lipid accumulation, inflammation, and fibrosis in the liver. Moreover, these findings were successfully validated in an additional cohort. This pioneering study elucidated the important functions of cultivated E. faecium B6 and its bioactive metabolite (tyramine) in exacerbating NAFLD. These findings advance the comprehensive understanding of NAFLD pathogenesis and provide new insights for the development of microbe/metabolite-based therapeutic strategies.
Collapse
Affiliation(s)
- Jia Wei
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Jiayou Luo
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Fei Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang, Hunan, China
| | - Xiangling Feng
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Ming Zeng
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Wen Dai
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Xiongfeng Pan
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Yue Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang, Hunan, China
| | - Yamei Li
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang, Hunan, China
| | - Yamei Duan
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Xiang Xiao
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Ping Ye
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Zhenzhen Yao
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Yixu Liu
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Zhihang Huang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Jiajia Zhang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Yan Zhong
- Institute of Children Health, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Ningan Xu
- Institute of Children Health, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Miyang Luo
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| |
Collapse
|
25
|
Zhang R, Qiu W, Sun X, Li J, Geng X, Yu S, Liu Y, Huang H, Li M, Fan Z, Li M, Lv G. Gut microbiota dynamics in a 1-year follow-up after adult liver transplantation in Northeast China. Front Physiol 2023; 14:1266635. [PMID: 38187130 PMCID: PMC10766776 DOI: 10.3389/fphys.2023.1266635] [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: 07/25/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Background: Liver transplantation (LTx) is the most effective treatment for end-stage liver diseases. Gut microorganisms influence the host physiology. We aim to profile the dynamics of gut microbiota in the perioperative period and a 1-year follow-up of LTx recipients in Northeast China. Methods: A total of 257 fecal samples were longitudinally collected from 85 LTx patients using anal swabs from pre-LTx to 1-year post-LTx. A total of 48 fecal samples from end-stage liver disease patients without LTx served as the control. 16S rRNA sequencing was used to analyze gut microbiota diversity, bacterial genera, phenotype classification, and metabolic pathways. Results: The diversity of gut microbiota decreased significantly after transplantation, accompanied by a profound change in the microbial structure, which is characterized by increased abundance of facultative anaerobic bacteria dominated by g_Enterococcus and reduced anaerobic bacteria composition. Predicted functional analysis also revealed disturbances in the metabolic pathway of the gut microbiota. After LTx, the diversity of microbiota gradually recovered but to a less preoperative level after 1 year of recovery. Compared with pre-transplantation, the microbiome structure was characterized by an increase in Acidaminococcus and Acidithiobacillus after 1 year of transplantation. Conclusion: LTx and perioperative treatment triggered gut microbial dysbiosis. The gut microbiota was restructured after LTx to near to but significantly differed from that of pre-LTx.
Collapse
Affiliation(s)
- Ruoyan Zhang
- Department of Hepatobiliary and Pancreatic Surgery I, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Qiu
- Department of Hepatobiliary and Pancreatic Surgery I, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaodong Sun
- Department of Hepatobiliary and Pancreatic Surgery I, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jing Li
- Department of Hepatobiliary and Pancreatic Surgery I, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaochen Geng
- Department of Hepatobiliary and Pancreatic Surgery I, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Shichao Yu
- The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Ying Liu
- Department of Hepatobiliary and Pancreatic Surgery I, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Heyu Huang
- Department of Hepatobiliary and Pancreatic Surgery I, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Mingyue Li
- Department of Hepatobiliary and Pancreatic Surgery I, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhongqi Fan
- Department of Hepatobiliary and Pancreatic Surgery I, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Mingqian Li
- Department of Hepatobiliary and Pancreatic Surgery I, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery I, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
26
|
Song W, Wu F, Yan Y, Li Y, Wang Q, Hu X, Li Y. Gut microbiota landscape and potential biomarker identification in female patients with systemic lupus erythematosus using machine learning. Front Cell Infect Microbiol 2023; 13:1289124. [PMID: 38169617 PMCID: PMC10758415 DOI: 10.3389/fcimb.2023.1289124] [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: 09/05/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
Objectives Systemic Lupus Erythematosus (SLE) is a complex autoimmune disease that disproportionately affects women. Early diagnosis and prevention are crucial for women's health, and the gut microbiota has been found to be strongly associated with SLE. This study aimed to identify potential biomarkers for SLE by characterizing the gut microbiota landscape using feature selection and exploring the use of machine learning (ML) algorithms with significantly dysregulated microbiotas (SDMs) for early identification of SLE patients. Additionally, we used the SHapley Additive exPlanations (SHAP) interpretability framework to visualize the impact of SDMs on the risk of developing SLE in females. Methods Stool samples were collected from 54 SLE patients and 55 Negative Controls (NC) for microbiota analysis using 16S rRNA sequencing. Feature selection was performed using Elastic Net and Boruta on species-level taxonomy. Subsequently, four ML algorithms, namely logistic regression (LR), Adaptive Boosting (AdaBoost), Random Forest (RF), and eXtreme gradient boosting (XGBoost), were used to achieve early identification of SLE with SDMs. Finally, the best-performing algorithm was combined with SHAP to explore how SDMs affect the risk of developing SLE in females. Results Both alpha and beta diversity were found to be different in SLE group. Following feature selection, 68 and 21 microbiota were retained in Elastic Net and Boruta, respectively, with 16 microbiota overlapping between the two, i.e., SDMs for SLE. The four ML algorithms with SDMs could effectively identify SLE patients, with XGBoost performing the best, achieving Accuracy, Sensitivity, Specificity, Positive Predictive Value, Negative Predictive Value, and AUC values of 0.844, 0.750, 0.938, 0.923, 0.790, and 0.930, respectively. The SHAP interpretability framework showed a complex non-linear relationship between the relative abundance of SDMs and the risk of SLE, with Escherichia_fergusonii having the largest SHAP value. Conclusions This study revealed dysbiosis in the gut microbiota of female SLE patients. ML classifiers combined with SDMs can facilitate early identification of female patients with SLE, particularly XGBoost. The SHAP interpretability framework provides insight into the impact of SDMs on the risk of SLE and may inform future scientific treatment for SLE.
