1
|
Chen F, Guo Z, Chen Y, Li S, Chen P. Non-alcoholic fatty liver disease enhances the beneficial effect of renal denervation on gut microbiota aberrations in rats with heart failure. BMC Microbiol 2025; 25:311. [PMID: 40399816 PMCID: PMC12093722 DOI: 10.1186/s12866-025-04027-y] [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: 02/23/2024] [Accepted: 05/06/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Renal denervation (RDN) contributes to improving cardiac function by ameliorating aberrations of the gut microbiota, and non-alcoholic fatty liver disease (NAFLD) is associated with gut microbiota dysbiosis and is critically involved in the development of heart failure (HF). It is unclear whether the beneficial effect of RDN on gut microbiota in HF can be affected by NAFLD and whether this effect changes with the severity of NAFLD. METHODS HF Sprague Dawley rats induced by transverse aortic constriction were fed a high-fat-fructose diet and underwent RDN, and sequencing of 16S rRNA gene in fecal samples was detected. RESULTS The dissimilarity coefficients and sample distances of the intestinal microbiome were elevated in HF rats with NAFLD. After RDN, HF rats with NAFLD had fewer bacteria harmful to cardiac function, such as Alphaproteobacteria, Bacteroidota and Prevotella-9, and more bacteria beneficial to HF, such as Monoglobaceae, Proteobacteria and Monoglobales, than HF rats without NAFLD (all p < 0.05). This tendency also existed but was much less significant when compared between HF rats with non-alcoholic steatohepatitis (NASH) and without NAFLD. Predictive functional profiling of microbial communities revealed that after RDN, the abundance of membrane transport, environmental and genetic information processing was significantly higher, and glycan biosynthesis and metabolism was significantly lower in HF rats with NAFLD than in those without NAFLD. CONCLUSION NAFLD could further enhance the beneficial role of RDN in mitigating gut microbiota aberrations in HF rats by increasing beneficial bacteria and decreasing bacteria harmful to cardiac function, but this effect was not apparent in NASH rats.
Collapse
Affiliation(s)
- Fuyan Chen
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, 1 Panfu Road, Guangzhou, Guangdong, 510182, China
| | - Zhiqin Guo
- Department of Cardiology, Guangzhou First People's Hospital, Guangzhou, China
| | - Yufeng Chen
- Department of Cardiology, Maoming People's Hospital, Maoming, China
| | - Shun Li
- Department of Cardiology, Guangzhou First People's Hospital, Guangzhou, China
| | - Pingan Chen
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, 1 Panfu Road, Guangzhou, Guangdong, 510182, China.
| |
Collapse
|
2
|
Chen Y, Chen Z, Liang L, Li J, Meng L, Yuan W, Xie B, Zhang X, Feng L, Jia Y, Fu Z, Su P, Tong Z, Zhong J, Liu X. Multi-kingdom gut microbiota dysbiosis is associated with the development of pulmonary arterial hypertension. EBioMedicine 2025; 115:105686. [PMID: 40220715 PMCID: PMC12013117 DOI: 10.1016/j.ebiom.2025.105686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/07/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Gut microbiota dysbiosis has been implicated in pulmonary arterial hypertension (PAH). However, the exact roles and underlying mechanisms of multi-kingdom gut microbiota, including bacteria, archaea, and fungi, in PAH remain largely unclear. METHODS The shotgun metagenomics was used to analyse multi-kingdom gut microbial communities in patients with idiopathic PAH (IPAH) and healthy controls. Furthermore, fecal microbiota transplantation (FMT) was performed to transfer gut microbiota from IPAH patients or monocrotaline (MCT)-PAH rats to normal rats and from normal rats to MCT-PAH rats. FINDINGS Gut microbiota analysis revealed substantial alterations in the bacterial, archaeal, and fungal communities in patients with IPAH compared with healthy controls. Notably, FMT from IPAH patients or MCT-PAH rats induced PAH phenotypes in recipient rats. More intriguingly, FMT from normal rats to MCT-PAH rats significantly ameliorated PAH symptoms; restored gut bacteria, archaea, and fungi composition; and shifted the plasma metabolite profiles of MCT-PAH rats toward those of normal rats. In parallel, RNA-sequencing analysis demonstrated the expression of genes involved in key signalling pathways related to PAH. A panel of multi-kingdom markers exhibited superior diagnostic accuracy compared with single-kingdom panels for IPAH. INTERPRETATION Our findings established an association between multi-kingdom gut microbiota dysbiosis and PAH, thereby indicating the therapeutic potential of FMT in PAH. More importantly, apart from gut bacteria, gut archaea and fungi were also significantly associated with PAH pathogenesis, highlighting their indispensable role in PAH. FUNDING This work was supported by Noncommunicable Chronic Diseases-National Science and Technology Major Projects No. 2024ZD0531200, No. 2024ZD0531201 (Research on Prevention and Treatment of Cancer, Cardiovascular and Cerebrovascular Diseases, Respiratory Diseases, and Metabolic Diseases), the National Natural Science Foundation of China of China (No. 82170302, 82370432), Financial Budgeting Project of Beijing Institute of Respiratory Medicine (Ysbz2025004, Ysbz2025007), National clinical key speciality construction project Cardiovascular Surgery, Reform and Development Program of Beijing Institute of Respiratory Medicine (Ggyfz202417, Ggyfz202501), Clinical Research Incubation Program of Beijing Chaoyang Hospital Affiliated to Capital Medical University (CYFH202209).
Collapse
Affiliation(s)
- Yihang Chen
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zhenzhen Chen
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Lirong Liang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Clinical Epidemiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 100020, Beijing, China
| | - Jifeng Li
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Liukun Meng
- Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Wen Yuan
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Boqia Xie
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xun Zhang
- Department of Pharmacy, Chinese PLA General Hospital, 100039, Beijing, China
| | - Lin Feng
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Clinical Epidemiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 100020, Beijing, China
| | - Yanxiong Jia
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zhou Fu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Pixiong Su
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zhaohui Tong
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Jiuchang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Xiaoyan Liu
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
3
|
Gong X, Bai S, Lei E, Lu T, Chen Y, Cai J, Liu J. A bibliometric analysis of metabolic dysfunction-associated steatotic liver disease in children from 2004 to 2024. Front Pediatr 2025; 13:1468788. [PMID: 40356777 PMCID: PMC12066688 DOI: 10.3389/fped.2025.1468788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Background Metabolic dysfunction-associated steatotic liver disease (MASLD), once known as Non-alcoholic fatty liver disease, impacts between 3% and 10% of children and adolescents globally, as well as nearly one-third of obsessed boys and one-quarter of obsessed girls, and is the most frequent cause of pediatric liver disease associated with the obesity epidemic. With the growing attention and increasing volume of literature on pediatric MASLD, there is an urgent need for bibliometric analysis and visualization in the area of pediatric MASLD study in terms of dissecting study priorities. Methods Literature was searched in the Web of Science Core Collection database, followed by categorization, bibliometric study as well as visual analysis conducted by applying software including Citespace, VOSviewer, and the R language. The study concentrated on analyzing information related to key authors, spatial and temporal distribution, core keywords, and important citations. Results In total, 3,409 publications on pediatric MASLD were collected in the study, including 2,697 articles and 712 review articles. Between 2004 and 2024, the volume of publications had been constantly increasing per year. The country with the most numerous publications was the United States, which had extensive exchanges and collaborations with Italy, China, and England, followed by Italy. The Journal of Pediatric Gastroenterology and Nutrition had the greatest quantity of publications in this domain. The core literature was a clinical guideline. Insulin resistance, metabolic syndrome, steatohepatitis, hepatocellular carcinoma, cardiovascular risk, diabetes risk, diagnostic accuracy, lifestyle intervention, gut microbiome, probiotics, and metabolic dysfunction-associated steatotic liver disease were also hot topics and frontier trends in pediatric MASLD studies. Conclusion This research represents the inaugural application of bibliometric analysis to examine the developmental trajectory of pediatric MASLD studies over the past two decades, which reveals that the etiology, pathological changes of the liver, relationship with obesity, complications, comorbidities, diagnosis and treatments of pediatric MASLD are the key focuses and provides academic references for pediatric clinicians and scholars to grasp the hotspots, the cutting edge and the evolving trends in the area.
Collapse
Affiliation(s)
- Xiaowei Gong
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Pediatrics, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Siyu Bai
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Enze Lei
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Tao Lu
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yao Chen
- Department of Pediatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Key Laboratory of Liver and Kidney Research and Application of Traditional Chinese Medicine, Wuhan, China
| | - Jianxin Cai
- Department of Pediatrics, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Jianzhong Liu
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Pediatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Key Laboratory of Liver and Kidney Research and Application of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
4
|
Liu SJ, Duan JH, Chen YY, Gu SL, He YH, Xue MM, Yue JY. Unraveling the triglyceride-glucose index: a key predictor of liver fat content and the amplifying role of BMI: evidence from a large physical examination data. Front Endocrinol (Lausanne) 2025; 16:1555300. [PMID: 40352458 PMCID: PMC12061704 DOI: 10.3389/fendo.2025.1555300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Background The triglyceride-glucose (TyG) index is associated with the severity of metabolic-associated fatty liver disease (MASLD), but its link to liver fat content is not fully understood. This study investigates the relationship between the TyG index and liver fat content and explores the role of body mass index (BMI) as a mediator. Methods This cross-sectional study analyzed data from 12,750 participants who underwent health screenings at the first affiliated hospital of Xinxiang Medical University between January 2018 and December 2023. The TyG index, derived as Ln [triglycerides (mg/dl) * fasting plasma glucose (mg/dl)/2], was the independent variable, while liver fat content, measured by quantitative computed tomography (QCT), was the dependent variable. Participants were grouped into tertiles based on their TyG index. Univariate and multivariate analyses, smooth curve fitting (generalized additive models), threshold effect analysis, and subgroup analyses were used to assess the TyG-liver fat content relationship. BMI's mediating effect was also examined. Results Liver fat content increased steadily across TyG index tertiles. After adjusting for confounders, the TyG index remained independently associated with liver fat content [β = 1.42, 95% CI: 1.26-1.57]. Participants in the highest TyG tertile (T3) had a 1.58-fold higher liver fat content compared to those in the lowest tertile (T1) (95% CI: 1.37-1.80, P<0.001). A generalized additive model showed a nonlinear relationship between TyG index and liver fat content. When the TyG index ≤ 7.39, liver fat content increased gradually (β = 0.74, 95% CI: 0.50-0.99, P<0.001). Beyond this threshold, liver fat content rose sharply (β = 2.19, 95% CI: 1.92-2.46, P<0.001). Subgroup analysis indicated that the association between TyG index and liver fat content was stronger at higher BMI levels (P for interaction < 0.001). Mediation analysis revealed that BMI accounted for 26.68% of the observed effect. Conclusion The TyG index is positively associated with liver fat content in a nonlinear manner, with BMI amplifying this effect. These results suggest that the TyG index may be a useful marker for predicting liver fat content, and managing weight could help slow the progression of MASLD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jun-Yan Yue
- Department of Radiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
5
|
Meybodi SM, Rezazadeh Khabaz MJ, Vojdani A, Nasiri Z, Mazhari SA, Tabar FA, Javazm SA, Owrang M, Noori Z, Pishva MS, Badameh P, Maleki MH, Nadimi E. Bifidobacterium adolescentis prevents diabetes-induced liver injury via pyroptosis attenuation. Exp Cell Res 2025; 447:114518. [PMID: 40097086 DOI: 10.1016/j.yexcr.2025.114518] [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: 06/02/2024] [Revised: 02/24/2025] [Accepted: 03/11/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), along with non-alcoholic steatohepatitis (NASH), lacks definitive therapy and typically remains asymptomatic until reaching advanced stages. Lipid metabolism and inflammation management using probiotics such as Bifidobacterium adolescentis is suggested to alleviate or suppress NAFLD development. Hence, this study aims to investigate the effects of Bifidobacterium adolescentis treatment on mitigating pyroptosis, an inflammatory cell death pathway, in the liver of rats with NAFLD induced by high-fat diet (HFD) and streptozotocin (STZ) administration. METHODS Forty 8-week adult male Sprague Dawley rats were divided into four groups. Bifidobacterium adolescentis was administered for 8 and 16 weeks at 4 × 1010 CFU/day to rats fed a high-fat diet (HFD). Subsequently, the mRNA expression levels of pyroptotic-related genes including Cas1, Cas3, Cas11, NLRP3, GSDMD, IL-1β, and NF-κB were quantified in liver tissue using quantitative polymerase chain reaction (qPCR). Histopathological alterations and stereological changes in liver structure, as well as lipid profile (FBG, TG, TC, HDL, LDL), and liver indices (ALT, AST, ALP, LDH), were also evaluated across the different groups. RESULTS Bifidobacterium adolescentis administration significantly reduced the expression levels of NF-κB and pyroptotic-related genes. Additionally, this probiotic effectively reversed the adverse effects of the high-fat diet (HFD) on liver volume, Kupffer cell numbers, and hepatocyte nuclei. Furthermore, it improved the lipid profile and liver indices of rats fed with the HFD. CONCLUSION This study demonstrates that B. adolescentis supplementation prevents diabetes-induced liver injury by attenuating pyroptosis. These findings suggest that Bifidobacterium adolescentis may be a promising therapeutic approach for managing NAFLD and its associated complications, primarily by modulating key genes associated with pyroptosis and inflammation in rats fed with a high-fat diet.
