1
|
Dong Y, Meng F, Wang J, Wei J, Zhang K, Qin S, Li M, Wang F, Wang B, Liu T, Zhong W, Cao H. Desulfovibrio vulgaris flagellin exacerbates colorectal cancer through activating LRRC19/TRAF6/TAK1 pathway. Gut Microbes 2025; 17:2446376. [PMID: 39718561 DOI: 10.1080/19490976.2024.2446376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/22/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024] Open
Abstract
The initiation and progression of colorectal cancer (CRC) are intimately associated with genetic, environmental and biological factors. Desulfovibrio vulgaris (DSV), a sulfate-reducing bacterium, has been found excessive growth in CRC patients, suggesting a potential role in carcinogenesis. However, the precise mechanisms underlying this association remain incompletely understood. We have found Desulfovibrio was abundant in high-fat diet-induced Apcmin/+ mice, and DSV, a member of Desulfovibrio, triggered colonocyte proliferation of germ-free mice. Furthermore, the level of DSV progressively rose from healthy individuals to CRC patients. Flagella are important accessory structures of bacteria, which can help them colonize and enhance their invasive ability. We found that D. vulgaris flagellin (DVF) drove the proliferation, migration, and invasion of CRC cells and fostered the growth of CRC xenografts. DVF enriched the epithelial-mesenchymal transition (EMT)-associated genes and characterized the facilitation of DVF on EMT. Mechanistically, DVF induced EMT through a functional transmembrane receptor called leucine-rich repeat containing 19 (LRRC19). DVF interacted with LRRC19 to modulate the ubiquitination of tumor necrosis factor receptor-associated factor (TRAF)6, rather than TRAF2. This interaction drove the ubiquitination of pivotal molecule TAK1, further enhancing its autophosphorylation and ultimately contributing to EMT. Collectively, DVF interacts with LRRC19 to activate the TRAF6/TAK1 signaling pathway, thereby promoting the EMT of CRC. These data shed new light on the role of gut microbiota in CRC and establish a potential clinical therapeutic target.
Collapse
Affiliation(s)
- Yue Dong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Fanyi Meng
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jingyi Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jingge Wei
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Kexin Zhang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Siqi Qin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Mengfan Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Fucheng Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| |
Collapse
|
2
|
Wang W, Zhang K, Zhang K, Wu R, Tang Y, Li Y. Gut microbiota promotes cholesterol gallstone formation through the gut-metabolism-gene axis. Microb Pathog 2025; 203:107446. [PMID: 40118296 DOI: 10.1016/j.micpath.2025.107446] [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: 06/17/2024] [Revised: 02/17/2025] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Gallstone disease, arising from the interplay between host metabolism and gut microbiota, represents a significant health concern. Dysbiosis of the gut microbiome and disruptions in circadian rhythm contribute to the pathogenesis of gallstones. This study conducted a comprehensive analysis of gut microbiota and metabolites derived from stool and serum samples of 28 patients with cholesterol gallstones (CGS) and 19 healthy controls, employing methodologies such as 16S rRNA sequencing, metaproteomics, metabolomics, and host genetic analysis. Additionally, a retrospective cohort study was utilized to assess the efficacy of probiotics or ursodeoxycholic acid (UDCA) in preventing CGS formation post-bariatric surgery. RESULTS In CGS patients, gut microbiota diversity shifted, with harmful bacteria rising and beneficial ones declining. The altered microbiota primarily affected amino acid, lipid, nucleotide, and carbohydrate metabolism. Metabolic abnormalities were noted in amino acids, glucose, lipids, and bile acids with decreased levels of ursodeoxycholic, glycosodeoxycholic, and glycolithocholic acids, and increased glycohyodeoxycholic and allocholic acids. Glutamine and alanine levels dropped, while phenylalanine and tyrosine rosed. Animal studies confirmed gene changes in gallbladder tissues related to bile acid, energy, glucose, and lipid metabolism. Importantly, UDCA and probiotics effectively reduced CGS risk post-bariatric surgery, especially when combined. CONCLUSIONS Multi-omics can clarify CGS pathology, by focusing on the gut-metabolism-gene axis, paving the way for future studies on CGS prevention and treatment through gut microbiota or metabolic interventions.