Collapse
Affiliation(s)
- Wenzhu Song
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Feng Wu
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Yan Yan
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Yaheng Li
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, Shanxiuan, China
| | - Qian Wang
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, Shanxiuan, China
| | - Xueli Hu
- Department of Nephrology, Hejin People’s Hospital, Yuncheng, Shanxi, China
| | - Yafeng Li
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, Shanxiuan, China
- Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
- Academy of Microbial Ecology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
27
|
Zhang Z, Xu M, Wang L, Gu W, Li X, Han Z, Fu X, Wang X, Li X, Su Z. Continuous oral exposure to micro- and nanoplastics induced gut microbiota dysbiosis, intestinal barrier and immune dysfunction in adult mice. ENVIRONMENT INTERNATIONAL 2023; 182:108353. [PMID: 38035535 DOI: 10.1016/j.envint.2023.108353] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/16/2023] [Accepted: 11/24/2023] [Indexed: 12/02/2023]
Abstract
Micro/nanoplastics in the environment can be ingested by organisms and spread throughout the food chain, ultimately posing a threat to human health. However, the risk of continuous oral exposure in mammals remains unresolved. In this study, we utilized a continuous gavage mouse model to investigate the potential intestinal risks associated with oral exposure to polystyrene micro/nanoplastics (PS-MNPs) with environmentally relevant concentrations. The effects of PS-MNPs with different particle sizes on the gut microbiota, intestinal barrier, and intestinal immune function were evaluated. PS-MNPs can accumulate in the intestine after oral exposure and alter the composition of the gut microbiota. Exposure to PS-MNPs significantly reduced the ratio of Firmicutes to Bacteroidetes as well as the number of potentially beneficial bacteria in the gut, while the number of potentially harmful bacteria significantly increased. The short-chain fatty acids metabolized by gut microbiota were significantly changed by PS-MNPs. Exposure to PS-MNPs disrupts the function of the intestinal barrier and leads to inflammation in the intestines. The levels of secretory immunoglobulin A in the intestine and the differentiation of CD4+ and CD8+ T cells in mesenteric lymph nodes were significantly decreased by PS-MNPs. Moreover, the impact of PS-MNPs on mammalian intestinal health is influenced by the exposure duration and particle size, rather than the concentration. It also suggests that nanoplastics may pose more severe environmental risks.
Collapse
Affiliation(s)
- Zhichun Zhang
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingkai Xu
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; Key Laboratory of Superantigen Research of Liao Ning Province, Shenyang 110016, China.
| | - Lei Wang
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin 300350, China
| | - Wu Gu
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Li
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China
| | - Zhiyang Han
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuanhe Fu
- Department of Immunology, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Xiujuan Wang
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China
| | - Xu Li
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China
| | - Zhencheng Su
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; Key Laboratory of Superantigen Research of Liao Ning Province, Shenyang 110016, China
| |
Collapse
|
28
|
Ma L, La X, Zhang B, Xu W, Tian C, Fu Q, Wang M, Wu C, Chen Z, Chang H, Li JA. Total Astragalus saponins can reverse type 2 diabetes mellitus-related intestinal dysbiosis and hepatic insulin resistance in vivo. Front Endocrinol (Lausanne) 2023; 14:1190827. [PMID: 38053727 PMCID: PMC10694298 DOI: 10.3389/fendo.2023.1190827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023] Open
Abstract
Objective Intestinal flora homeostasis in rats with type 2 diabetes mellitus (T2DM) was evaluated to explore the effects of total Astragalus saponins (TAS) on hepatic insulin resistance (IR). Methods Six-week-old male Sprague-Dawley rats were fed high-fat and high-sugar diet for 4 weeks and intraperitoneally injected with streptozotocin to induce T2DM, and they were then randomly divided into control, model, metformin, and TAS groups. Stool, serum, colon, and liver samples were collected after 8 weeks of drug administration for relevant analyses. Results TAS reduced fasting blood glucose, 2-hour postprandial blood glucose, area under the curve of oral glucose tolerance test, glycated serum protein, homeostasis model assessment of insulin resistance, total cholesterol, triglyceride, and low-density lipoprotein cholesterol levels in T2DM rats but increased insulin, C-peptide, and high-density lipoprotein cholesterol levels. Moreover, TAS improved the morphology and structure of liver and colon tissues and improved the composition of the intestinal microbiome and bacterial community structure at different taxonomic levels. In addition, TAS increased the protein expression of hepatic IRS-1, PI3K, PDK1, and p-AKT and decreased the protein expression of p-GSK-3β. Meanwhile, TAS increased the mRNA expression of liver PDK1, PI3K, and GS and decreased the mRNA expression of GSK-3β. Conclusion TAS can ameliorate T2DM-related abnormal glucose and blood lipid metabolism, intestinal dysbiosis, and IR.
Collapse
Affiliation(s)
- Leilei Ma
- School of Public Health, North China University of Science and Technology, Tangshan, China
- He Bei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Xiaojin La
- He Bei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Biwei Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, China
- He Bei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Wenxuan Xu
- He Bei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Chunyu Tian
- He Bei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Qianru Fu
- He Bei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Meng Wang
- He Bei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Chenxi Wu
- He Bei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Zhen Chen
- Oriental Herbs Korlatolt felelossegu tarsasag, Budapest, Hungary
| | - Hong Chang
- He Bei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Ji-an Li
- School of Public Health, North China University of Science and Technology, Tangshan, China
- He Bei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
29
|
Cao X, Wang X, Ren Y, Sun Y, Yang Z, Ge J, Ping W. Lonicera caerulea L. polyphenols improve short-chain fatty acid levels by reshaping the microbial structure of fermented feces in vitro. Front Microbiol 2023; 14:1228700. [PMID: 37965545 PMCID: PMC10641692 DOI: 10.3389/fmicb.2023.1228700] [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: 05/25/2023] [Accepted: 10/04/2023] [Indexed: 11/16/2023] Open
Abstract
Increasing evidence suggests that the pathogenesis of type 2 diabetes mellitus (T2DM) is closely related to the gut microbiota. Polyphenols have been shown to alleviate T2DM, but the effects of L. caerulea L. polyphenols (LPs) on the gut microbiota and metabolites remain elusive. In this study, the inhibitory effects of fermented L. caerulea L. polyphenols (FLPs) and unfermented L. caerulea L. polyphenols (ULPs) on α-amylase and α-glucosidase and the impact of LP on the gut microbiota and metabolites were investigated. Furthermore, the relationship between the two was revealed through correlation analysis. The results showed that ULP and FLP had the highest inhibitory rates against α-amylase and α-glucosidase at 4 mg ml-1, indicating a strong inhibitory ability. In addition, LP plays a regulatory role in the concentration of short-chain fatty acids (SCFAs) and tends to restore them to their normal levels. LP reversed the dysbiosis of the gut microbiota caused by T2DM, as evidenced by an increase in the abundance of bacterial genera such as Lactobacillus, Blautia, and Bacteroides and a decrease in the abundance of bacterial genera such as Escherichia-Shigella and Streptococcus. Similarly, after LP intervention, the relationships among microbial species became more complex and interconnected. In addition, the correlation between the gut microbiota and metabolites was established through correlation analysis. These further findings clarify the mechanism of action of LP against T2DM and provide a new target for T2DM interventions.