Collapse
Affiliation(s)
- Seyed Mohammadmahdi Meybodi
- Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | | | - Andia Vojdani
- Department of Microbiology, School of Biology, University of Tehran, Tehran, Iran.
| | - Zahra Nasiri
- Department of Cellular and Molecular Biology, Faculty of Materials, Najafabad Branch, Islamic Azad University, Isfahan, Iran.
| | | | - Farideh Akhlaghi Tabar
- Department of Genetics, Faculty of Basic Science, Qom Branch, Islamic Azad University, Qom, Iran.
| | - Sara Abdizadeh Javazm
- Department of Microbiology, Faculty of Sciences, Karaj Branch, Islamic Azad University, Karaj, Iran.
| | - Marzieh Owrang
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Zahra Noori
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Maryam Sadat Pishva
- University of Tehran, Kish International Campus, School of Biology, Kish Island, Iran.
| | - Parisa Badameh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mohammad Hasan Maleki
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Elham Nadimi
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Medical Biotechnology Department, School of Advanced, Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
6
|
Hu L, Du H, Zhou Q, Liu C, Zhang T, Yuan M. Web of Science-Based Visualization of Metabolic Dysfunction-Associated Fatty Liver Disease in Pediatric and Adolescent Populations: A Bibliometric Study. Health Sci Rep 2025; 8:e70409. [PMID: 39897463 PMCID: PMC11779742 DOI: 10.1002/hsr2.70409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/31/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
Background and Aims The prevalence of metabolism-associated fatty liver disease (MAFLD) in children is on the rise. This study employs visualization and analysis to evaluate the research implications, current advancements, and emerging trends in pediatric MAFLD, with the aim of elucidating its pathogenesis and informing the development of clinical treatment strategies. Methods Using visualization software, we conducted a visual analysis and mapping of the journal distribution, leading institutions, prominent authors, annual publication trends, and keyword frequencies among the 1179 scholarly articles retrieved from the Web of Science Core Collection for this study. Results The overall publication volume demonstrated an upward trend, with a total of 200 journals, contributions from 63 countries, 882 research institutions, and 5605 authors involved, including 84 who were identified as core authors. The main research team is led by Nobili, Valerio. The main research institutions are concentrated in Italy, the United States, and China. A total of 473 keywords were included, and the keywords with high frequency and medium centricity are insulin resistance, metabolic syndrome, children, steatohepatitis, adolescents, hepatic steatosis, nash, obesity, diagnosis, and fibrosis, which resulted in six keyword clusters. Conclusion MAFLD represents a significant public health concern. Research on children and adolescents with MAFLD continues to attract high interest. Noninvasive diagnostic methods, pathogenesis (intestinal microbiota research), disease prediction (gene research) are current research hotspots.
Collapse
Affiliation(s)
- Liangyu Hu
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Huarong Du
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - QianQian Zhou
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Chunlei Liu
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Tiansong Zhang
- Department of Traditional Chinese Medicine, Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Min Yuan
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
7
|
Pan X, Song Y, Liang Y, Feng G, Wang Z. Roseburia intestinalis: A possible target for vascular calcification. Heliyon 2024; 10:e39865. [PMID: 39524709 PMCID: PMC11550659 DOI: 10.1016/j.heliyon.2024.e39865] [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: 01/21/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
With the advancement of metagenomics and metabolomics techniques, the crucial role of the gut microbiome in intestinal, cardiovascular, and metabolic disorders has been extensively explored. Vascular calcification (VC) is common in atherosclerosis, hypertension, diabetes mellitus, and chronic kidney disease. Moreover, it is a significant cause of cardiovascular diseases and mortality. Roseburia intestinalis, as a promising candidate for the next generation of probiotics, plays a substantial role in inhibiting the systemic inflammatory response and holds great potential in the treatment of intestinal diseases, cardiovascular diseases, and metabolic disorders. Its primary metabolite, butyrate, acts on specific receptors (GPR43, GPR41, GPR109a). It enters cells via transporters (MCT1, SMCT1), affecting gene expression through HDACs, PPARγ and Nrf2, promoting energy metabolism and changing the concentration of other metabolites (including AGEs, LPS, BHB) in the circulation to affect the body's life activities. In this paper, we focus on the possible mechanism of the primary metabolite butyrate of Roseburia intestinalis in inhibiting VC, which may become a potential therapeutic target for the treatment of VC and the ways to enhance its effect.
Collapse
Affiliation(s)
- Xinyun Pan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| | - Yunjian Song
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| | - Yapeng Liang
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Guoquan Feng
- Department of Imaging, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| |
Collapse
|
8
|
Vieira JIG, Braga LG, Chud TCS, Ferreira PH, Guimarães SEF, Martins MF, do Carmo Panetto JC, Machado MA, Silva DBDS, Bonafé CM, Magalhães AFB, da Silva MVGB, Verardo LL. Resequencing of Brazilian locally adapted cattle breeds revealed variants in candidate genes and transcription factors for meat fatty acid profile. J Anim Breed Genet 2024; 141:628-642. [PMID: 38686591 DOI: 10.1111/jbg.12869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
The beef cattle industry has experienced a shift driven by a market demand for healthier meat, cost efficiency and environmental sustainability in recent years. Consequently, there has been a growing focus on the fatty acids content and functions of meat in cattle breeding programmes. Besides, a deeper understanding of the biological mechanisms influencing the expression of different phenotypes related to fatty acid profiles is crucial. In this study, we aimed to identify Single-Nucleotide Variants (SNV) and Insertion/Deletion (InDels) DNA variants in candidate genes related to fatty acid profiles described in genomic, transcriptomic and proteomic studies conducted in beef cattle breeds. Utilizing whole-genome re-sequencing data from Brazilian locally adapted bovine breeds, namely Caracu and Pantaneiro, we identified SNVs and InDels associated with 23,947 genes. From these, we identified 318 candidate genes related to fatty acid profiles that contain variants. Subsequently, we select only genes with SNVs and InDels in their promoter, 5' UTR and coding region. Through the gene-biological process network, approximately 19 genes were highlighted. Furthermore, considering the studied trait and a literature review, we selected the main transcription factors (TF). Functional analysis via gene-TF network allowed us to identify the 30 most likely candidate genes for meat fatty acid profile in cattle. LIPE, MFSD2A and SREBF1 genes were highlighted in networks due to their biological importance. Further dissection of these genes revealed 15 new variants found in promoter regions of Caracu and Pantaneiro sequences. The gene networks facilitated a better functional understanding of genes and TF, enabling the identification of variants potentially related to the expression of candidate genes for meat fatty acid profiles in cattle.
Collapse
Affiliation(s)
| | - Larissa Graciano Braga
- Departamento de Engenharia e Ciências Exatas, Universidade Estadual Paulista, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | - Lucas Lima Verardo
- Universidade Federal dos Vales Do Jequitinhonha e Mucuri, Diamantina, Minas Gerais, Brazil
| |
Collapse
|
9
|
Zhai Z, Yang Y, Chen S, Wu Z. Long-Term Exposure to Polystyrene Microspheres and High-Fat Diet-Induced Obesity in Mice: Evaluating a Role for Microbiota Dysbiosis. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:97002. [PMID: 39226184 PMCID: PMC11370995 DOI: 10.1289/ehp13913] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 07/23/2024] [Accepted: 08/08/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Microplastics (MPs) have become a global environmental problem, emerging as contaminants with potentially alarming consequences. However, long-term exposure to polystyrene microspheres (PS-MS) and its effects on diet-induced obesity are not yet fully understood. OBJECTIVES We aimed to investigate the effect of PS-MS exposure on high-fat diet (HFD)-induced obesity and underlying mechanisms. METHODS In the present study, C57BL/6J mice were fed a normal diet (ND) or a HFD in the absence or presence of PS-MS via oral administration for 8 wk. Antibiotic depletion of the microbiota and fecal microbiota transplantation (FMT) were performed to assess the influence of PS-MS on intestinal microbial ecology. We performed 16S rRNA sequencing to dissect microbial discrepancies and investigated the dysbiosis-associated intestinal integrity and inflammation in serum. RESULTS Compared with HFD mice, mice fed the HFD with PS-MS exhibited higher body weight, liver weight, metabolic dysfunction-associated steatotic liver disease (MASLD) activity scores, and mass of white adipose tissue, as well as higher blood glucose and serum lipid concentrations. Furthermore, 16S rRNA sequencing of the fecal microbiota revealed that mice fed the HFD with PS-MS had greater α -diversity and greater relative abundances of Lachnospiraceae, Oscillospiraceae, Bacteroidaceae, Akkermansiaceae, Marinifilaceae, Deferribacteres, and Desulfovibrio, but lower relative abundances of Atopobiaceae, Bifidobacterium, and Parabacteroides. Mice fed the HFD with PS-MS exhibited lower expression of MUC2 mucin and higher levels of lipopolysaccharide and inflammatory cytokines [tumor necrosis factor-α (TNF-α ), interleukin-6 (IL-6), IL-1β , and IL-17A] in serum. Correlation analyses revealed that differences in the microbial flora of mice exposed to PS-MS were associated with obesity. Interestingly, microbiota-depleted mice did not show the same PS-MS-associated differences in Muc2 and Tjp1 expression in the distal colon, expression of inflammatory cytokines in serum, or obesity outcomes between HFD and HFD + PS-MS. Importantly, transplantation of feces from HFD + PS-MS mice to microbiota-depleted HFD-fed mice resulted in a lower expression of mucus proteins, higher expression of inflammatory cytokines, and obesity outcomes, similar to the findings in HFD + PS-MS mice. CONCLUSIONS Our findings provide a new gut microbiota-driven mechanism for PS-MS-induced obesity in HFD-fed mice, suggesting the need to reevaluate the adverse health effects of MPs commonly found in daily life, particularly in susceptible populations. https://doi.org/10.1289/EHP13913.
Collapse
Affiliation(s)
- Zhian Zhai
- Department of Companion Animal Science, State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Ying Yang
- Department of Companion Animal Science, State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Sheng Chen
- State Key Lab of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Food Science and Nutrition, Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Zhenlong Wu
- Department of Companion Animal Science, State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| |
Collapse
|
10
|
Du Y, He C, An Y, Huang Y, Zhang H, Fu W, Wang M, Shan Z, Xie J, Yang Y, Zhao B. The Role of Short Chain Fatty Acids in Inflammation and Body Health. Int J Mol Sci 2024; 25:7379. [PMID: 39000498 PMCID: PMC11242198 DOI: 10.3390/ijms25137379] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Short chain fatty acids (SCFAs), mainly including acetate, propionate and butyrate, are produced by intestinal bacteria during the fermentation of partially digested and indigestible polysaccharides. SCFAs play an important role in regulating intestinal energy metabolism and maintaining the homeostasis of the intestinal environment and also play an important regulatory role in organs and tissues outside the gut. In recent years, many studies have shown that SCFAs can regulate inflammation and affect host health, and two main signaling mechanisms have also been identified: the activation of G-protein coupled receptors (GPCRs) and inhibition of histone deacetylase (HDAC). In addition, a growing body of evidence highlights the importance of every SCFA in influencing health maintenance and disease development. In this review, we summarized the recent advances concerning the biological properties of SCFAs and their signaling pathways in inflammation and body health. Hopefully, it can provide a systematic theoretical basis for the nutritional prevention and treatment of human diseases.