Collapse
Affiliation(s)
- Wei Wang
- Department of Interventional, The Second Hospital of Shandong University, Shandong, 250033, China
| | - Kai Zhang
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Shandong University, Shandong, 250033, China
| | - Kun Zhang
- Shanghai Biotree Biotech Co., Ltd., Shanghai, China
| | - Rui Wu
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Shandong University, Shandong, 250033, China
| | - Yu Tang
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Yuliang Li
- Department of Interventional, The Second Hospital of Shandong University, Shandong, 250033, China.
| |
Collapse
|
3
|
Zhang Y, Dai X, Duan R, Wei L. Research progress in the treatment of gallstones with laparoscopic and endoscopic surgery: a narrative review. BMC Surg 2025; 25:238. [PMID: 40442755 PMCID: PMC12123860 DOI: 10.1186/s12893-025-02977-8] [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: 01/05/2025] [Accepted: 05/27/2025] [Indexed: 06/02/2025] Open
Abstract
Cholelithiasis, the formation of stones in the gallbladder, is a common surgical condition with an increasing incidence. Laparoscopic cholecystectomy has become the gold standard for the treatment of symptomatic gallstones due to its favorable outcomes. However, with increasing recognition of the gallbladder's importance, this procedure no longer aligns with patients' desire to preserve the organ. Technological advancements and surgical innovations have led to emerging approaches such as natural orifice transluminal endoscopic surgery (NOTES), robot-assisted laparoscopic cholecystectomy, and cholangioscopy. This narrative review examines the current landscape of surgical interventions for gallstones, highlighting both gallbladder-preserving and removal approaches, and aims to provide insight into their respective outcomes and clinical implications.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun, PR China
| | - Xinyu Dai
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun, PR China
| | - Ruifeng Duan
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun, PR China
| | - Lijuan Wei
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun, PR China.
| |
Collapse
|
4
|
Cheng S, Zhang Y, Jiang L, Li J, Jiang R, Wang F. The association between inflammatory biomarkers and gallstones in Americans under 50 years old. BMC Gastroenterol 2025; 25:393. [PMID: 40399796 PMCID: PMC12096754 DOI: 10.1186/s12876-025-03994-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 05/14/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND This study aimed to assess the link between inflammatory biomarkers (like the neutrophil-to-lymphocyte ratio (NLR), systemic immune-inflammatory index (SII), and systemic inflammation response index (SIRI)) and gallstones in American individuals aged under 50 years. METHODS This study utilized data from the National Health and Nutrition Examination Survey (NHANES) covering 2017 to 2020, centering on individuals under 50 years with comprehensive data on NLR, SII, SIRI, and gallstones. It employed a weighted multiple logistic regression approach to investigate the link between inflammatory biomarkers and gallstones. Furthermore, dose-response relationships and threshold effects were evaluated utilizing restricted cubic spline (RCS) methods and segmented linear regression models. Subgroup examinations and interaction assessments were conducted, too. RESULTS The investigation encompassed a total of 3,295 individuals. Upon comprehensive adjustment for variables, multivariate logistic regression revealed a positive relationship between inflammatory biomarkers and gallstones: ln-NLR (OR = 1.68, 95% CI: 1.06-2.66, p = 0.033), ln-SII (OR = 1.79, 95% CI: 1.08-2.98, p = 0.032), and ln-SIRI (OR = 1.46, 95% CI: 1.07-1.99, p = 0.025). A non-linear association, shaped like an inverse "U", was noted between ln-SIRI and gallstones. To the left of the inflection point (ln-SIRI = 0.35, SIRI = 1.42), a positive link existed between ln-SIRI and gallstones (OR = 2.45, 95% CI: 1.20-5.03); whereas, to the right of the inflection point, the association was statistically insignificant (OR = 0.60, 95% CI: 0.21-1.73). CONCLUSION ln-NLR and ln-SII exhibited a linear and positive relationship with the likelihood of developing gallstones. ln-SIRI demonstrated a nonlinear dose-response relationship with gallstone risk, characterized by an inverted "U" shape.