Collapse
Affiliation(s)
- Xinbo Cao
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education and Heilongjiang Provincial Key Laboratory of Plant Genetic Engineering and Biological Fermentation Engineering for Cold Region and Key Laboratory of Microbiology, College of Heilongjiang Province and School of Life Sciences, Heilongjiang University, Harbin, China
| | - Xuemeng Wang
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education and Heilongjiang Provincial Key Laboratory of Plant Genetic Engineering and Biological Fermentation Engineering for Cold Region and Key Laboratory of Microbiology, College of Heilongjiang Province and School of Life Sciences, Heilongjiang University, Harbin, China
| | - Yanxin Ren
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education and Heilongjiang Provincial Key Laboratory of Plant Genetic Engineering and Biological Fermentation Engineering for Cold Region and Key Laboratory of Microbiology, College of Heilongjiang Province and School of Life Sciences, Heilongjiang University, Harbin, China
| | - Yangcun Sun
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education and Heilongjiang Provincial Key Laboratory of Plant Genetic Engineering and Biological Fermentation Engineering for Cold Region and Key Laboratory of Microbiology, College of Heilongjiang Province and School of Life Sciences, Heilongjiang University, Harbin, China
| | - Zhichao Yang
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education and Heilongjiang Provincial Key Laboratory of Plant Genetic Engineering and Biological Fermentation Engineering for Cold Region and Key Laboratory of Microbiology, College of Heilongjiang Province and School of Life Sciences, Heilongjiang University, Harbin, China
| | - Jingping Ge
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education and Heilongjiang Provincial Key Laboratory of Plant Genetic Engineering and Biological Fermentation Engineering for Cold Region and Key Laboratory of Microbiology, College of Heilongjiang Province and School of Life Sciences, Heilongjiang University, Harbin, China
- Hebei Key Laboratory of Agroecological Safety, Hebei University of Environmental Engineering, Qinhuangdao, China
| | - Wenxiang Ping
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education and Heilongjiang Provincial Key Laboratory of Plant Genetic Engineering and Biological Fermentation Engineering for Cold Region and Key Laboratory of Microbiology, College of Heilongjiang Province and School of Life Sciences, Heilongjiang University, Harbin, China
- Hebei Key Laboratory of Agroecological Safety, Hebei University of Environmental Engineering, Qinhuangdao, China
| |
Collapse
|
30
|
Jiang L, Fan JG. The role of the gut microbiome in chronic liver diseases: Present insights and future outlook. Hepatobiliary Pancreat Dis Int 2023; 22:441-443. [PMID: 37690926 DOI: 10.1016/j.hbpd.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023]
Affiliation(s)
- Lu Jiang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jian-Gao Fan
- Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
31
|
Xu J, Zhou Y, Cheng S, Zhao Y, Yan J, Wang Y, Cai W, Jiang L. Lactobacillus johnsonii Attenuates Liver Steatosis and Bile Acid Dysregulation in Parenteral Nutrition-Fed Rats. Metabolites 2023; 13:1043. [PMID: 37887368 PMCID: PMC10608838 DOI: 10.3390/metabo13101043] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/24/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
Parenteral nutrition (PN), a vital therapy for patients with intestinal failure, can lead to the development of parenteral nutrition-associated liver disease (PNALD). In this study, we aimed to investigate the role of Lactobacillus johnsonii (L. johnsonii) in a rat model of PNALD. Total parenteral nutrition (TPN)-fed rats were used to assess the role of L. johnsonii in liver steatosis, bile acid metabolism, gut microbiota, and hepatocyte apoptosis. We observed a depletion of L. johnsonii that was negatively correlated with the accumulation of glycochenodeoxycholic acid (GCDCA), a known apoptosis inducer, in rats subjected to TPN. L. johnsonii attenuated TPN-induced liver steatosis by inhibiting fatty acid synthesis and promoting fatty acid oxidation. TPN resulted in a decrease in bile acid synthesis and biliary bile secretion, which were partially restored by L. johnsonii treatment. The gut microbial profile revealed depletion of pathogenic bacteria in L. johnsonii-treated rats. L. johnsonii treatment reduced both hepatic GCDCA levels and hepatocyte apoptosis compared with the TPN group. In vitro, L. johnsonii treatment inhibited GCDCA-induced hepatocyte apoptosis via its bile salt hydrolase (BSH) activity. Our findings suggest that L. johnsonii protects against liver steatosis, bile acid dysregulation, and hepatocyte apoptosis in TPN-fed rats.
Collapse
Affiliation(s)
- Juan Xu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (J.X.); (J.Y.); (Y.W.)
| | - Yongchang Zhou
- Shanghai Institute for Pediatric Research, Shanghai 200092, China;
| | - Siyang Cheng
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (S.C.); (Y.Z.)
| | - Yuling Zhao
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (S.C.); (Y.Z.)
| | - Junkai Yan
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (J.X.); (J.Y.); (Y.W.)
- Shanghai Institute for Pediatric Research, Shanghai 200092, China;
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (J.X.); (J.Y.); (Y.W.)
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| | - Wei Cai
- Shanghai Institute for Pediatric Research, Shanghai 200092, China;
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (S.C.); (Y.Z.)
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| | - Lu Jiang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (J.X.); (J.Y.); (Y.W.)
- Shanghai Institute for Pediatric Research, Shanghai 200092, China;
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| |
Collapse
|
32
|
Wang C, Huang L, Jin S, Hou R, Chen M, Liu Y, Tang W, Li T, Yin Y, He L. d-Aspartate in Low-Protein Diets Improves the Pork Quality by Regulating Energy and Lipid Metabolism via the Gut Microbes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:12417-12430. [PMID: 37578298 DOI: 10.1021/acs.jafc.3c01974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
d-Aspartate is critical in maintaining hormone secretion and reproductive development in mammals. This study investigated the mechanism of different d-aspartate levels (0, 0.005, 0.05, and 0.5% d-aspartate) in low-protein diets on growth performance and meat quality by mediating the gut microbiota alteration in pigs. We found that adding 0.005% d-aspartate to a low-protein diet could dramatically improve the growth performance during the weaned and growing periods. Dietary d-aspartate with different levels markedly increased the back fat, and 0.5% d-aspartate significantly increased the redness in 24 h and reduced the shear force of the longissimus dorsi (LD) muscle. Moreover, d-aspartate treatments decreased the mRNA expression of MyHC II a and MyHC IIx in the LD muscle. The protein expression of MyH1, MyH7, TFAM, FOXO1, CAR, UCP2, and p-AMPK was upregulated by 0.005% d-aspartate. Additionally, the abundance of Alistipes, Akkermansia, and the [Eubacterium]_coprostanoligenes_group in the intestinal chyme of pigs was significantly decreased by d-aspartate treatments at the genus level, which was also accompanied by a significant decrease in acetate content. These differential microorganisms were significantly correlated with meat quality characteristics. These results indicated that d-aspartate in low-protein diets could improve the growth performance and meat quality in pigs by regulating energy and lipid metabolism via the alteration of gut microbiota.