Collapse
Affiliation(s)
- Yuhang Du
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Changhao He
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yongcheng An
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yan Huang
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huilin Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wanxin Fu
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Menglu Wang
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ziyi Shan
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiamei Xie
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yang Yang
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
11
|
Luo J, Luo M, Kaminga AC, Wei J, Dai W, Peng Y, Zhao K, Duan Y, Xiao X, Ouyang S, Yao Z, Liu Y, Pan X. Integrative metabolomics highlights gut microbiota metabolites as novel NAFLD-related candidate biomarkers in children. Microbiol Spectr 2024; 12:e0523022. [PMID: 38445874 PMCID: PMC10986516 DOI: 10.1128/spectrum.05230-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 12/29/2023] [Indexed: 03/07/2024] Open
Abstract
Altered gut microbiota and metabolites are important for non-alcoholic fatty liver disease (NAFLD) in children. We aimed to comprehensively examine the effects of gut metabolites on NAFLD progression. We performed integrative metabolomics (untargeted discovery and targeted validation) analysis of non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), and obesity in children. Fecal samples were collected from 75 subjects in the discovery cohort (25 NAFL, 25 NASH, and 25 obese control children) and 145 subjects in an independent validation cohort (53 NAFL, 39 NASH, and 53 obese control children). Among 2,491 metabolites, untargeted metabolomics revealed a complete NAFLD metabolic map containing 318 increased and 123 decreased metabolites. Then, machine learning selected 65 important metabolites that can distinguish the severity of the NAFLD. Furthermore, precision-targeted metabolomics selected 5 novel gut metabolites from 20 typical metabolites. The functionality of candidate metabolites was validated in hepatocyte cell lines. In the end, this study annotated two novel elevated pathogenic metabolites (dodecanoic acid and creatinine) and a relationship between depleted protective gut microbiota (Butyricicoccus and Alistipes), increased inflammation (IL-1β), lipid metabolism (TG), and liver function (ALT and AST). This study demonstrates the role of novel gut metabolites (dodecanoic acid and creatinine), as the fatty acid metabolism regulator contributing to NAFLD development through its influence on inflammation and liver function. IMPORTANCE Altered gut microbiota and metabolites are a major cause of non-alcoholic fatty liver disease (NAFLD) in children. This study demonstrated a complete gut metabolic map of children with NAFLD, containing 318 increased and 123 decreased metabolites by untargeted metabolomic. Multiple validation approaches (machine learning and targeted metabolomic) selected five novel gut metabolites for targeted metabolomics, which can distinguish NAFLD status and severity. The gut microbiota (Butyricicoccus and Alistipes) and metabolites (creatinine and dodecanoic acid) were novel biomarkers associated with impaired liver function and inflammation and validated by experiments of hepatocyte cell lines. The data provide a better understanding of the importance of gut microbiota and metabolite alterations in NAFLD, which implies that the altered gut microbiota and metabolites may represent a potential target to prevent NAFLD development.
Collapse
Affiliation(s)
- Jiayou Luo
- Pediatrics Research Institute of Hunan Province, Hunan Children’s Hospital, Changsha, China
- Department of Maternal and Child Health, Xiangya School of Public Health, Central South University, Changsha, China
| | - Miyang Luo
- Department of Maternal and Child Health, Xiangya School of Public Health, Central South University, Changsha, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | | | - Jia Wei
- Department of Maternal and Child Health, Xiangya School of Public Health, Central South University, Changsha, China
| | - Wen Dai
- Department of Maternal and Child Health, Xiangya School of Public Health, Central South University, Changsha, China
| | - Yunlong Peng
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Kunyan Zhao
- School of Public Health, University of South China, Hengyang, China
| | - Yamei Duan
- Department of Maternal and Child Health, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xiang Xiao
- Department of Maternal and Child Health, Xiangya School of Public Health, Central South University, Changsha, China
| | - SiSi Ouyang
- Department of Maternal and Child Health, Xiangya School of Public Health, Central South University, Changsha, China
| | - Zhenzhen Yao
- Department of Maternal and Child Health, Xiangya School of Public Health, Central South University, Changsha, China
| | - Yixu Liu
- Department of Maternal and Child Health, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xiongfeng Pan
- Pediatrics Research Institute of Hunan Province, Hunan Children’s Hospital, Changsha, China
- Department of Maternal and Child Health, Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
12
|
Cai W, Qiu T, Hu W, Fang T. Changes in the intestinal microbiota of individuals with non-alcoholic fatty liver disease based on sequencing: An updated systematic review and meta-analysis. PLoS One 2024; 19:e0299946. [PMID: 38547205 PMCID: PMC10977702 DOI: 10.1371/journal.pone.0299946] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 02/20/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Alterations in the composition and abundance of the intestinal microbiota occur in non-alcoholic fatty liver disease (NAFLD). However, the results are inconsistent because of differences in the study design, subject area, and sequencing methodology. In this study, we compared the diversity and abundance of the intestinal microbiota of patients with NAFLD and healthy individuals through a systematic review and meta-analysis. METHODS Three databases (PubMed, EMBASE, and Cochrane Library) were searched from their inception to March 20, 2023. A meta-analysis was performed using Stata software to analyze variations in the richness and abundance of the intestinal microbiota in patients with NAFLD. The Newcastle-Ottawa Quality Assessment Scale (NOS) was used for quality assessment. RESULTS A total of 28 articles were included. Shannon diversity was reduced in patients with NAFLD (SMD = -0.24 (95% CI -0.43-0.05, I2 = 71.7%). The relative abundance of Ruminococcus, Faecalibacterium, and Coprococcus all decreased, with total SMDs of -0.96 (95% CI -1.29 to -0.63, I2 = 4.8%), -1.13 (95% CI -2.07 to -0.19, I2 = 80.5%), and -1.66 (95% CI -3.04 to -0.28, I2 = 91.5%). Escherichia was increased in individuals with NAFLD (SMD = 1.78, 95% CI 0.12 to 3.45, I2 = 94.4%). CONCLUSION Increasing the species diversity and altering the abundance of specific gut microbiota, including Coprococcus, Faecalibacterium, Ruminococcus, and Escherichia, may be beneficial for improving NAFLD.
Collapse
Affiliation(s)
- Wenpin Cai
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Ting Qiu
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Weitao Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
13
|
Tsai CC, Chiu MH, Kek HP, Yang MC, Su YT, Liu HK, Wu MS, Yeh YT. The Reduced Gut Lachnospira Species Is Linked to Liver Enzyme Elevation and Insulin Resistance in Pediatric Fatty Liver Disease. Int J Mol Sci 2024; 25:3640. [PMID: 38612453 PMCID: PMC11011648 DOI: 10.3390/ijms25073640] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
The objective of this study was to investigate gut dysbiosis and its metabolic and inflammatory implications in pediatric metabolic dysfunction-associated fatty liver disease (MAFLD). This study included 105 children and utilized anthropometric measurements, blood tests, the Ultrasound Fatty Liver Index, and fecal DNA sequencing to assess the relationship between gut microbiota and pediatric MAFLD. Notable decreases in Lachnospira spp., Faecalibacterium spp., Oscillospira spp., and Akkermansia spp. were found in the MAFLD group. Lachnospira spp. was particularly reduced in children with MAFLD and hepatitis compared to controls. Both MAFLD groups showed a reduction in flavone and flavonol biosynthesis sequences. Lachnospira spp. correlated positively with flavone and flavonol biosynthesis and negatively with insulin levels and insulin resistance. Body weight, body mass index (BMI), and total cholesterol levels were inversely correlated with flavone and flavonol biosynthesis. Reduced Lachnospira spp. in children with MAFLD may exacerbate insulin resistance and inflammation through reduced flavone and flavonol biosynthesis, offering potential therapeutic targets.
Collapse
Affiliation(s)
- Ching-Chung Tsai
- Department of Pediatrics, E-Da Hospital, I-Shou University, No. 1, Yi-Da Road, Yan-Chao District, Kaohsiung City 82445, Taiwan; (C.-C.T.); (H.-P.K.); (M.-C.Y.); (Y.-T.S.); (H.-K.L.)
- School of Medicine for International Students, College of Medicine, I-Shou University, No. 8, Yi-Da Road, Yan-Chao District, Kaohsiung City 82445, Taiwan
| | - Min-Hsi Chiu
- Aging and Disease Prevention Research Center, Fooyin University, No. 151, Jinxue Road, Daliao District, Kaohsiung City 83102, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Fooyin University, No. 151, Jinxue Road, Daliao District, Kaohsiung City 83102, Taiwan
| | - Ho-Poh Kek
- Department of Pediatrics, E-Da Hospital, I-Shou University, No. 1, Yi-Da Road, Yan-Chao District, Kaohsiung City 82445, Taiwan; (C.-C.T.); (H.-P.K.); (M.-C.Y.); (Y.-T.S.); (H.-K.L.)
| | - Ming-Chun Yang
- Department of Pediatrics, E-Da Hospital, I-Shou University, No. 1, Yi-Da Road, Yan-Chao District, Kaohsiung City 82445, Taiwan; (C.-C.T.); (H.-P.K.); (M.-C.Y.); (Y.-T.S.); (H.-K.L.)
- School of Medicine, College of Medicine, I-Shou University, No. 8, Yi-Da Road, Yan-Chao District, Kaohsiung City 82445, Taiwan
| | - Yu-Tsun Su
- Department of Pediatrics, E-Da Hospital, I-Shou University, No. 1, Yi-Da Road, Yan-Chao District, Kaohsiung City 82445, Taiwan; (C.-C.T.); (H.-P.K.); (M.-C.Y.); (Y.-T.S.); (H.-K.L.)
- School of Medicine for International Students, College of Medicine, I-Shou University, No. 8, Yi-Da Road, Yan-Chao District, Kaohsiung City 82445, Taiwan
| | - Hsien-Kuan Liu
- Department of Pediatrics, E-Da Hospital, I-Shou University, No. 1, Yi-Da Road, Yan-Chao District, Kaohsiung City 82445, Taiwan; (C.-C.T.); (H.-P.K.); (M.-C.Y.); (Y.-T.S.); (H.-K.L.)
- School of Medicine, College of Medicine, I-Shou University, No. 8, Yi-Da Road, Yan-Chao District, Kaohsiung City 82445, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, No. 7, Zhongshan S. Road, Zhongzheng District, Taipei City 100225, Taiwan;
| | - Yao-Tsung Yeh
- Aging and Disease Prevention Research Center, Fooyin University, No. 151, Jinxue Road, Daliao District, Kaohsiung City 83102, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Fooyin University, No. 151, Jinxue Road, Daliao District, Kaohsiung City 83102, Taiwan
| |
Collapse
|
14
|
Zeng F, Su X, Liang X, Liao M, Zhong H, Xu J, Gou W, Zhang X, Shen L, Zheng JS, Chen YM. Gut microbiome features and metabolites in non-alcoholic fatty liver disease among community-dwelling middle-aged and older adults. BMC Med 2024; 22:104. [PMID: 38454425 PMCID: PMC10921631 DOI: 10.1186/s12916-024-03317-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND The specific microbiota and associated metabolites linked to non-alcoholic fatty liver disease (NAFLD) are still controversial. Thus, we aimed to understand how the core gut microbiota and metabolites impact NAFLD. METHODS The data for the discovery cohort were collected from the Guangzhou Nutrition and Health Study (GNHS) follow-up conducted between 2014 and 2018. We collected 272 metadata points from 1546 individuals. The metadata were input into four interpretable machine learning models to identify important gut microbiota associated with NAFLD. These models were subsequently applied to two validation cohorts [the internal validation cohort (n = 377), and the prospective validation cohort (n = 749)] to assess generalizability. We constructed an individual microbiome risk score (MRS) based on the identified gut microbiota and conducted animal faecal microbiome transplantation experiment using faecal samples from individuals with different levels of MRS to determine the relationship between MRS and NAFLD. Additionally, we conducted targeted metabolomic sequencing of faecal samples to analyse potential metabolites. RESULTS Among the four machine learning models used, the lightGBM algorithm achieved the best performance. A total of 12 taxa-related features of the microbiota were selected by the lightGBM algorithm and further used to calculate the MRS. Increased MRS was positively associated with the presence of NAFLD, with odds ratio (OR) of 1.86 (1.72, 2.02) per 1-unit increase in MRS. An elevated abundance of the faecal microbiota (f__veillonellaceae) was associated with increased NAFLD risk, whereas f__rikenellaceae, f__barnesiellaceae, and s__adolescentis were associated with a decreased presence of NAFLD. Higher levels of specific gut microbiota-derived metabolites of bile acids (taurocholic acid) might be positively associated with both a higher MRS and NAFLD risk. FMT in mice further confirmed a causal association between a higher MRS and the development of NAFLD. CONCLUSIONS We confirmed that an alteration in the composition of the core gut microbiota might be biologically relevant to NAFLD development. Our work demonstrated the role of the microbiota in the development of NAFLD.
Collapse
Affiliation(s)
- Fangfang Zeng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Road West, Guangzhou, 510632, China.