Collapse
Affiliation(s)
- Shuqin Cheng
- First Central Hospital of Tianjin Medical University, Tianjin, 300070, China
| | - Yaping Zhang
- The Third Central Clinical College of Tianjin Medical University, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Nankai University Affiliated Third Central Hospital, Tianjin, 300072, China
| | - Lu Jiang
- First Central Hospital of Tianjin Medical University, Tianjin, 300070, China
| | - Jiana Li
- First Central Hospital of Tianjin Medical University, Tianjin, 300070, China
- Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin First Central Hospital, No. 24 Fukang Road, Nankai District, Tianjin, 300192, China
| | - Runci Jiang
- First Central Hospital of Tianjin Medical University, Tianjin, 300070, China
- Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin First Central Hospital, No. 24 Fukang Road, Nankai District, Tianjin, 300192, China
| | - Fengmei Wang
- Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin First Central Hospital, No. 24 Fukang Road, Nankai District, Tianjin, 300192, China.
- Department of Gastroenterology and Hepatology, Tianjin First Central Hospital, Tianjin, 300192, China.
| |
Collapse
|
5
|
Liu X, Wang H, Xie Z, Li L, He Y, Meng Z, Li J, Yu J, Du Z, Zheng Y, Liu T, Hao C, Xue D, Wang L, Gao E. Whole Transcriptome-wide Analysis Combined With Summary Data-Based Mendelian Randomization Identifies High-Risk Genes for Cholelithiasis Incidence. Clin Transl Gastroenterol 2025; 16:e00800. [PMID: 39840844 PMCID: PMC12101918 DOI: 10.14309/ctg.0000000000000800] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/04/2024] [Indexed: 01/23/2025] Open
Abstract
INTRODUCTION Cholelithiasis is influenced by various factors, including genetic elements identified in genomewide association studies, but their biological functions are not fully understood. METHODS Analyzing data from the Finngen database with 37,041 cholelithiasis cases and 330,903 controls, this study combined SNP data from GTEx v8 and linkage disequilibriums data from the 1000 Genomes Project. Using the Transcriptomewide Association Studies FUSION protocol and summary data-based Mendelian randomization analysis, it investigated the relationship between gene expression and cholelithiasis, using colocalization tests and conditional analyses to explore causality. RESULTS The study identified genes associated with cholelithiasis in the liver and whole blood, such as LINC01595, TTC39B, and UGT1A3, with several showing colocalization traits. Notably, RP11-378A13.1 and adenosine deaminase acting on RNA (ADAR) were significantly associated with the disease in both tissues. DISCUSSION This research provides insights into the genetic underpinnings of cholelithiasis, highlighting the significant role of gene expression in its development. It establishes new gene associations and identifies potential genetic markers for the disease.