Collapse
Affiliation(s)
- Chenyu Wang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Le Huang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Shunshun Jin
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Ruoxin Hou
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Mingzhe Chen
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yonghui Liu
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Tiejun Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Yulong Yin
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Liuqin He
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| |
Collapse
|
33
|
Lai Y, Deng H, Fang Q, Ma L, Lei H, Guo X, Chen Y, Song C. Water-Insoluble Polysaccharide Extracted from Poria cocos Alleviates Antibiotic-Associated Diarrhea Based on Regulating the Gut Microbiota in Mice. Foods 2023; 12:3080. [PMID: 37628079 PMCID: PMC10453245 DOI: 10.3390/foods12163080] [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: 06/22/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Antibiotics are very effective in treating a variety of bacterial infections, while clinical overuse of antibiotics can lead to diseases such as antibiotic-associated diarrhea. Numerous studies have shown that natural polysaccharides can be used as prebiotics to alleviate antibiotic-associated diarrhea (AAD). Poria cocos is a medicinal and edible mushroom widely used for thousands of years in China, and our former study demonstrated that water-insoluble polysaccharide (PCY) has the potential prebiotic function. Therefore, we simulated the digestion and fermentation of PCY using feces from volunteers, and then administered it to C57BL/6 mice with AAD to study its effects on the gut microbiota and metabolites. The results indicated that PCY effectively alleviated the symptoms of AAD in mice, restored the intestinal barrier function, improved the content of short-chain fatty acids (SCFAs), decreased the level of inflammatory cytokines, and changed the structure of gut microbiota by increasing the relative abundance of norank_f__Muribaculaceae and unclassified_f__Lachnospiraceae, and decreasing that of Escherichia-Shigella, Staphylococcus and Acinetobacter. This study further demonstrated that PCY is an effective functional prebiotic for improving AAD disease, and provided a new avenue and insight for developing PCY as a functional food or prebiotic for alleviating gastrointestinal diseases.
Collapse
Affiliation(s)
- Yong Lai
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| | - Huiling Deng
- Chongqing Academy of Science and Technology, Chongqing 401121, China; (H.D.); (Y.C.)
- Key Laboratory of Condiment Supervision Technology for State Market Regulation, Chongqing Institute for Food and Drug Administration, Chongqing 401121, China
| | - Qi Fang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| | - Linhua Ma
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| | - Hui Lei
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| | - Xiurong Guo
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| | - Ya Chen
- Chongqing Academy of Science and Technology, Chongqing 401121, China; (H.D.); (Y.C.)
| | - Can Song
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (Y.L.); (Q.F.); (L.M.); (H.L.); (X.G.)
| |
Collapse
|
34
|
Aggeletopoulou I, Kalafateli M, Tsounis EP, Triantos C. Epigenetic Regulation in Lean Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2023; 24:12864. [PMID: 37629043 PMCID: PMC10454848 DOI: 10.3390/ijms241612864] [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: 07/31/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), the most prominent cause of chronic liver disease worldwide, is a rapidly growing epidemic. It consists of a wide range of liver diseases, from steatosis to nonalcoholic steatohepatitis, and predisposes patients to liver fibrosis, cirrhosis, and even hepatocellular carcinoma. NAFLD is strongly correlated with obesity; however, it has been extensively reported among lean/nonobese individuals in recent years. Although lean patients demonstrate a lower prevalence of diabetes mellitus, central obesity, dyslipidemia, hypertension, and metabolic syndrome, a percentage of these patients may develop steatohepatitis, advanced liver fibrosis, and cardiovascular disease, and have increased all-cause mortality. The pathophysiological mechanisms of lean NAFLD remain vague. Studies have reported that lean NAFLD demonstrates a close association with environmental factors, genetic predisposition, and epigenetic modifications. In this review, we aim to discuss and summarize the epigenetic mechanisms involved in lean NAFLD and to introduce the interaction between epigenetic patterns and genetic or non genetic factors. Several epigenetic mechanisms have been implicated in the regulation of lean NAFLD. These include DNA methylation, histone modifications, and noncoding-RNA-mediated gene regulation. Epigenetics is an area of special interest in the setting of lean NAFLD as it could provide new insights into the therapeutic options and noninvasive biomarkers that target this under-recognized and challenging disorder.
Collapse
Affiliation(s)
- Ioanna Aggeletopoulou
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (I.A.); (E.P.T.)
| | - Maria Kalafateli
- Department of Gastroenterology, General Hospital of Patras, 26332 Patras, Greece;
| | - Efthymios P. Tsounis
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (I.A.); (E.P.T.)
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (I.A.); (E.P.T.)
| |
Collapse
|
35
|
Shen B, Gu T, Shen Z, Zhou C, Guo Y, Wang J, Li B, Xu X, Li F, Zhang Q, Cai X, Dong H, Lu L. Escherichia coli Promotes Endothelial to Mesenchymal Transformation of Liver Sinusoidal Endothelial Cells and Exacerbates Nonalcoholic Fatty Liver Disease Via Its Flagellin. Cell Mol Gastroenterol Hepatol 2023; 16:857-879. [PMID: 37572735 PMCID: PMC10598062 DOI: 10.1016/j.jcmgh.2023.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND&AIMS: Gut bacteria translocate into the liver through a disrupted gut vascular barrier, which is an early and common event in the development of nonalcoholic fatty liver disease (NAFLD). Liver sinusoidal endothelial cells (LSECs) are directly exposed to translocated gut microbiota in portal vein blood. Escherichia coli, a commensal gut bacterium with flagella, is markedly enriched in the gut microbiota of patients with NAFLD. However, the impact of E coli on NAFLD progression and its underlying mechanisms remains unclear. METHODS The abundance of E coli was analyzed by using 16S ribosomal RNA sequencing in a cohort of patients with NAFLD and healthy controls. The role of E coli was assessed in NAFLD mice after 16 weeks of administration, and the features of NAFLD were evaluated. Endothelial to mesenchymal transition (EndMT) in LSECs induced by E coli was analyzed through Western blotting and immunofluorescence. RESULTS The abundance of gut Enterobacteriaceae increased in NAFLD patients with severe fat deposition and fibrosis. Importantly, translocated E coli in the liver aggravated hepatic steatosis, inflammation, and fibrosis in NAFLD mice. Mechanistically, E coli induced EndMT in LSECs through the TLR5/MYD88/TWIST1 pathway during NAFLD development. The toll-like receptor 5 inhibitor attenuated E coli-induced EndMT in LSECs and liver injury in NAFLD mice. Interestingly, flagellin-deficient E coli promoted less EndMT in LSECs and liver injury in NAFLD mice. CONCLUSIONS E coli promoted the development of NAFLD and promoted EndMT in LSECs through toll-like receptor 5/nuclear factor kappa B-dependent activation of TWIST1 mediated by flagellin. Therapeutic interventions targeting E coli and/or flagellin may represent a promising candidate for NAFLD treatment.