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou, 510275, China.
| | - Xin Su
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Road West, Guangzhou, 510632, China
| | - Xinxiu Liang
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, 310030, China
| | - Minqi Liao
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Haili Zhong
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Jinjian Xu
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Wanglong Gou
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, 310030, China
| | - Xiangzhou Zhang
- Big Data Decision Institute, Jinan University, No.601 Huangpu Road West, Guangzhou, 510632, China
| | - Luqi Shen
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, 310030, China
| | - Ju-Sheng Zheng
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, 310030, China.
| | - Yu-Ming Chen
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou, 510275, China.
| |
Collapse
|
15
|
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
|
16
|
Hizo GH, Rampelotto PH. The Impact of Probiotic Bifidobacterium on Liver Diseases and the Microbiota. Life (Basel) 2024; 14:239. [PMID: 38398748 PMCID: PMC10890151 DOI: 10.3390/life14020239] [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: 12/26/2023] [Revised: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Recent studies have shown the promising potential of probiotics, especially the bacterial genus Bifidobacterium, in the treatment of liver diseases. In this work, a systematic review was conducted, with a focus on studies that employed advanced Next Generation Sequencing (NGS) technologies to explore the potential of Bifidobacterium as a probiotic for treating liver pathologies such as Non-Alcoholic Fatty Liver Disease (NAFLD), Non-Alcoholic Steatohepatitis (NASH), Alcoholic Liver Disease (ALD), Cirrhosis, and Hepatocelullar Carcinoma (HCC) and its impact on the microbiota. Our results indicate that Bifidobacterium is a safe and effective probiotic for treating liver lesions. It successfully restored balance to the intestinal microbiota and improved biochemical and clinical parameters in NAFLD, ALD, and Cirrhosis. No significant adverse effects were identified. While more research is needed to establish its efficacy in treating NASH and HCC, the evidence suggests that Bifidobacterium is a promising probiotic for managing liver lesions.
Collapse
Affiliation(s)
- Gabriel Henrique Hizo
- Graduate Program in Gastroenterology and Hepatology Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-907, Brazil
| |
Collapse
|
17
|
Zhang J, Shi M, Zhao C, Liang G, Li C, Ge X, Pei C, Kong Y, Li D, Yang W, Cao B, Fu L, Yan Y, Wu J, Zhou J, Fang Y, Meng X, Li Y, Wang L. Role of intestinal flora in the development of nonalcoholic fatty liver disease in children. Microbiol Spectr 2024; 12:e0100623. [PMID: 38189294 PMCID: PMC10846053 DOI: 10.1128/spectrum.01006-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 11/11/2023] [Indexed: 01/09/2024] Open
Abstract
In China, 45% of adolescents with obesity develop fatty liver disease, a condition that increases the long-term risk of developing cirrhosis and liver cancer. Although the factors triggering nonalcoholic fatty liver disease (NAFLD) vary in children, the composition of intestinal microflora has been found to play an increasingly important role. However, evidence is limited on the prevalence of nonalcoholic fatty liver (NAFL) and nonalcoholic steatohepatitis (NASH) in Chinese children. Therefore, this study aimed to evaluate the fecal microbiome of Chinese children with NAFLD and further analyze the potential of flora in regulating NAFLD-related symptoms and metabolic functions. Specifically, the study applied a 16S rRNA and metagenomic sequencing to the fecal samples of pediatric patients with NAFLD, NASH, and NAFL, as well as healthy controls, to explore the correlation among NAFLD-related indexes, metabolic pathways, and gut flora. The findings showed that some fecal microbiota had a negative correlation with body mass index, and various NAFLD-related bacteria, including Lachnoclostridium, Escherichia-Shigella, and Faecalibacterium prausnitzii, were detected. Consequently, the study concluded that the variation in gut microbiota might be more important in improving NAFLD/NASH compared with single species, providing a microbiota diagnostic profile of NAFLD/NASH.IMPORTANCEThis study aims to characterize the gut microbiota in Chinese children with nonalcoholic fatty liver disease (NAFLD) through 16S rRNA and metagenomic sequencing. The results highlight the association between fecal microbiota and NAFLD in Chinese children, demonstrating distinct characteristics compared to adults and children from other countries. Based on the sequencing data from our cohort's fecal samples, we propose a microbiota model with a high area under the curve for distinguishing between NAFLD and healthy individuals. Furthermore, our follow-up study reveals that changes in the relative abundance of microbial biomarkers in this model are consistent with variations in patients' body mass index. These findings suggest the potential utility of the microbiota model and microbial biomarkers for diagnosing and treating NAFLD in children.
Collapse
Affiliation(s)
- Jing Zhang
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Mengxuan Shi
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Chunna Zhao
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Guangcai Liang
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Department of Scientific Research, Microvita Medical Technology Co., Beijing, China
| | - Chuan Li
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Department of Scientific Research, Microvita Medical Technology Co., Beijing, China
| | - Xiaomeng Ge
- Microbial Resources and Big Data Center, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Caixia Pei
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yawei Kong
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Dongdan Li
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Wenli Yang
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Bingyan Cao
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Libing Fu
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Yinkun Yan
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Jie Wu
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Jin Zhou
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Yongli Fang
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Xi Meng
- National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Yong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liming Wang
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
18
|
Gilley SP, Zarate MA, Zheng L, Jambal P, Yazza DN, Chintapalli SV, MacLean PS, Wright CJ, Rozance PJ, Shankar K. Metabolic and fecal microbial changes in adult fetal growth restricted mice. Pediatr Res 2024; 95:647-659. [PMID: 37935884 PMCID: PMC10899111 DOI: 10.1038/s41390-023-02869-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Fetal growth restriction (FGR) increases risk for development of obesity and type 2 diabetes. Using a mouse model of FGR, we tested whether metabolic outcomes were exacerbated by high-fat diet challenge or associated with fecal microbial taxa. METHODS FGR was induced by maternal calorie restriction from gestation day 9 to 19. Control and FGR offspring were weaned to control (CON) or 45% fat diet (HFD). At age 16 weeks, offspring underwent intraperitoneal glucose tolerance testing, quantitative MRI body composition assessment, and energy balance studies. Total microbial DNA was used for amplification of the V4 variable region of the 16 S rRNA gene. Multivariable associations between groups and genera abundance were assessed using MaAsLin2. RESULTS Adult male FGR mice fed HFD gained weight faster and had impaired glucose tolerance compared to control HFD males, without differences among females. Irrespective of weaning diet, adult FGR males had depletion of Akkermansia, a mucin-residing genus known to be associated with weight gain and glucose handling. FGR females had diminished Bifidobacterium. Metabolic changes in FGR offspring were associated with persistent gut microbial changes. CONCLUSION FGR results in persistent gut microbial dysbiosis that may be a therapeutic target to improve metabolic outcomes. IMPACT Fetal growth restriction increases risk for metabolic syndrome later in life, especially if followed by rapid postnatal weight gain. We report that a high fat diet impacts weight and glucose handling in a mouse model of fetal growth restriction in a sexually dimorphic manner. Adult growth-restricted offspring had persistent changes in fecal microbial taxa known to be associated with weight, glucose homeostasis, and bile acid metabolism, particularly Akkermansia, Bilophilia and Bifidobacteria. The gut microbiome may represent a therapeutic target to improve long-term metabolic outcomes related to fetal growth restriction.
Collapse
Affiliation(s)
- Stephanie P Gilley
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Miguel A Zarate
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lijun Zheng
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Purevsuren Jambal
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Deaunabah N Yazza
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Paul S MacLean
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Clyde J Wright
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Paul J Rozance
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
19
|
Mai H, Yang X, Xie Y, Zhou J, Wang Q, Wei Y, Yang Y, Lu D, Ye L, Cui P, Liang H, Huang J. The role of gut microbiota in the occurrence and progression of non-alcoholic fatty liver disease. Front Microbiol 2024; 14:1257903. [PMID: 38249477 PMCID: PMC10797006 DOI: 10.3389/fmicb.2023.1257903] [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: 07/13/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is the most prevalent cause of chronic liver disease worldwide, and gut microbes are associated with the development and progression of NAFLD. Despite numerous studies exploring the changes in gut microbes associated with NAFLD, there was no consistent pattern of changes. Method We retrieved studies on the human fecal microbiota sequenced by 16S rRNA gene amplification associated with NAFLD from the NCBI database up to April 2023, and re-analyzed them using bioinformatic methods. Results We finally screened 12 relevant studies related to NAFLD, which included a total of 1,189 study subjects (NAFLD, n = 654; healthy control, n = 398; obesity, n = 137). Our results revealed a significant decrease in gut microbial diversity with the occurrence and progression of NAFLD (SMD = -0.32; 95% CI -0.42 to -0.21; p < 0.001). Alpha diversity and the increased abundance of several crucial genera, including Desulfovibrio, Negativibacillus, and Prevotella, can serve as an indication of their predictive risk ability for the occurrence and progression of NAFLD (all AUC > 0.7). The occurrence and progression of NAFLD are significantly associated with higher levels of LPS biosynthesis, tryptophan metabolism, glutathione metabolism, and lipid metabolism. Conclusion This study elucidated gut microbes relevance to disease development and identified potential risk-associated microbes and functional pathways associated with NAFLD occurrence and progression.
Collapse
Affiliation(s)
- Huanzhuo Mai
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Xing Yang
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Yulan Xie
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Jie Zhou
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Qing Wang
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Yiru Wei
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Yuecong Yang
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Dongjia Lu
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Li Ye
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Nanning, China
| | - Ping Cui
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Nanning, China
- Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Hao Liang
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Nanning, China
- Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Jiegang Huang
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| |
Collapse
|
20
|
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
|
21
|
Ji J, Sun J, Li J, Xie J, Xi B, Zhao M. Altered gut microbiome associated with metabolic-associated fatty liver disease in Chinese children. Clin Nutr 2024; 43:187-196. [PMID: 38070210 DOI: 10.1016/j.clnu.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND & AIM Limited studies have investigated the association between gut microbiota and metabolic dysfunction-associated fatty liver disease (MAFLD) in children and adolescents. We aimed to identify differences in gut microbiota composition and diversity between children with MAFLD and healthy counterparts. METHODS Data were collected from a nested case-control study (October to December, 2021) of the "Huantai Childhood Cardiovascular Health Cohort Study" in Huantai County, Zibo City, China. The study included 52 children aged 5-11 years with new-onset MAFLD and 52 healthy children matched by age and sex. Stool samples were collected and analyzed using 16S rRNA gene sequencing. Shannon index and Chao index were used to assess the α diversity of gut microbiota and Principal coordinates analysis (PCoA) was performed to evaluate β diversity between the two groups. The differences in the relative abundance of gut microbiota between MAFLD group and control group were compared by the Wilcoxon rank-sum test after false discovery rate (FDR) correction. Additionally, the gut-microbial metabolic pathways were identified using the phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt). RESULTS We found that children with MAFLD had significant different gut microbiota composition and reduced α diversity compared with the control group. PCoA showed that the two groups can be significantly distinguished based on the unweighted unifrac distance algorithm. Gut microbiota at the phylum level such as Verrucomicrobia and Desulfobacterial, genus level such as Blautia, Lachnospiraceae_NK4A136_group, Coprococcus, Erysipelotrichaceae_UCG-003, UCG-002 and Akkermansia, and species level such as Bifidobacterium_longum abundances were significantly decreased in children with MAFLD compared with that in children without MAFLD. Notably, the abundance of these bacteria were found to be associated with HDL-C, SBP, DBP, WC, BMI, etc. In addition, our analysis of gut-microbial metabolic pathways identified differences in carbohydrate transport and metabolism, as well as amino acid transport and metabolism between the two groups. CONCLUSION Significant differences in gut microbiota composition are observed between children with and without MAFLD, which indicate that gut microbiota may be a potential contributor to the development of MAFLD in childhood.
Collapse
Affiliation(s)
- Jing Ji
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jiahong Sun
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, China
| | - Juan Li
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jintang Xie
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Bo Xi
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Min Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
22
|
Su X, Chen S, Liu J, Feng Y, Han E, Hao X, Liao M, Cai J, Zhang S, Niu J, He S, Huang S, Lo K, Zeng F. Composition of gut microbiota and non-alcoholic fatty liver disease: A systematic review and meta-analysis. Obes Rev 2024; 25:e13646. [PMID: 37813400 DOI: 10.1111/obr.13646] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 08/03/2023] [Accepted: 09/09/2023] [Indexed: 10/11/2023]
Abstract
The present systematic review and meta-analysis aimed to summarize the associations between gut microbiota composition and non-alcoholic fatty liver disease. To compare the differences between individuals with or without NAFLD, the standardized mean difference and 95% confidence interval were computed for each α-diversity index and relative abundance of gut microbes. The β-diversity indices were summarized in a qualitative manner. A total of 54 studies with 8894 participants were included. Overall, patients with NAFLD had moderate reduction in α-diversity indices including Shannon (SMD = -0.36, 95% CI = [-0.53, -0.19], p < 0.001) and Chao 1 (SMD = -0.42, 95% CI = [-0.68, -0.17], p = 0.001), but no significant differences were found for Simpson, observed species, phylogenetic diversity, richness, abundance-based coverage estimator, and evenness (p ranged from 0.081 to 0.953). Over 75% of the included studies reported significant differences in β-diversity. Although there was substantial interstudy heterogeneity, especially for analyses at the phylum, class, and family levels, the majority of the included studies showed alterations in the depletion of anti-inflammatory microbes (i.e., Ruminococcaceae and Coprococcus) and the enrichment of proinflammatory microbes (i.e., Fusobacterium and Escherichia) in patients with NAFLD. Perturbations in gut microbiota were associated with NAFLD, commonly reflected by a reduction in beneficial species and an increase in the pathogenic species.