Collapse
Affiliation(s)
- Xuxu Liu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Heming Wang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhihong Xie
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lianghao Li
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yuanhang He
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziang Meng
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiachen Li
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jingjing Yu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhiwei Du
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yi Zheng
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Tianming Liu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chenjun Hao
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Dongbo Xue
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Liyi Wang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Enjun Gao
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
6
|
Pan H, Bi J, Hu H, Huang Y, Li A, Zhang H, Wan Y, Zhan K, Wang K, Zhao Z, Bai X. Chinese herbal medicine improves antioxidant capacity of chicken liver at high stocking density involved gut-liver microbiota axis based on multi-omics technologies. Poult Sci 2025; 104:105015. [PMID: 40106906 PMCID: PMC11964641 DOI: 10.1016/j.psj.2025.105015] [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: 12/20/2024] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Traditional Chinese Medicine (TCM), such as artemisinin, berberine and proanthocyanidin, has been considered an effective additive for broiler production. High density farming (HDF), which is the primary modern mode of chicken production, is associated with animal health problems. This work aimed to evaluate the effects of dietary TCMs (dihydroartemisinin, hydrochloride, and oligomeric proanthocyanidins) on improving the antioxidant capacity of chickens under HDF and their underlying mechanisms. A total of 360 Wuding chickens (134-day-old) were divided into five experimental groups: one normal stocking density (8 birds/m2, control group) and four high stocking density (16 birds/m2), with six replicates for each group. For four HDF groups, one group was fed the basal diet, and the other three groups were fed the basal diet supplemented with 80 mg/kg dihydroartemisinin, 600 mg/kg berberine hydrochloride, and 250 mg/kg grape oligomeric proanthocyanidins, respectively. HDF increased malondialadehyde level, but decreased superoxide dismutase, glutathione and glutathione peroxidase levels in the liver of broiler; however, dietary TCMs apparently alleviated this attenuation. Dietary TCMs significantly decreased the expression of genes involved in cholesterol synthesis in the liver and the levels of tripepides in the intestine of the HDF chickens. Meanwhile, dietary TCMs significantly altered the composition of the liver microbiome in the HDF chickens, expressing by reduced Pseudomonas but enriched Bradyrhizobium. The gut microbiota of the HDF chickens was also altered following dietary TCM administration, with a decreased abundance of Microbacter margulisiae and an increased abundance of acetate synthesis genes. Association analysis of the multi-omics results revealed negative correlations between liver cholesterol synthesis and antioxidant factors that could be regulated by gut microbiota-produced short-chain fatty acids. Furthermore, alleviating of oxidative stress by dietary TCMs also showed significant correlations with the liver microbiome, which could be mediated by tripeptides produced by the gut microbiota. These results indicated that dietary TCM is beneficial in improving antioxidant defenses in HDF chickens and interpreted the mechanisms of action of TCM from the perspective of modern science.
Collapse
Affiliation(s)
- Hongbin Pan
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, PR China
| | - Junlong Bi
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, PR China
| | - Hong Hu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, PR China
| | - Ying Huang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, PR China
| | - Anjian Li
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, PR China
| | - Hao Zhang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing 100193, PR China
| | - Yi Wan
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, Anhui 230031, PR China
| | - Kai Zhan
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, Anhui 230031, PR China
| | - Kunping Wang
- College of Animal Science, Anhui Science and Technology University, Bengbu, 233000, PR China
| | - Zelong Zhao
- Shanghai BIOZERON Biotechnology Co., Ltd., Shanghai, 201800, PR China
| | - Xi Bai
- College of Animal Science, Anhui Science and Technology University, Bengbu, 233000, PR China.
| |
Collapse
|
7
|
Shi C, Yu J, Meng Z, Lu D, Ding H, Sun H, Shi G, Xue D, Meng X. PCSK9 and APOA4: The Dynamic Duo in TMAO-induced Cholesterol Metabolism and Cholelithiasis. J Clin Transl Hepatol 2025; 13:295-305. [PMID: 40206272 PMCID: PMC11976434 DOI: 10.14218/jcth.2024.00403] [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: 11/05/2024] [Revised: 12/31/2024] [Accepted: 01/22/2025] [Indexed: 04/11/2025] Open
Abstract
Background and Aims Cholesterol synthesis and gallstone formation are promoted by trimethylamine-N-oxide (TMAO), a derivative of trimethylamine, which is a metabolite of gut microbiota. However, the underlying mechanisms of TMAO-induced lithogenesis remain incompletely understood. This study aimed to explore the specific molecular mechanisms through which TMAO promotes gallstone formation. Methods Enzyme-linked immunosorbent assays were used to compare serum concentrations of TMAO, apolipoprotein A4 (APOA4), and proprotein convertase subtilisin/kexin type 9 (PCSK9) between patients with cholelithiasis and normal controls. A murine model of TMAO-induced cholelithiasis was employed, incorporating assays of gallstone weight and bile cholesterol content, along with RNA sequencing of murine hepatic tissue. A TMAO-induced AML12 hepatocyte line was constructed and transfected with targeted small interfering RNAs and overexpression plasmids. In vivo and in vitro experiments were performed to determine the expression and regulation of genes related to cholesterol metabolism. Results Serum TMAO and PCSK9 levels were elevated, whereas APOA4 levels were reduced in patients with cholelithiasis. Furthermore, our murine model demonstrated that TMAO upregulated hepatic expression of PCSK9, 3-hydroxy-3-methylglutaryl-CoA reductase, and ATP-binding cassette sub-family G member 5/8, while reducing APOA4 expression, thereby modulating cholesterol metabolism and promoting lithogenesis. PCSK9 and APOA4 were identified as key regulatory genes in the cholesterol metabolic pathway. PCSK9 knockdown increased APOA4 expression, while APOA4 overexpression led to reduced PCSK9 expression. Conclusions TMAO upregulated hepatic PCSK9 expression and reduced APOA4 expression, initiating a feedback loop that dysregulated cholesterol metabolism and promoted lithogenesis.