Collapse
Affiliation(s)
- Bo Shen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyi Gu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyang Shen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cui Zhou
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuecheng Guo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjun Wang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Binghang Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xianjun Xu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qidi Zhang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobo Cai
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Dong
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lungen Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
36
|
Wang H, Shen H, Seo W, Hwang S. Experimental models of fatty liver diseases: Status and appraisal. Hepatol Commun 2023; 7:e00200. [PMID: 37378635 DOI: 10.1097/hc9.0000000000000200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
Fatty liver diseases, including alcohol-associated liver disease (ALD) and nonalcoholic fatty liver disease nonalcoholic fatty liver disease (NAFLD), affect a large number of people worldwide and become one of the major causes of end-stage liver disease, such as liver cirrhosis and hepatocellular carcinoma (HCC). Unfortunately, there are currently no approved pharmacological treatments for ALD or NAFLD. This situation highlights the urgent need to explore new intervention targets and discover effective therapeutics for ALD and NAFLD. The lack of properly validated preclinical disease models is a major obstacle to the development of clinical therapies. ALD and NAFLD models have been in the development for decades, but there are still no models that recapitulate the full spectrum of ALD and NAFLD. Throughout this review, we summarize the current in vitro and in vivo models used for research on fatty liver diseases and discuss the advantages and limitations of these models.
Collapse
Affiliation(s)
- Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Haiyuan Shen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Wonhyo Seo
- Laboratory of Hepatotoxicity, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
37
|
Zhang B, Li J, Fu J, Shao L, Yang L, Shi J. Interaction between mucus layer and gut microbiota in non-alcoholic fatty liver disease: Soil and seeds. Chin Med J (Engl) 2023; 136:1390-1400. [PMID: 37200041 PMCID: PMC10278733 DOI: 10.1097/cm9.0000000000002711] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Indexed: 05/19/2023] Open
Abstract
ABSTRACT The intestinal mucus layer is a barrier that separates intestinal contents and epithelial cells, as well as acts as the "mucus layer-soil" for intestinal flora adhesion and colonization. Its structural and functional integrity is crucial to human health. Intestinal mucus is regulated by factors such as diet, living habits, hormones, neurotransmitters, cytokines, and intestinal flora. The mucus layer's thickness, viscosity, porosity, growth rate, and glycosylation status affect the structure of the gut flora colonized on it. The interaction between "mucus layer-soil" and "gut bacteria-seed" is an important factor leading to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Probiotics, prebiotics, fecal microbiota transplantation (FMT), and wash microbial transplantation are efficient methods for managing NAFLD, but their long-term efficacy is poor. FMT is focused on achieving the goal of treating diseases by enhancing the "gut bacteria-seed". However, a lack of effective repair and management of the "mucus layer-soil" may be a reason why "seeds" cannot be well colonized and grow in the host gut, as the thinning and destruction of the "mucus layer-soil" is an early symptom of NAFLD. This review summarizes the existing correlation between intestinal mucus and gut microbiota, as well as the pathogenesis of NAFLD, and proposes a new perspective that "mucus layer-soil" restoration combined with "gut bacteria-seed" FMT may be one of the most effective future strategies for enhancing the long-term efficacy of NAFLD treatment.
Collapse
Affiliation(s)
- Binbin Zhang
- Department of Translational Medicine Platform, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, China
- Department of School of Life Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310053, China
| | - Jie Li
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Jinlong Fu
- Department of School of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Li Shao
- Department of Translational Medicine Platform, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, China
- Department of School of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Luping Yang
- Department of Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Junping Shi
- Department of Translational Medicine Platform, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, China
- Department of Infectious & Hepatology Diseases, Metabolic Disease Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, China
| |
Collapse
|
38
|
Liu T, Zhao M, Zhang Y, Wang Z, Yuan B, Zhao C, Wang M. Integrated microbiota and metabolite profiling analysis of prebiotic characteristics of Phellinus linteus polysaccharide in vitro fermentation. Int J Biol Macromol 2023; 242:124854. [PMID: 37182617 DOI: 10.1016/j.ijbiomac.2023.124854] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/10/2023] [Accepted: 05/09/2023] [Indexed: 05/16/2023]
Abstract
Phellinus linteus polysaccharide (PLP) had received increasing attention due to its multiple biological activities. Herein, the extraction, characterization and in vitro fermentation of PLP were studied to explore its physiochemical properties and the interaction mechanism between the gut microbiota and PLP. The results obtained demonstrated that PLP was mainly composed of 9 monosaccharides, with three gel chromatographic peaks and molecular weights (Mw) of 308.45 kDa, 13.58 kD and 3.33 kDa, respectively. After 48 h fermentation, the Mw, total sugar, reducing sugar, pH and monosaccharides composition were decreased. Furthermore, PLP regulated the composition of gut microbiota, such as promoting the proliferation of beneficial bacteria such as Bacteroides, Prevotella and Butyricimonas, while preventing the growth of pathogenic bacteria such as Escherichia-Shigella, Morganella and Intestinimonas. Gut microbiota metabolites regulated by PLP such as short-chain fatty acids were the main regulators that impact the host health. Bioinformatics analysis indicated that butyrate, bile acid and purine metabolism were the main metabolic pathways of PLP regulating host health, and the Bacteroides was the key genus to regulate these metabolic pathways. In conclusion, our finding suggested that PLP may be used as a prebiotic agent for human health because of its ability to regulate gut microbiota.