Collapse
Affiliation(s)
- Xin Su
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Shiyun Chen
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jiazi Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yonghui Feng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Eerdun Han
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiaolei Hao
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Minqi Liao
- Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Epidemiology, Neuherberg, PR, Germany
| | - Jun Cai
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Shiwen Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jianxiang Niu
- General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Shihua He
- Department of Infectious Disease, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Shaofen Huang
- Shenzhen Qianhai Shekou Free Zone Hospital, Shenzhen, China
| | - Kenneth Lo
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Research Institute for Future Food, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Fangfang Zeng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
23
|
Banerjee G, Papri SR, Satapathy SK, Banerjee P. Akkermansia muciniphila - A Potential Next-generation Probiotic for Non-alcoholic Fatty Liver Disease. Curr Pharm Biotechnol 2024; 25:426-433. [PMID: 37724669 DOI: 10.2174/1389201025666230915103052] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/20/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a spectrum of liver conditions, and its growing prevalence is a serious concern worldwide, especially in Western countries. Researchers have pointed out several genetic mutations associated with NAFLD; however, the imbalance of the gut microbial community also plays a critical role in the progression of NAFLD. Due to the lack of approved medicine, probiotics gain special attention in controlling metabolic disorders like NAFLD. Among these probiotics, Akkermansia muciniphila (a member of natural gut microflora) is considered one of the most efficient and important bacterium in maintaining gut health, energy homeostasis, and lipid metabolism. In this perspective, we discussed the probable molecular mechanism of A. muciniphila in controlling the progression of NAFLD and restoring liver health. The therapeutic potential of A. muciniphila in NAFLD has been tested primarily on animal models, and thus, more randomized human trials should be conducted to prove its efficacy.
Collapse
Affiliation(s)
- Goutam Banerjee
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Suraya R Papri
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Sanjaya K Satapathy
- 2Department of Medicine, Northwell Health Center for Liver Disease & Transplantation, North Shore, University Hospital/Northwell Health, 400 Community Drive, Manhasset, NY 11030, USA
| | - Pratik Banerjee
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| |
Collapse
|
24
|
Yang Z, Yang M, Deehan EC, Cai C, Madsen KL, Wine E, Li G, Li J, Liu J, Zhang Z. Dietary fiber for the prevention of childhood obesity: a focus on the involvement of the gut microbiota. Gut Microbes 2024; 16:2387796. [PMID: 39163556 PMCID: PMC11340751 DOI: 10.1080/19490976.2024.2387796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/22/2024] Open
Abstract
Given the worldwide epidemic of overweight and obesity among children, evidence-based dietary recommendations are fundamentally important for obesity prevention. Although the significance of the human gut microbiome in shaping the physiological effects of diet and obesity has been widely recognized, nutritional therapeutics for the mitigation of pediatric obesity globally are only just starting to leverage advancements in the nutritional microbiology field. In this review, we extracted data from PubMed, EMBASE, Scopus, Web of Science, Google Scholar, CNKI, Cochrane Library and Wiley online library that focuses on the characterization of gut microbiota (including bacteria, fungi, viruses, and archaea) in children with obesity. We further review host-microbe interactions as mechanisms mediating the physiological effects of dietary fibers and how fibers alter the gut microbiota in children with obesity. Contemporary nutritional recommendations for the prevention of pediatric obesity are also discussed from a gut microbiological perspective. Finally, we propose an experimental framework for integrating gut microbiota into nutritional interventions for children with obesity and provide recommendations for the design of future studies on precision nutrition for pediatric obesity.
Collapse
Affiliation(s)
- Zhongmin Yang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
| | - Mingyue Yang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
| | - Edward C. Deehan
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska, Lincoln, NE, USA
| | - Chenxi Cai
- School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Karen L. Madsen
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Eytan Wine
- Division of Pediatric Gastroenterology, Departments of Pediatrics and Physiology, University of Alberta, Edmonton, AB, Canada
| | - Guiling Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian, China
| | - Jian Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian, China
| | - Jingwen Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
| | - Zhengxiao Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian, China
| |
Collapse
|
25
|
Hizo GH, Rampelotto PH. The Role of Bifidobacterium in Liver Diseases: A Systematic Review of Next-Generation Sequencing Studies. Microorganisms 2023; 11:2999. [PMID: 38138143 PMCID: PMC10745637 DOI: 10.3390/microorganisms11122999] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
The physiopathology of liver diseases is complex and can be caused by various factors. Bifidobacterium is a bacterial genus commonly found in the human gut microbiome and has been shown to influence the development of different stages of liver diseases significantly. This study investigated the relationship between the Bifidobacterium genus and liver injury. In this work, we performed a systematic review in major databases using the key terms "Bifidobacterium", "ALD", "NAFLD", "NASH", "cirrhosis", and "HCC" to achieve our purpose. In total, 31 articles were selected for analysis. In particular, we focused on studies that used next-generation sequencing (NGS) technologies. The studies focused on assessing Bifidobacterium levels in the diseases and interventional aimed at examining the therapeutic potential of Bifidobacterium in the mentioned conditions. Overall, the abundance of Bifidobacterium was reduced in hepatic pathologies. Low levels of Bifidobacterium were associated with harmful biochemical and physiological parameters, as well as an adverse clinical outcome. However, interventional studies using different drugs and treatments were able to increase the abundance of the genus and improve clinical outcomes. These results strongly support the hypothesis that changes in the abundance of Bifidobacterium significantly influence both the pathophysiology of hepatic diseases and the related clinical outcomes. In addition, our critical assessment of the NGS methods and related statistical analyses employed in each study highlights concerns with the methods used to define the differential abundance of Bifidobacterium, including potential biases and the omission of relevant information.
Collapse
Affiliation(s)
- Gabriel Henrique Hizo
- Graduate Program in Gastroenterology and Hepatology Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-907, Brazil
| |
Collapse
|
26
|
Li J, Ma J, Wang W, Du H, Tang S, Li Y, Zhu W, Zhang R, Wan J. Alterations of ileal mucosa-associated microbiota in hypercholesterolemia patients. Heliyon 2023; 9:e22116. [PMID: 38076161 PMCID: PMC10709208 DOI: 10.1016/j.heliyon.2023.e22116] [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: 06/17/2023] [Revised: 09/11/2023] [Accepted: 11/05/2023] [Indexed: 01/03/2025] Open
Abstract
Many metabolic diseases have been demonstrated to be associated with changes in the microbiome. However, no studies have yet been conducted to examine the characteristics of the mucosal microbiota of patients with hypercholesterolemia. We aimed to examine mucosa-associated microbiota in subjects with hypercholesterolemia. We conducted a case-control study, in which ileal mucosal samples were collected from 13 hypercholesterolemia patients and 13 controls for 16S rRNA gene sequencing. There were differences in the composition of ileal mucosal microbiota based on beta diversity between the hypercholesterolemia and control groups (P < 0.05). Compared with the control group, the phylum Bacteroidetes and the genera Bacteroides, Butyricicoccus, Parasutterella, Candidatus_Soleaferrea, and norank_f__norank_o__Izemoplasmatales were less abundant in the hypercholesterolemia group (P < 0.05), while the genus Anaerovibrio was enriched in the hypercholesterolemia group (P < 0.05). The relative abundance of Bacteroides was negatively correlated with total cholesterol and low-density lipoprotein cholesterol (P < 0.01). The relative abundance of Coprococcus was negatively correlated with triglycerides and body mass index (all P < 0.05). PICRUSt functional prediction analysis showed that pathways related to Glycerophospholipid metabolism, ABC transporters, Phosphotransferase system, and Biofilm formation - Escherichia coli, and infectious diseases of pathogenic Escherichia coli were enriched in the hypercholesterolemia group. This work suggests a potential role of ileal mucosal microbiota in the development of hypercholesterolemia.
Collapse
Affiliation(s)
- Jia Li
- Department of Gastroenterology, The 983rd Hospital of Joint Logistic Support Force of PLA, No. 60, Huangwei Road, Hebei District, Tianjin 300142, China
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Jinxia Ma
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Weihua Wang
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Haitao Du
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Shuai Tang
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Yi Li
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Wenya Zhu
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Ru Zhang
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Jun Wan
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| |
Collapse
|
27
|
Spiezia C, Di Rosa C, Fintini D, Ferrara P, De Gara L, Khazrai YM. Nutritional Approaches in Children with Overweight or Obesity and Hepatic Steatosis. Nutrients 2023; 15:nu15112435. [PMID: 37299398 DOI: 10.3390/nu15112435] [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: 04/17/2023] [Revised: 05/11/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Childhood obesity is a global public health problem. Worldwide, 41 million children under 5 years and 340 million children and adolescents between 5 and 19 years are overweight. In addition, the recent COVID-19 epidemic has further amplified this social phenomenon. Obesity is a condition associated with various comorbidities, such as nonalcoholic fatty liver disease (NAFLD). The pathophysiology of NAFLD in obesity is intricate and involves the interaction and dysregulation of several mechanisms, such as insulin resistance, cytokine signaling, and alteration of the gut microbiota. NAFLD is defined as the presence of hepatic steatosis in more than 5% of hepatocytes, evaluated by histological analysis. It can evolve from hepatic steatosis to steatohepatitis, fibrosis, cirrhosis, hepatocellular carcinoma, and end-stage liver failure. Body weight reduction through lifestyle modification remains the first-line intervention for the management of pediatric NAFLD. Indeed, studies suggest that diets low in fat and sugar and conversely rich in dietary fibers promote the improvement of metabolic parameters. This review aims to evaluate the existing relationship between obesity and NAFLD in the pediatric population and to assess the dietary patterns and nutritional supplementations that can be recommended to prevent and manage obesity and its comorbidities.
Collapse
Affiliation(s)
- Chiara Spiezia
- Research Unit of Food Science and Human Nutrition, Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128 Roma, Italy
| | - Claudia Di Rosa
- Research Unit of Food Science and Human Nutrition, Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128 Roma, Italy
| | - Danilo Fintini
- Endocrinology and Diabetology Unit, Bambino Gesù Children's Hospital, IRCCS L.go S.Onofrio, 4-00165 Roma, Italy
| | - Pietro Ferrara
- Operative Research Unit of Pediatrics, Department of Medicine and Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200-00128 Roma, Italy
| | - Laura De Gara
- Research Unit of Food Science and Human Nutrition, Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128 Roma, Italy
| | - Yeganeh Manon Khazrai
- Research Unit of Food Science and Human Nutrition, Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128 Roma, Italy
- Operative Research Unit of Nutrition and Prevention, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200-00128 Roma, Italy
| |
Collapse
|
28
|
Saenz M, McDonough JC, Bloom-Saldana E, Irimia JM, Cauble EL, Castillo A, Fueger PT, Treviño LS. Longitudinal analysis of a dietary mouse model of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.19.540989. [PMID: 37293034 PMCID: PMC10245692 DOI: 10.1101/2023.05.19.540989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD), and resultant non-alcoholic steatohepatitis (NASH), incidence and prevalence are rising globally due to increasing rates of obesity and diabetes. Currently, there are no approved pharmacological treatments for NAFLD, highlighting a need for additional mechanistic studies to develop prevention and/or therapeutic strategies. Diet-induced preclinical models of NAFLD can be used to examine the dynamic changes that occur during NAFLD development and progression throughout the lifespan. To date, most studies utilizing such models have focused exclusively on terminal time points and have likely missed critical early and late changes that are important for NAFLD progression (i.e, worsening). We performed a longitudinal analysis of histopathological, biochemical, transcriptomic, and microbiome changes that occurred in adult male mice fed either a control diet or a NASH-promoting diet (high in fat, fructose, and cholesterol) for up to 30 weeks. We observed progressive development of NAFLD in mice fed the NASH diet compared to the control diet. Differential expression of immune-related genes was observed at an early stage of diet-induced NAFLD development (10 weeks) and persisted into the later stages of the disease (20 and 30 weeks). Differential expression of xenobiotic metabolism related genes was observed at the late stage of diet-induced NAFLD development (30 weeks). Microbiome analysis revealed an increased abundance of Bacteroides at an early stage (10 weeks) that persisted into the later stages of the disease (20 and 30 weeks). These data provide insight into the progressive changes that occur during NAFLD/NASH development and progression in the context of a typical Western diet. Furthermore, these data are consistent with what has been reported in patients with NAFLD/NASH, supporting the preclinical use of this diet-induced model for development of strategies to prevent or treat the disease.
Collapse
Affiliation(s)
- Marissa Saenz
- Center for Comparative Medicine, City of Hope, Duarte, CA
- Department of Molecular and Cellular Endocrinology, City of Hope, Duarte, CA
| | - Jillian C. McDonough
- Division of Health Inequities, Department of Population Sciences, City of Hope, Duarte, CA
| | - Elizabeth Bloom-Saldana
- Department of Molecular and Cellular Endocrinology, City of Hope, Duarte, CA
- Comprehensive Metabolic Phenotyping Core, Beckman Research Institute, City of Hope, Duarte, CA
| | - Jose M. Irimia
- Department of Molecular and Cellular Endocrinology, City of Hope, Duarte, CA
- Comprehensive Metabolic Phenotyping Core, Beckman Research Institute, City of Hope, Duarte, CA
| | - Emily L. Cauble
- Division of Health Inequities, Department of Population Sciences, City of Hope, Duarte, CA
| | - Ashly Castillo
- Eugene and Ruth Roberts Summer Student Academy, Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA
| | - Patrick T. Fueger
- Department of Molecular and Cellular Endocrinology, City of Hope, Duarte, CA
- Comprehensive Metabolic Phenotyping Core, Beckman Research Institute, City of Hope, Duarte, CA
| | - Lindsey S. Treviño
- Division of Health Inequities, Department of Population Sciences, City of Hope, Duarte, CA
- Cancer Control and Population Sciences, Comprehensive Cancer Center, City of Hope, Duarte, CA
| |
Collapse
|
29
|
Rodpai R, Sanpool O, Janwan P, Boonroumkaew P, Sadaow L, Thanchomnang T, Intapan PM, Maleewong W. Gut microbiota diversity in human strongyloidiasis differs little in two different regions in endemic areas of Thailand. PLoS One 2022; 17:e0279766. [PMID: 36584127 PMCID: PMC9803247 DOI: 10.1371/journal.pone.0279766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Human gastrointestinal helminthic infections have a direct and/or indirect effect on the composition of the host gut microbial flora. Here, we investigated the effect of infection with a soil-transmitted intestinal nematode, Strongyloides stercoralis, on the gut microbiota of the human host. We also investigated whether composition of the microbiota in infected persons might vary across endemic regions. Fecal samples were obtained from volunteers from two areas endemic for strongyloidiasis, Khon Kaen Province in northeastern Thailand and Nakhon Si Thammarat Province in southern Thailand. Samples from Khon Kaen were from infected (SsNE) and uninfected (NegNE) individuals. Similarly, samples from the latter province were from infected (SsST) and uninfected (NegST) individuals. DNA sequences of the V3-V4 regions of the bacterial 16S rRNA gene were obtained from the fecal samples. No statistical difference in alpha diversity between groups in terms of richness or diversity were found. Statistical difference in beta diversity was observed only between NegNE and NegST. Some significant differences in species abundance were noted between geographical isolates. The SsNE group had a higher abundance of Tetragenococcus holophilus than did the SsST group, whereas Bradyrhizobium sp. was less abundant in the SsNE than the SsST group. For the uninfected groups, the NegNE had a higher abundance of T. holophilus than the NegST group. Our data showed that S. stercoralis infection leads to only minor alterations in the relative abundance of individual bacterial species in the human gut: no detectable effect was observed on community structure and diversity.
Collapse
Affiliation(s)
- Rutchanee Rodpai
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Oranuch Sanpool
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Penchom Janwan
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
| | - Patcharaporn Boonroumkaew
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Lakkhana Sadaow
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Tongjit Thanchomnang
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Faculty of Medicine, Mahasarakham University, Mahasarakham, Thailand
| | - Pewpan M. Intapan
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Wanchai Maleewong
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen, Thailand
- * E-mail:
| |
Collapse
|
30
|
Liang T, Li D, Zunong J, Li M, Amaerjiang N, Xiao H, Khattab NM, Vermund SH, Hu Y. Interplay of Lymphocytes with the Intestinal Microbiota in Children with Nonalcoholic Fatty Liver Disease. Nutrients 2022; 14:nu14214641. [PMID: 36364902 PMCID: PMC9657134 DOI: 10.3390/nu14214641] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/24/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Abnormally high lymphocyte counts are seen in persons with nonalcoholic fatty liver disease (NAFLD). Gut microbiota dysbiosis is a risk factor for NAFLD. We assessed the gut microbiota of 63 healthy children and 63 children with NAFLD using 16S rRNA gene and metagenomic sequencing to explore the relationships. Compared with healthy children (HC group), the Bacteroidetes, Verrucomicrobia, and Akkermansia were less abundant, while the Actinobacteria were more abundant in children with NAFLD (FLD group). To understand the effect of lymphocytes on the gut microbiota of children with NAFLD, we compared the microbiota of 41 children with NAFLD and high numbers of lymphocytes (FLD_HL group) and 22 children with NAFLD and low numbers of lymphocytes (FLD_LL group). The abundances of Bacteroidetes, Verrucobacterium, and Akkermansia increased and Actinobacteria decreased in the FLD_LL group compared to the FLD_HL group. Akkermansia was negatively correlated with lymphocyte count. NAFLD may disturb the gut microbiota in children through reducing the abundance of Akkermansia and increasing the abundance of proinflammatory bacteria, such as Escherichia-Shigella. Conclusions: High lymphocyte counts are associated with disturbances of gut microbiota and emergence of opportunistic pathogens in children with NAFLD.
Collapse
Affiliation(s)
- Tian Liang
- Department of Child, Adolescent Health and Maternal Care, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Dan Li
- Yale School of Public Health, Yale University, New Haven, CT 06510-3201, USA
| | - Jiawulan Zunong
- Department of Child, Adolescent Health and Maternal Care, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Menglong Li
- Department of Child, Adolescent Health and Maternal Care, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Nubiya Amaerjiang
- Department of Child, Adolescent Health and Maternal Care, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Huidi Xiao
- Department of Child, Adolescent Health and Maternal Care, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Nourhan M. Khattab
- Department of Child, Adolescent Health and Maternal Care, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Sten H. Vermund
- Yale School of Public Health, Yale University, New Haven, CT 06510-3201, USA
| | - Yifei Hu
- Department of Child, Adolescent Health and Maternal Care, School of Public Health, Capital Medical University, Beijing 100069, China
- Correspondence: or ; Tel.: +86-10-83911747
| |
Collapse
|
31
|
Vyletelová V, Nováková M, Pašková Ľ. Alterations of HDL's to piHDL's Proteome in Patients with Chronic Inflammatory Diseases, and HDL-Targeted Therapies. Pharmaceuticals (Basel) 2022; 15:1278. [PMID: 36297390 PMCID: PMC9611871 DOI: 10.3390/ph15101278] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/03/2022] [Accepted: 10/14/2022] [Indexed: 09/10/2023] Open
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis, steatohepatitis, periodontitis, chronic kidney disease, and others are associated with an increased risk of atherosclerotic cardiovascular disease, which persists even after accounting for traditional cardiac risk factors. The common factor linking these diseases to accelerated atherosclerosis is chronic systemic low-grade inflammation triggering changes in lipoprotein structure and metabolism. HDL, an independent marker of cardiovascular risk, is a lipoprotein particle with numerous important anti-atherogenic properties. Besides the essential role in reverse cholesterol transport, HDL possesses antioxidative, anti-inflammatory, antiapoptotic, and antithrombotic properties. Inflammation and inflammation-associated pathologies can cause modifications in HDL's proteome and lipidome, transforming HDL from atheroprotective into a pro-atherosclerotic lipoprotein. Therefore, a simple increase in HDL concentration in patients with inflammatory diseases has not led to the desired anti-atherogenic outcome. In this review, the functions of individual protein components of HDL, rendering them either anti-inflammatory or pro-inflammatory are described in detail. Alterations of HDL proteome (such as replacing atheroprotective proteins by pro-inflammatory proteins, or posttranslational modifications) in patients with chronic inflammatory diseases and their impact on cardiovascular health are discussed. Finally, molecular, and clinical aspects of HDL-targeted therapies, including those used in therapeutical practice, drugs in clinical trials, and experimental drugs are comprehensively summarised.
Collapse
Affiliation(s)
| | | | - Ľudmila Pašková
- Department of Cell and Molecular Biology of Drugs, Faculty of Pharmacy, Comenius University, 83232 Bratislava, Slovakia
| |
Collapse
|
32
|
Zhang T, Zhu T, Wen J, Chen Y, Wang L, Lv X, Yang W, Jia Y, Qu C, Li H, Wang H, Qu L, Ning Z. Gut microbiota and transcriptome analysis reveals a genetic component to dropping moisture in chickens. Poult Sci 2022; 102:102242. [PMID: 36931071 PMCID: PMC10036737 DOI: 10.1016/j.psj.2022.102242] [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: 12/09/2021] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 03/12/2023] Open
Abstract
High dropping moisture (DM) in poultry production has deleterious effects on the environment, feeding cost, and public health of people and animals. To explore the contributing genetic components, we classified DM of 67-wk-old Rhode Island Red (RIR) hens at 4 different levels and evaluated the underlying genetic heritability. We found the heritability of DM to be 0.219, indicating a moderately heritable trait. We then selected chickens with the highest and lowest DM levels. Using transcriptome, we only detected 12 differentially expressed genes (DEGs) between these 2 groups from the spleen, and 1,507 DEGs from intestinal tissues (jejunum and cecum). The low number of DEGs observed in the spleen suggests that differing moisture levels are not attributed to pathogenic infection. Fourteen of the intestinal high expressed genes are associated with water-salt metabolism (WSM). We also investigated the gut microbial composition by 16S rRNA gene amplicon sequencing. Six different microbial operational taxonomic units (OTUs) (Cetobacterium, Sterolibacterium, Elusimicrobium, Roseburia, Faecalicoccus, and Megamonas) between the 2 groups from jejunum and cecum are potentially biomarkers related to DM levels. Our results identify a genetic component to chicken DM, and can guide breeding strategies.
Collapse
Affiliation(s)
- Tongyu Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Tao Zhu
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Junhui Wen
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yu Chen
- Beijing Animal Husbandry and Veterinary Station, Beijing, China
| | - Liang Wang
- Beijing Animal Husbandry and Veterinary Station, Beijing, China
| | - Xueze Lv
- Beijing Animal Husbandry and Veterinary Station, Beijing, China
| | - Weifang Yang
- Beijing Animal Husbandry and Veterinary Station, Beijing, China
| | - Yaxiong Jia
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Changqing Qu
- Engineering Technology Research Center of Anti-aging Chinese Herbal Medicine of Anhui Province, Fuyang Normal University, Fuyang, China
| | - Haiying Li
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| | - Huie Wang
- College of Animal Science, Tarim University, Xinjiang, China
| | - Lujiang Qu
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| | - Zhonghua Ning
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| |
Collapse
|
33
|
Abudujilile D, Wang W, Aimaier A, Chang L, Dong Y, Wang Y, Fan X, Ma Y, Wang Y, Ziyayiding D, Ma Y, Lv J, Li J. Cistanche tubulosa phenylethanoid glycosides suppressed adipogenesis in 3T3-L1 adipocytes and improved obesity and insulin resistance in high-fat diet induced obese mice. BMC Complement Med Ther 2022; 22:270. [PMID: 36229811 PMCID: PMC9564091 DOI: 10.1186/s12906-022-03743-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 09/23/2022] [Indexed: 11/10/2022] Open
Abstract
Background Cistanche tubulosa is an editable and medicinal traditional Chinese herb and phenylethanoid glycosides are its major components, which have shown various beneficial effects such as anti-tumor, anti-oxidant and neuroprotective activities. However, the anti-obesity effect of C. tubulosa phenylethanoid glycosides (CTPG) and their regulatory effect on gut microbiota are still unclear. In the present study, we investigated its anti-obesity effect and regulatory effect on gut microbiota by 3T3-L1 cell model and obesity mouse model. Methods 3T3-L1 adipocytes were used to evaluate CTPG effects on adipogenesis and lipids accumulation. Insulin resistant 3T3-L1 cells were induced and used to measure CTPG effects on glucose consumption and insulin sensitivity. High-fat diet (HFD)-induced C57BL/6 obese mice were used to investigate CTPG effects on fat deposition, glucose and lipid metabolism, insulin resistance and intestinal microorganism. Results In vitro data showed that CTPG significantly decreased the triglyceride (TG) and non-esterified fatty acid (NEFA) contents of the differentiated 3T3-L1 adipocytes in a concentration-dependent manner without cytotoxicity, and high concentration (100 µg/ml) of CTPG treatment dramatically suppressed the level of monocyte chemoattractant protein-1 (MCP-1) in 3T3-L1 mature adipocytes. Meanwhile, CTPG increased glucose consumption and decreased NEFA level in insulin resistant 3T3-L1 cells. We further found that CTPG protected mice from the development of obesity by inhibiting the expansion of adipose tissue and adipocyte hypertrophy, and improved hepatic steatosis by activating AMPKα to reduce hepatic fat accumulation. CTPG ameliorated HFD-induced hyperinsulinemia, hyperglycemia, inflammation and insulin resistance by activating IRS1/Akt/GLUT4 insulin signaling pathway in white adipose tissue. Moreover, gut microbiota structure and metabolic functions in HFD-induced obese mice was changed by CTPG, especially short chain fatty acids-producing bacteria including Blautia, Roseburia, Butyrivibrio and Bacteriodes were significantly increased by CTPG treatment. Conclusions CTPG effectively suppressed adipogenesis and lipid accumulation in 3T3-L1 adipocytes and ameliorated HFD-induced obesity and insulin resistance through activating AMPKα and IRS1/AKT/GLUT4 signaling pathway and regulating the composition and metabolic functions of gut microbiota. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03743-6.