Collapse
Affiliation(s)
- Tingting Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Min Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Yumeng Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Zheyong Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Bo Yuan
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Chunjie Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China.
| | - Miao Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China.
| |
Collapse
|
39
|
Xiao Q, Huang W, Wu Q, Xu H, Zhang Y, Yang J, Bian S, Tan H, Nie S. The effects of pectin on the gut microbiota and serum metabolites in mice fed with a high fat diet and exposed to low-dose antibiotics. Food Funct 2023; 14:4752-4762. [PMID: 37114890 DOI: 10.1039/d2fo03966d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
A sedentary lifestyle, unhealthy diet, and antibiotic use among other environmental factors have been associated with an increased incidence of metabolic disorders and inflammation, as well as gut dysbiosis. Pectin is an edible polysaccharide that exists widely in the cell wall of plants. Our previous study has shown that pectin with various degrees of esterification displayed different effects on preventing acute colitis and regulating the gut microbiome and serum metabolome. This study aimed to further explore the differential effects of pectin with various degrees of esterification on mice simultaneously treated with a high-fat diet and low-dose antibiotics. The results showed that low-esterified pectin L102 improved the biomarkers of metabolic disorders including blood glucose and body weight. The high-esterified pectin H121 and the low-esterified pectin L13 ameliorated inflammatory markers such as superoxide dismutase (SOD). The enrichment of probiotic bacteria such as Lactobacillus by pectin L102, reduction of conditional pathogens such as Klebsiella by pectin L13, and changes in circulating metabolites like L-tryptophan and 3-indoleacrylate by all three types of pectins were detected. These data provide evidence for a differential effect of different types of pectin on the gut microbiota and metabolic health.
Collapse
Affiliation(s)
- Qianhuang Xiao
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| | - Wenqi Huang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| | - Quanyong Wu
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| | - Hedi Xu
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| | - Yanli Zhang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| | - Jingrui Yang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| | - Shuigen Bian
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| | - Huizi Tan
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
40
|
Hu S, Lin Z, Zhao S, Zhang B, Luo L, Zeng L. Pu-erh tea alleviated colitis-mediated brain dysfunction by promoting butyric acid production. Food Chem Toxicol 2023; 172:113594. [PMID: 36592713 DOI: 10.1016/j.fct.2022.113594] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/18/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022]
Abstract
Brain inflammation develops with increased colitis. Pu-erh tea is considered a potential dietary intervention to improve colitis. However, it's unclear whether Pu-erh tea helps alleviate colitis-mediated brain dysfunction. Here, we found that colitis triggered brain dysfunction and increased the risk of depression. Pu-erh tea improved gut-brain barrier function (increased ZO-1 and Occludin) and restored short-chain fatty acids (SCFAs) as well as neurotransmitter release (γ-GABA, 5-HT, and dopamine), which stemmed from the production of butyric acid (BA). Pu-erh tea and BA promoted the production of SCFAs by reshaping the gut microbes (increased Lactobacillus, Akkermansia, Faecalibaculum), thereby downregulating gut inflammatory protein expression (PI3K/AKT/NF-κB). SCFAs, especially BA, intervened directly in the blood-brain barrier via the gut-brain axis to restore neurotransmitter release. Collectively, our results highlighted that increasing BA through Pu-erh tea consumption may be a key mechanism for improving colitis-mediated brain dysfunction by lowering gut inflammation and balancing gut microbe-gut-brain axis homeostasis. These results provide a promising step that might encourage further investigations of Pu-erh tea as a protective agent for brain function in colitis patients.
Collapse
Affiliation(s)
- Shanshan Hu
- College of Food Science, Southwest University, Beibei, Chongqing, 400715, China.
| | - Zhiyuan Lin
- College of Food Science, Southwest University, Beibei, Chongqing, 400715, China.
| | - Sibo Zhao
- College of Food Science, Southwest University, Beibei, Chongqing, 400715, China.
| | - Bowen Zhang
- College of Food Science, Southwest University, Beibei, Chongqing, 400715, China.
| | - Liyong Luo
- College of Food Science, Southwest University, Beibei, Chongqing, 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Beibei, Chongqing, 400715, China.
| | - Liang Zeng
- College of Food Science, Southwest University, Beibei, Chongqing, 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Beibei, Chongqing, 400715, China.
| |
Collapse
|
41
|
Wu J, Yu C, Shen S, Ren Y, Cheng H, Xiao H, Liu D, Chen S, Ye X, Chen J. RGI-Type Pectic Polysaccharides Modulate Gut Microbiota in a Molecular Weight-Dependent Manner In Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:2160-2172. [PMID: 36648986 DOI: 10.1021/acs.jafc.2c07675] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
In this study, the fermentation characteristics of high rhamnogalacturonan I pectic polysaccharides (RGI) and free-radical degraded RGI (DRGI) were evaluated by a human fecal batch-fermentation model, and their structural properties were also investigated. As a result, the Mw of RGI decreased from 246.8 to 11.6 kDa, and the branches were broken dramatically. Fermentation showed that RGI degraded faster and produced more acetate and propionate than DRGI. Both of them reduced the Firmicutes/Bacteroidetes ratio and promoted the development of Bacteroides, Bifidobacterium, and Lactobacillus, bringing benefits to the gut ecosystem. However, the composition and metabolic pathways of the microbiota in RGI and DRGI were different. Most of the dominant bacteria of RGI (such as [Eubacterium]_eligens_group) participated in carbohydrate utilization, leading to better performance in glucolipid metabolism and energy metabolism. This work elucidated that large molecular weight matters in the gut microbiota modulatory effect of RGI-type pectic polysaccharides in vitro.
Collapse
Affiliation(s)
- Jiaxiong Wu
- College of Biosystems Engineering and Food Science, Ningbo Innovation Center, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agri-Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou310058, China
| | - Chengxiao Yu
- College of Biosystems Engineering and Food Science, Ningbo Innovation Center, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agri-Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou310058, China
| | - Sihuan Shen
- College of Biosystems Engineering and Food Science, Ningbo Innovation Center, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agri-Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou310058, China
| | - Yanming Ren
- College of Biosystems Engineering and Food Science, Ningbo Innovation Center, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agri-Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou310058, China
| | - Huan Cheng
- College of Biosystems Engineering and Food Science, Ningbo Innovation Center, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agri-Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou310058, China
- Zhejiang University Zhongyuan Institute, Zhengzhou 450000, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
| | - Hang Xiao
- College of Biosystems Engineering and Food Science, Ningbo Innovation Center, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agri-Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou310058, China
| | - Donghong Liu
- College of Biosystems Engineering and Food Science, Ningbo Innovation Center, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agri-Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou310058, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
| | - Shiguo Chen
- College of Biosystems Engineering and Food Science, Ningbo Innovation Center, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agri-Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou310058, China
- Zhejiang University Zhongyuan Institute, Zhengzhou 450000, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, Ningbo Innovation Center, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agri-Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou310058, China
- Zhejiang University Zhongyuan Institute, Zhengzhou 450000, China
| | - Jianle Chen
- College of Biosystems Engineering and Food Science, Ningbo Innovation Center, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agri-Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou310058, China
- NingboTech University, Ningbo315100, China
- Zhejiang University Zhongyuan Institute, Zhengzhou 450000, China
| |
Collapse
|
42
|
Crosstalk between imbalanced gut microbiota caused by antibiotic exposure and rotavirus replication in the intestine. Heliyon 2023; 9:e12718. [PMID: 36685479 PMCID: PMC9850052 DOI: 10.1016/j.heliyon.2022.e12718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 12/26/2022] [Accepted: 12/26/2022] [Indexed: 01/03/2023] Open
Abstract
Objective Rotavirus (RV), one of non-enveloped double-strained RNA viruses, can cause infantile diarrheal illness. It is widely accepted that RV is transmitted mainly via feces-oral route. However, infected asymptomatic adults are becoming the source of infection. It is necessary to explore the underlying mechanism of RV replication in adult's intestine. Methods After recruiting healthy volunteers and RV asymptomatic carriers, we firstly investigated the association of animal-derived food intake with antibiotic level in urine samples. Secondly, we compared the difference in the structure of gut microbiota, and identified the taxa that most likely explained the difference. Finally, we investigated the impact of lipopolysaccharide (LPS), produced by gram-negative bacteria, on RV replication in vivo and in vitro. Results We found that 10% of participants were RV asymptomatic carriers in our study. High intake of animal-derived food was positively correlated to antibiotic level in urine samples. The disrupted gut microbiota in RV carriers was characterized by high abundance of antibiotic resistant gram-negative bacteria and high level of LPS. The disrupted gut microbiota caused by penicillin treatment was benefit to RV replication in vivo. LPS enhanced RV thermal stability in vitro. Conclusions Our findings suggest that the imbalanced gut microbiota caused by antibiotic exposure plays an important role in RV replication, and brings risk to health complications.
Collapse
|
43
|
He Y, Song Z, Ji Y, Tso P, Wu Z. Preventive Effects of l-Glutamine on High-Fat Diet-Induced Metabolic Disorders Linking with Regulation of Intestinal Barrier Integrity, Hepatic Lipid Metabolism, and Gut Microbiota in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:11923-11934. [PMID: 36122193 DOI: 10.1021/acs.jafc.2c01975] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The present study was conducted to investigate the effects of l-glutamine (Gln) on a high-fat diet (HFD)-induced lipid metabolic abnormality and explore its possible mechanisms. The results demonstrated that Gln administration reduced body weight, improved serum lipids, and decreased glucose tolerance in HFD-fed rats. Meanwhile, Gln administration alleviated liver injury, reduced the hepatic inflammatory response by inhibiting NLRP3 inflammasome activation, and decreased hepatic lipid accumulation by promoting VLDL secretion and fatty acid β-oxidation, as well as reduced bile acid synthesis by activating hepatic and ileal FXR in HFD-fed rats. Moreover, Gln administration restored HFD-induced intestinal barrier dysfunction, promoted intestinal fat absorption, suppressed intestinal inflammation, and also reshaped the gut microbiota composition in HFD-fed rats by downregulating the abundance of potential pathogens Escherichia-Shigella and upregulating the abundance of beneficial bacteria such as Akkermansia. To conclude, the present results showed that Gln may be a potential option for preventing HFD-induced metabolic disorders via the gut-liver axis.
Collapse
Affiliation(s)
- Yu He
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, China
| | - Zhuan Song
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, China
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, 2120 E. Galbraith Road, Building A, Cincinnati, Ohio 45237, United States
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| |
Collapse
|
44
|
Miyajima Y, Karashima S, Ogai K, Taniguchi K, Ogura K, Kawakami M, Nambo H, Kometani M, Aono D, Demura M, Yoneda T, Tsujiguchi H, Hara A, Nakamura H, Okamoto S. Impact of gut microbiome on dyslipidemia in japanese adults: Assessment of the Shika-machi super preventive health examination results for causal inference. Front Cell Infect Microbiol 2022; 12:908997. [PMID: 36118024 PMCID: PMC9479221 DOI: 10.3389/fcimb.2022.908997] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Dyslipidemia (DL) is one of the most common lifestyle-related diseases. There are few reports showing the causal relationship between gut microbiota (GM) and DL. In the present study, we used a linear non-Gaussian acyclic model (LiNGAM) to evaluate the causal relationship between GM and DL. A total of 79 men and 82 women aged 40 years or older living in Shika-machi, Ishikawa Prefecture, Japan were included in the analysis, and their clinical information was investigated. DNA extracted from the GM was processed to sequence the 16S rRNA gene using next-generation sequencing. Participants were divided into four groups based on sex and lipid profile information. The results of one-way analysis of covariance, linear discriminant analysis effect size, and least absolute value reduction and selection operator logistic regression model indicated that several bacteria between men and women may be associated with DL. The LiNGAM showed a presumed causal relationship between different bacteria and lipid profiles in men and women. In men, Prevotella 9 and Bacteroides were shown to be potentially associated with changes in low- and high-density lipoprotein cholesterol levels. In women, the LiNGAM results showed two bacteria, Akkermansia and Escherichia/Shigella, had a presumptive causal relationship with lipid profiles. These results may provide a new sex-based strategy to reduce the risk of developing DL and to treat DL through the regulation of the intestinal environment using specific GM.
Collapse
Affiliation(s)
- Yuna Miyajima
- Department of Clinical Laboratory Science, Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Shigehiro Karashima
- Institute of Liberal Arts and Science, Kanazawa University, Kanazawa, Japan
- *Correspondence: Shigehiro Karashima, ; Shigefumi Okamoto,
| | - Kazuhiro Ogai
- AI Hospital/Macro Signal Dynamics Research and Development Center, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kouki Taniguchi
- Department of Clinical Laboratory Science, Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kohei Ogura
- Advanced Health Care Science Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Masaki Kawakami
- School of Electrical, Information and Communication Engineering, College of Science and Engineering, Kanazawa University, Kanazawa, Japan
| | - Hidetaka Nambo
- School of Electrical, Information and Communication Engineering, College of Science and Engineering, Kanazawa University, Kanazawa, Japan
| | - Mitsuhiro Kometani
- Department of Endocrinology and Metabolism, Kanazawa University Hospital, Kanazawa, Japan
| | - Daisuke Aono
- Department of Endocrinology and Metabolism, Kanazawa University Hospital, Kanazawa, Japan
| | - Masashi Demura
- Department of Hygiene, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Takashi Yoneda
- Department of Endocrinology and Metabolism, Kanazawa University Hospital, Kanazawa, Japan
- Department of Health Promotion and Medicine of the Future, Kanazawa University, Kanazawa, Japan
- Faculty of Transdisciplinary Sciences, Institute of Transdisciplinary Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiromasa Tsujiguchi
- Department of Public Health, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Akinori Hara
- Department of Public Health, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroyuki Nakamura
- Department of Public Health, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shigefumi Okamoto
- Department of Clinical Laboratory Science, Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
- Advanced Health Care Science Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
- *Correspondence: Shigehiro Karashima, ; Shigefumi Okamoto,
| |
Collapse
|
45
|
Xu R, Pan J, Zhou W, Ji G, Dang Y. Recent advances in lean NAFLD. Biomed Pharmacother 2022; 153:113331. [PMID: 35779422 DOI: 10.1016/j.biopha.2022.113331] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 02/09/2023] Open
Abstract
As the predominant type of chronic liver disease, the growing prevalence of nonalcoholic fatty liver disease (NAFLD) has become a concern worldwide. Although obesity plays the most pivotal role in NAFLD, approximately 10-20% of individuals with NAFLD who are not overweight or obese (BMI < 25 kg/m2, or BMI < 23 kg/m2 in Asians) have "lean NAFLD." Lean individuals with NAFLD have a lower prevalence of diabetes, hypertension, hypertriglyceridemia, central obesity, and metabolic syndrome than nonlean individuals with NAFLD, but higher fibrosis scores and rates of cardiovascular morbidity and all-cause mortality in advanced stages. The pathophysiological mechanisms of lean NAFLD remain poorly understood. Studies have shown that lean NAFLD is more correlated with factors such as environmental, genetic susceptibility, and epigenetic regulation. This review will examine the way in which the research progress and characteristic of lean NAFLD, and explore the function of epigenetic modification to provide the basis for the clinical treatment and diagnosis of lean NAFLD.