Collapse
Affiliation(s)
- Dilinazi Abudujilile
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| | - Weilan Wang
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| | - Alimu Aimaier
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| | - Lili Chang
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| | - Yuliang Dong
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| | - Yiye Wang
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| | - Xu Fan
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| | - Yu Ma
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| | - Yongli Wang
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| | - Dilinigeer Ziyayiding
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| | - Yuan Ma
- grid.413254.50000 0000 9544 7024College of Resource and Environment Sciences, Xinjiang University, Urumqi, 830017 China
| | - Jie Lv
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| | - Jinyao Li
- grid.413254.50000 0000 9544 7024Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830017 China
| |
Collapse
|
34
|
Han Y, Li L, Wang B. Role of Akkermansia muciniphila in the development of nonalcoholic fatty liver disease: current knowledge and perspectives. Front Med 2022; 16:667-685. [PMID: 36318353 DOI: 10.1007/s11684-022-0960-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/06/2022] [Indexed: 11/19/2022]
|
35
|
Zhang C, Ma K, Nie K, Deng M, Luo W, Wu X, Huang Y, Wang X. Assessment of the safety and probiotic properties of Roseburia intestinalis: A potential “Next Generation Probiotic”. Front Microbiol 2022; 13:973046. [PMID: 36160246 PMCID: PMC9493362 DOI: 10.3389/fmicb.2022.973046] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/05/2022] [Indexed: 12/04/2022] Open
Abstract
Roseburia intestinalis is an anaerobic bacterium that produces butyric acid and belongs to the phylum Firmicutes. There is increasing evidence that this bacterium has positive effects on several diseases, including inflammatory bowel disease, atherosclerosis, alcoholic fatty liver, colorectal cancer, and metabolic syndrome, making it a potential “Next Generation Probiotic.” We investigated the genomic characteristics, probiotic properties, cytotoxicity, oral toxicity, colonization characteristics of the bacterium, and its effect on the gut microbiota. The genome contains few genes encoding virulence factors, three clustered regularly interspaced short palindromic repeat (CRISPR) sequences, two Cas genes, no toxic biogenic amine synthesis genes, and several essential amino acid and vitamin synthesis genes. Seven prophages and 41 genomic islands were predicted. In addition to a bacteriocin (Zoocin A), the bacterium encodes four metabolic gene clusters that synthesize short-chain fatty acids and 222 carbohydrate-active enzyme modules. This bacterium is sensitive to antibiotics specified by the European Food Safety Authority, does not exhibit hemolytic or gelatinase activity, and exhibits some acid resistance. R. intestinalis adheres to intestinal epithelial cells and inhibits the invasion of certain pathogens. In vitro experiments showed that the bacterium was not cytotoxic. R. intestinalis did not affect the diversity or abundance of the gut flora. Using the fluorescent labelling method, we discovered that R. intestinalis colonizes the cecum and mucus of the colon. An oral toxicity study did not reveal any obvious adverse effects. The lethal dose (LD)50 of R. intestinalis exceeded 1.9 × 109 colony forming units (CFU)/kg, whereas the no observed adverse effect level (NOAEL) derived from this study was 1.32 × 109 CFU/kg/day for 28 days. The current research shows that, R. intestinalis is a suitable next-generation probiotic considering its probiotic properties and safety.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Kejia Ma
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Kai Nie
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Minzi Deng
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Weiwei Luo
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xing Wu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yujun Huang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoyan Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
- *Correspondence: Xiaoyan Wang,
| |
Collapse
|
36
|
Gilley SP, Ruebel ML, Sims C, Zhong Y, Turner D, Lan RS, Pack LM, Piccolo BD, Chintapalli SV, Abraham A, Bode L, Andres A, Shankar K. Associations between maternal obesity and offspring gut microbiome in the first year of life. Pediatr Obes 2022; 17:e12921. [PMID: 35478493 PMCID: PMC9641193 DOI: 10.1111/ijpo.12921] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 03/04/2022] [Accepted: 03/24/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Maternal obesity is an important determinant of offspring obesity risk, which may be mediated via changes in the infant microbiome. OBJECTIVES We examined infant faecal microbiome, short-chain fatty acids (SCFA), and maternal human milk oligosaccharides (HMO) in mothers with overweight/obese body mass index (BMI) (OW) compared with normal weight (NW) (Clinicaltrials.gov NCT01131117). METHODS Infant stool samples at 1, 6, and 12 months were analysed by 16S rRNA sequencing. Maternal (BODPOD) and infant (quantitative nuclear magnetic resonance [QMR]) adiposity were measured. HMOs at 2 months postpartum and faecal SCFAs at 1 month were also assessed. Statistical analyses included multivariable and mixed linear models for assessment of microbiome diversity, composition, and associations of taxonomic abundance with metabolic and anthropometric variables. RESULTS At 1 month, offspring of women with obesity had lower abundance of SCFA-producing bacteria (including Ruminococcus and Turicibacter) and lower faecal butyric acid levels. Lachnospiraceae abundance was lower in OW group at 6 months, and infant fat mass was negatively associated with the levels of Sutterella. Gradient boosting machine models indicated that higher α-diversity and specific microbial taxa at 1 month predicted elevated adiposity at 12 months with overall accuracy of 76.5%. Associations between maternal HMO concentrations and infant bacterial taxa differed between NW and OW groups. CONCLUSIONS Elevated maternal BMI is associated with relative depletion of butyrate-producing microbes and faecal butyrate in the early infant faecal microbiome. Overall microbial richness may aid in prediction of elevated adiposity in later infancy.
Collapse
Affiliation(s)
- Stephanie P Gilley
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Meghan L Ruebel
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Clark Sims
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ying Zhong
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Donald Turner
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Renny S Lan
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Lindsay M Pack
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Brian D Piccolo
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ann Abraham
- Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California San Diego, La Jolla, California, USA
| | - Lars Bode
- Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California San Diego, La Jolla, California, USA
| | - Aline Andres
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, Colorado, USA
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
37
|
A Polyclonal Aptamer Library for the Specific Binding of the Gut Bacterium Roseburia intestinalis in Mixtures with Other Gut Microbiome Bacteria and Human Stool Samples. Int J Mol Sci 2022; 23:ijms23147744. [PMID: 35887092 PMCID: PMC9317077 DOI: 10.3390/ijms23147744] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 02/07/2023] Open
Abstract
Roseburia intestinalis has received attention as a potential probiotic bacterium. Recent studies have demonstrated that changes in its intestinal abundance can cause various diseases, such as obesity, enteritis and atherosclerosis. Probiotic administration or fecal transplantation alter the structure of the intestinal flora, offering possibilities for the prevention and treatment of these diseases. However, current monitoring methods, such as 16S rRNA sequencing, are complex and costly and require specialized personnel to perform the tests, making it difficult to continuously monitor patients during treatment. Hence, the rapid and cost-effective quantification of intestinal bacteria has become an urgent problem to be solved. Aptamers are of emerging interest because their stability, low immunogenicity and ease of modification are attractive properties for a variety of applications. We report a FluCell-SELEX polyclonal aptamer library specific for R. intestinalis isolated after seven evolution rounds, that can bind and label this organism for fluorescence microscopy and binding assays. Moreover, R. intestinalis can be distinguished from other major intestinal bacteria in complex defined mixtures and in human stool samples. We believe that this preliminary evidence opens new avenues towards aptamer-based electronic biosensors as new powerful and inexpensive diagnostic tools for the relative quantitative monitoring of R. intestinalis in gut microbiomes.
Collapse
|
38
|
Leng J, Tian HJ, Fang Y, Hu YY, Peng JH. Amelioration of Non-Alcoholic Steatohepatitis by Atractylodes macrocephala Polysaccharide, Chlorogenic Acid, and Geniposide Combination Is Associated With Reducing Endotoxin Gut Leakage. Front Cell Infect Microbiol 2022; 12:827516. [PMID: 35865826 PMCID: PMC9294165 DOI: 10.3389/fcimb.2022.827516] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 06/03/2022] [Indexed: 01/01/2023] Open
Abstract
Gut-derived lipopolysaccharide (LPS) leaking through the dysfunctional intestinal barrier contributes to the onset of non-alcoholic steatohepatitis (NASH) by triggering inflammation in the liver. In the present study, a combination consisting of Atractylodes macrocephala polysaccharide (A), chlorogenic acid (C), and geniposide (G) (together, ACG), was shown to ameliorate NASH in mice and reduce hepatic LPS signaling and endotoxemia without decreasing the abundance of identified Gram-negative bacteria through restoring the intestinal tight junctions. Our data indicated that inhibition of LPS gut leakage by the ACG combination contributed to its amelioration of NASH.
Collapse
Affiliation(s)
- Jing Leng
- Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua-jie Tian
- Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Yi Fang
- Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Yi-yang Hu
- Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Institute of Clinical Pharmacology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Jing-hua Peng, ; Yi-yang Hu,
| | - Jing-hua Peng
- Institute of Liver Diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- *Correspondence: Jing-hua Peng, ; Yi-yang Hu,
| |
Collapse
|
39
|
Dai Y, Zhu W, Zhou J, Shen T. The combination of berberine and evodiamine ameliorates high-fat diet-induced non-alcoholic fatty liver disease associated with modulation of gut microbiota in rats. Braz J Med Biol Res 2022; 55:e12096. [PMID: 35584453 PMCID: PMC9113531 DOI: 10.1590/1414-431x2022e12096] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/26/2022] [Indexed: 11/21/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is considered to be a manifestation of hepatic metabolic syndrome. Some studies on the pathogenesis of NAFLD by targeting gut microbiota have attracted wide attention. Previous studies have demonstrated the positive effects of berberine and evodiamine on metabolic diseases and gut microbiota dysbiosis. However, it is not known whether the combination of berberine and evodiamine (BE) can prevent the development of high-fat diet (HFD)-induced NAFLD. Therefore, we aimed to explore the protective effects of BE on the development of HFD-induced NAFLD from the perspective of the gut microbiota. Gut microbiota profiles were established by high throughput sequencing of the bacterial 16S ribosomal RNA gene. The effects of BE on liver and intestinal tissue, intestinal barrier integrity, and hepatic inflammation were also investigated. The results showed that the abundance and diversity of gut microbiota were enriched by BE treatment, with an increase in beneficial bacteria, such as Lactobacillus, Ruminococcus, and Prevotella, and a decrease in pathogenic bacteria such as Fusobacterium and Lachnospira. In addition, BE effectively improved liver fat accumulation and tissue damage, inhibited the apoptosis of intestinal epithelial cells, increased the contents of intestinal tight junction proteins, and decreased the expression of pro-inflammatory factors. Consequently, BE treatment could be an effective and alternative strategy for alleviating NAFLD by modulating gut microbiota and safeguarding the intestinal barrier.