Collapse
Affiliation(s)
- Ruohui Xu
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jiashu Pan
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Department of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Yanqi Dang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
46
|
Zhang XL, Wang TY, Targher G, Byrne CD, Zheng MH. Lifestyle Interventions for Non-Obese Patients Both with, and at Risk, of Non-Alcoholic Fatty Liver Disease. Diabetes Metab J 2022; 46:391-401. [PMID: 35656562 PMCID: PMC9171159 DOI: 10.4093/dmj.2022.0048] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 04/29/2022] [Indexed: 12/26/2022] Open
Abstract
Non-alcoholic fatty liver disease occurring in non-obese subjects (the so-called non-obese NAFLD) is a highly prevalent but neglected liver condition, which is closely associated with metabolic disorders and suboptimal lifestyles. Landmark studies have shown that lifestyle interventions are potentially beneficial in decreasing the risk of developing non-obese NAFLD and in ameliorating NAFLD in non-obese individuals with pre-existing NAFLD. Lifestyle interventions usually refer to changes in eating habits and physical activity, both of which have a powerful effect on non-obese NAFLD and on risk factors for non-obese NAFLD. However, to date, patients and health-care professionals have a poor awareness and understanding of non-obese NAFLD and the beneficial effects of lifestyle interventions in this patient population. The aim of this narrative review is to briefly discuss the evidence for the effects of lifestyle changes and what changes are needed amongst medical personnel and other stakeholders in order to raise awareness of non-obese NAFLD.
Collapse
Affiliation(s)
- Xin-Lei Zhang
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ting-Yao Wang
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Verona, Verona, Italy
| | - Christopher D. Byrne
- Southampton National Institute for Health Research Biomedical Research Centre, University Hospital Southampton, Southampton General Hospital, Southampton, UK
- Nutrition and Metabolism, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Ming-Hua Zheng
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| |
Collapse
|
47
|
Hu J, Guo P, Mao R, Ren Z, Wen J, Yang Q, Yan T, Yu J, Zhang T, Liu Y. Gut Microbiota Signature of Obese Adults Across Different Classifications. Diabetes Metab Syndr Obes 2022; 15:3933-3947. [PMID: 36601354 PMCID: PMC9807070 DOI: 10.2147/dmso.s387523] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/28/2022] [Indexed: 12/29/2022] Open
Abstract
PURPOSE Obesity is currently a major global public health issue. It has been shown by many that gut microbiota and microbial factors regulate the pathogenesis of obesity and metabolic abnormalities, but little is known about their roles in the different degrees of obesity. Here, we sought to investigate the microbial signatures of obesity of various severities. PATIENTS AND METHODS We did this by characterizing the intestinal microbiome signature in a Chinese cohort of obese patients and healthy controls using 16S rRNA gene sequencing. To this end, obesity was sub-divided into four subgroups, including "Overweight", Class I, Class II, and Class III obesity, based on body mass index (BMI). RESULTS Microbial diversity decreased in obese subjects, and the reduction trend was correlated with the severity of obesity. We detected an expansion of Escherichia shigella in obese patients compared to healthy controls. The family Eubacterium coprostanoligenes and Tannerellaceae, the genera Eubacterium coprostanoligenes, Lachnospiraceae NK4A136, Parabacteroides, and Akkermansia, and the species Prevotella copri were microbial biomarkers of healthy people. Gammaproteobacteria and Enterobacterales were biomarkers of being "Overweight". Erysipelatoclostridiaceae was a biomarker of Class I obesity. The class Bacilli and the order Lactobacillales were both biomarkers of Class II obesity. Negativicutes was a biomarker of Class III obesity. We further established relationships between this microbiome data and other biochemical data, including albumin, low-density lipoprotein (LDL), high-density lipoprotein (HDL), vitamin folic acid (FA) and vitamin B12 (VB12), and Interleukin-6 (IL-6) levels. Function prediction results showed a marked energy metabolism dysbiosis in obesity, especially in patients with Class III obesity. CONCLUSION These results suggested that people with different levels of obesity had distinct gut microbial signatures. Decreased microbial diversity, depletion of some specific taxa, and deviation in potential functions mirrored the severity of obesity in this cohort.
Collapse
Affiliation(s)
- Junqing Hu
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
- Medical Research Center, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
| | - Pengsen Guo
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
| | - Rui Mao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Zhengyun Ren
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
- College of Medicine, Southwest Jiaotong University, Chengdu, People’s Republic of China
| | - Jun Wen
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
- College of Medicine, Southwest Jiaotong University, Chengdu, People’s Republic of China
| | - Qin Yang
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
- Department of General Surgery, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
| | - Tong Yan
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
| | - Jiahui Yu
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
| | - Tongtong Zhang
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
- Medical Research Center, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
- Correspondence: Tongtong Zhang, Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, & Medical Research Center, the Third People’s Hospital of Chengdu, Chengdu, People’s Republic of China, Email ; Yanjun Liu, Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, the Third People’s Hospital of Chengdu, No. 82, Qinglong Street, Qingyang District, Chengdu, 610031, People’s Republic of China, Emai
| | - Yanjun Liu
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, People’s Republic of China
| |
Collapse
|