Collapse
Affiliation(s)
- Yufan Dai
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenyu Zhu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | | - Tao Shen
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
40
|
Liu Y, Tan Y, Huang J, Wu C, Fan X, Stalin A, Lu S, Wang H, Zhang J, Zhang F, Wu Z, Li B, Huang Z, Chen M, Cheng G, Mou Y, Wu J. Revealing the Mechanism of Huazhi Rougan Granule in the Treatment of Nonalcoholic Fatty Liver Through Intestinal Flora Based on 16S rRNA, Metagenomic Sequencing and Network Pharmacology. Front Pharmacol 2022; 13:875700. [PMID: 35559233 PMCID: PMC9086680 DOI: 10.3389/fphar.2022.875700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/16/2022] [Indexed: 12/12/2022] Open
Abstract
Background: The incidence of Nonalcoholic Fatty Liver (NAFL) is increasing year by year, growing evidence suggests that the intestinal flora plays a causative role in NAFL. Huazhi Rougan Granule (HRG) is commonly used in the clinical treatment of NAFL. It is reported that it can reduce lipids and protect the liver, but no research has confirmed whether the drug's effect is related to the intestinal flora. Therefore, we investigated whether the effect of HRG is related to the regulation of intestinal flora to further explore the mechanism of HRG in the treatment of NAFL through intestinal flora. Methods: In this study, C57BL/6J mice were fed a high-fat diet for 8 weeks, and the high-fat diet plus HRG or polyene phosphatidylcholine capsules were each administered by gavage for 4 weeks. High-throughput sequencing, network pharmacology, and molecular docking were used to explore the mechanism of HRG in the treatment of NAFL through intestinal flora. Results: HRG treatment can reduce body weight gain, lipid accumulation in liver and lipogenesis and reduce serum biochemical indexes in high-fat-fed mice. Analysis of intestinal flora showed that HRG changed the composition of intestinal flora, which was characterized by a decrease in the Firmicutes/Bacteroidetes ratio. Moreover, the species distribution was significantly correlated with AKP, HDL-C, and TG. Metagenetic analysis showed that HRG altered the functional composition and functional diversity of microorganisms, which was mainly characterized by an increase in the abundance of metabolic pathways. The network pharmacology results show that the mechanism of HRG in the treatment of NAFL through intestinal flora is mainly reflected in the biological process of gene function and related to infectious diseases, immune systems, and signal transduction pathways, such as cytokine-cytokine receptor interaction, Chagas disease, IL-17 signaling pathway and other signaling pathways. Conclusion: These results strongly suggest that HRG may alleviate NAFL by preventing IFD.
Collapse
Affiliation(s)
- Yingying Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Tan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaqi Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotian Fan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Shan Lu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Haojia Wang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fanqin Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhishan Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Bing Li
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Zhihong Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Meilin Chen
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guoliang Cheng
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Yanfang Mou
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
41
|
Yan J, Nie Y, Liu Y, Li J, Wu L, Chen Z, He B. Yiqi-Bushen-Tiaozhi Recipe Attenuated High-Fat and High-Fructose Diet Induced Nonalcoholic Steatohepatitis in Mice via Gut Microbiota. Front Cell Infect Microbiol 2022; 12:824597. [PMID: 35531334 PMCID: PMC9072834 DOI: 10.3389/fcimb.2022.824597] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/21/2022] [Indexed: 12/19/2022] Open
Abstract
Aim To investigate the treating effect of Yiqi-Bushen-Tiaozhi (YBT) recipe on nonalcoholic steatohepatitis (NASH) mice, determine whether the outcome was associated with gut microbiota, and clarify the regulating mechanism. Methods NASH mice were induced by high-fat and high-fructose diets (HFFD). In the fifth week, mice in the YBT group were orally administrated YBT (22.12g·kg-1·d-1) daily for 12 weeks. Fresh stool of mice was collected at the 16th week for fecal 16S rDNA analysis. Hepatic pathology and biochemical indicators were used to reflect the improvement of YBT on hepatic inflammation and lipid metabolism in NASH mice. Quantitative real-time PCR (qRT-PCR) was used to verify the results of PICRUSt analysis. Results Results of the pathological and biochemical index showed that YBT could improve NASH mice. Compared with improving inflammation and hepatocyte damage, YBT may be more focused on enhancing metabolic disorders in mice, such as increasing HDL-c level. The diversity and richness of the gut microbiota of NASH mice induced by HFFD are significantly different from the normal control (NC) group. After YBT treatment, the diversity and richness of the mice microbiota will be increased to similar NC mice. Intestinimonas, Acetatifactor, Alistipes, Intestinimonas, Acetatifactor, and Alistipes have the most significant changes in the class level. PICRUSt analysis was performed to predict genomic functions based on the 16S rDNA results and reference sequencing. The efficacy of YBT in the treatment of NASH can be achieved by regulating the diversity and richness of gut microbiota. PICRUSt analysis results showed that the most relevant function of the microbiota construction variations is α- Linolenic acid (ALA) metabolism. Results of qRT-PCR showed significant differences between groups in the expression of Fatty acid desaturase 1 (FADS1), Fatty acid desaturase 2 (FADS2), Acyl-CoA Oxidase 1 (ACOX1), and Acyl-CoA Oxidase 2 (ACOX2) related to ALA metabolism. The expression of the above genes will be inhibited in the liver and small intestine of the HFFD group mice, and the expression can be restored after YBT treatment. Conclusion YBT could treat NASH mice by improving the diversity and richness of gut microbiota and further the improvement of ALA metabolism.
Collapse
Affiliation(s)
- Junbin Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- The Second Central Laboratory, Key Lab of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yunmeng Nie
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuan Liu
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- The Second Central Laboratory, Key Lab of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingya Li
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- The Second Central Laboratory, Key Lab of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Liyan Wu
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zhiyun Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- The Second Central Laboratory, Key Lab of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Beihui He
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- The Second Central Laboratory, Key Lab of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
42
|
Liao CA, Huang CH, Ho HH, Chen JF, Kuo YW, Lin JH, Tsai SY, Tsai HY, Yeh YT. A Combined Supplement of Probiotic Strains AP-32, bv-77, and CP-9 Increased Akkermansia mucinphila and Reduced Non-Esterified Fatty Acids and Energy Metabolism in HFD-Induced Obese Rats. Nutrients 2022; 14:nu14030527. [PMID: 35276886 PMCID: PMC8839477 DOI: 10.3390/nu14030527] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Obesity is referred to as a condition in which excess body fat has accumulated to an extent that it causes negative impacts on health. The formation of body fat is regulated by complicated networks in relation to energy metabolism, and gut microbiota have been regarded as a key player. Studies have shown that supplements of probiotics provide benefits to health, including an improvement in metabolic syndrome and the control of body weight. In the present study, three probiotic strains, AP-32, bv-77, and CP-9, stood out from nine candidates using a lipid consumption assay, and were subsequently introduced to further animal tests. A rodent model of obesity was induced by a high-fat diet (HFD) in Sprague-Dawley (SD) rats, and three probiotic strains were administered either separately or in a mixture. A low dose (5 × 109 CFU/kg/day) and a high dose (2.5 × 1010 CFU/kg/day) of probiotics were orally provided to obese rats. The bioeffects of the probiotic supplements were evaluated based on five aspects: (1) the body weight and growth rate; (2) ketone bodies, non-esterified fatty acids (NEFAs), and feed efficiency; (3) blood biochemistry; (4) fat content; and (5) gut microbiota composition. Our results demonstrated that the supplement of AP-32, CP-9, and bv-77 alleviated the increasing rate of body weight and prevented the elevation of NEFAs and ketone bodies in obese rats. Although the effect on fat content showed a minor improvement, the supplement of probiotics displayed significant improvements in HFD-induced poor blood biochemical characteristics, such as alanine aminotransferase (ALT), aspartate Transaminase (AST), and uric acid, within 4 weeks. Furthermore, the combined supplement of three strains significantly increased Akkermansia mucinphila as compared with three individual strains, while its enrichment was negatively correlated with NEFAs and energy metabolism. In general, a mixture of three probiotic strains delivered a better outcome than a single strain, and the high dose of supplements provided a more profound benefit than the low dose. In conclusion, three probiotic strains, AP-32, bv-77, and CP-9, can alleviate body fat formation in obese rats. Furthermore, a combined supplement of these three probiotic strains may have potential in treating or controlling metabolic disorders.
Collapse
Affiliation(s)
- Chorng-An Liao
- Aging and Diseases Prevention Research Center, Fooyin University, Kaohsiung 83102, Taiwan; (C.-A.L.); (C.-H.H.)
- Biomed Analysis Center, Fooyin Hospital, Pingtung 92847, Taiwan
| | - Cheng-Hsieh Huang
- Aging and Diseases Prevention Research Center, Fooyin University, Kaohsiung 83102, Taiwan; (C.-A.L.); (C.-H.H.)
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hsieh-Hsun Ho
- Department of Research and Design, glac Biotech Co., Ltd., Tainan 74442, Taiwan; (H.-H.H.); (J.-F.C.); (Y.-W.K.); (J.-H.L.); (S.-Y.T.)
| | - Jui-Fen Chen
- Department of Research and Design, glac Biotech Co., Ltd., Tainan 74442, Taiwan; (H.-H.H.); (J.-F.C.); (Y.-W.K.); (J.-H.L.); (S.-Y.T.)
| | - Yi-Wei Kuo
- Department of Research and Design, glac Biotech Co., Ltd., Tainan 74442, Taiwan; (H.-H.H.); (J.-F.C.); (Y.-W.K.); (J.-H.L.); (S.-Y.T.)
| | - Jia-Hung Lin
- Department of Research and Design, glac Biotech Co., Ltd., Tainan 74442, Taiwan; (H.-H.H.); (J.-F.C.); (Y.-W.K.); (J.-H.L.); (S.-Y.T.)
| | - Shin-Yu Tsai
- Department of Research and Design, glac Biotech Co., Ltd., Tainan 74442, Taiwan; (H.-H.H.); (J.-F.C.); (Y.-W.K.); (J.-H.L.); (S.-Y.T.)
| | - Hui-Yun Tsai
- Aging and Diseases Prevention Research Center, Fooyin University, Kaohsiung 83102, Taiwan; (C.-A.L.); (C.-H.H.)
- Department of Nutrition and Health Science, Fooyin University, Kaohsiung 83102, Taiwan
- Correspondence: (H.-Y.T.); (Y.-T.Y.); Tel.: +886-7-781-1151 (ext. 6800) (H.-Y.T. & Y.-T.Y.)
| | - Yao-Tsung Yeh
- Aging and Diseases Prevention Research Center, Fooyin University, Kaohsiung 83102, Taiwan; (C.-A.L.); (C.-H.H.)
- Biomed Analysis Center, Fooyin Hospital, Pingtung 92847, Taiwan
- Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung 83102, Taiwan
- Correspondence: (H.-Y.T.); (Y.-T.Y.); Tel.: +886-7-781-1151 (ext. 6800) (H.-Y.T. & Y.-T.Y.)
| |
Collapse
|
43
|
Wu N, Zhou J, Mo H, Mu Q, Su H, Li M, Yu Y, Liu A, Zhang Q, Xu J, Yu W, Liu P, Liu G. The Gut Microbial Signature of Gestational Diabetes Mellitus and the Association With Diet Intervention. Front Cell Infect Microbiol 2022; 11:800865. [PMID: 35096649 PMCID: PMC8795975 DOI: 10.3389/fcimb.2021.800865] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/24/2021] [Indexed: 12/11/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is a high-risk pregnancy complication that is associated with metabolic disorder phenotypes, such as abnormal blood glucose and obesity. The link between microbiota and diet management contributes to metabolic homeostasis in GDM. Therefore, it is crucial to understand the structure of the gut microbiota in GDM and to explore the effect of dietary management on the microbiota structure. In this study, we analyzed the composition of the gut microbiota between 27 GDM and 30 healthy subjects at two time points using Illumina HiSeq 2500 platform. The taxonomy analyses suggested that the overall bacteria clustered by diabetes status, rather than diet intervention. Of particular interest, the phylum Acidobacteria in GDM was significantly increased, and positively correlated with blood glucose levels. Moreover, Partial least-squares discriminant analysis (PLS-DA) revealed that certain genera in the phyla Firmicutes, Bacteroidetes, Proteobacteria, and Lentisphaerae characterized the GDM gut microbiota. Correlation analysis indicated that blood glucose levels and BMI index were correlated with the relative abundance of SCFAS-producing genera. Through the comparison between the GDM and healthy samples with or without diet intervention, we discovered that the role of short-term diet management in GDM processes is associated with the change in the Firmicutes/Bacteroidetes ratio and some specific taxa, rather than an alternative gut microbial pattern. Our study have important implications for understanding the beneficial effects of diet intervention on the specific gut microbiota and thus possibly their metabolism in pregnant women with GDM.
Collapse
Affiliation(s)
- Na Wu
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, China
| | - Jingwei Zhou
- Department of Gynecology and Obstetrics, Peking University People’s Hospital, Beijing, China
| | - Heng Mo
- Department of Stomatology, Peking University People’s Hospital, Beijing, China
| | - Qing Mu
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, China
| | - Huiting Su
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, China
| | - Mei Li
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, China
| | - Yimeng Yu
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, China
| | - Aiyu Liu
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, China
| | - Qi Zhang
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, China
| | - Jun Xu
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, China
| | - Weidong Yu
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, China
| | - Peng Liu
- Department of Clinical Nutrition, Peking University People’s Hospital, Beijing, China
| | - Guoli Liu
- Department of Gynecology and Obstetrics, Peking University People’s Hospital, Beijing, China
| |
Collapse
|