Collapse
Affiliation(s)
- Chao Shi
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Ambulatory Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jingjing Yu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziang Meng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Dongxu Lu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Haoran Ding
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Haijun Sun
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Ambulatory Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Guangxin Shi
- Department of Internal Medicine, The Third Hospital of Changtu County, Tieling, Liaoning, China
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xianzhi Meng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
8
|
Baruah C, Jorvekar SB, Sarma A, Gogoi G, Roy N, Dutta U, Khanna S, Borkar RM, Kumar A, Barah P. Gallstone Physicochemical Properties and Heavy Metal Concentrations Associated with Gallbladder Carcinogenesis in Assam, India. Chem Res Toxicol 2025; 38:598-608. [PMID: 40172547 DOI: 10.1021/acs.chemrestox.4c00392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Gallbladder cancer (GBC) is an aggressive malignancy, with gallstone disease (GSD) recognized as the primary risk factor. Although the precise mechanism linking GSD to GBC remains unclear, evidence suggests that gallstone characteristics play a significant role. This study investigates the physicochemical characteristics of gallstones critical for GBC development. We analyzed 40 gallstone samples from 30 GSD and 10 GBC with GSD (GBCGS) patients using advanced spectroscopic and imaging techniques such as fourier transform infrared (FTIR), powder X-ray diffraction (PXRD), nuclear magnetic resonance (NMR), and scanning electron microscopy energy-dispersive X-ray (SEM-EDX)). Subsequently, elemental analysis of 10 gallstones each from GBCGS and GSD was conducted via inductively coupled plasma-mass spectrometry (ICP-MS). Gallstones from the GSD group were identified as cholesterol (70%), mixed (13.3%), pigment (6.7%), and calcium carbonate (10%), while the GBCGS group included only cholesterol (70%) and mixed (30%) types. Cholesterol was the dominant organic component in most gallstones, with the cholesterol and mixed types exhibiting highly crystalline phases characterized by a stacked plate-like microstructure, particularly prominent in the GBCGS group. Additionally, the GBCGS group revealed significantly higher concentrations of carcinogenic elements such as arsenic, chromium, mercury, iron, and lead (p < 0.05), suggesting their accumulation in the gallbladder and gallstones. Consequently, our findings highlight that the physicochemical properties of cholesterol-rich gallstones and exposure to carcinogenic elements play a key role in the pathogenesis of GBC in Assam. These results emphasize the need for further research into cholesterol dysregulation and its link to elemental toxicity.
Collapse
Affiliation(s)
- Cinmoyee Baruah
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam 784028, India
| | - Sachin B Jorvekar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Guwahati, Assam 781101, India
| | - Anupam Sarma
- Department of Onco-pathology, Dr. Bhubaneswar Borooah Cancer Institute, Guwahati, Assam 781016, India
| | - Gayatri Gogoi
- Department of Pathology, Assam Medical College and Hospital, Dibrugarh, Assam 786002, India
| | - Nabanita Roy
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam 784028, India
| | - Utpal Dutta
- Department of Pathology, Assam Medical College and Hospital, Dibrugarh, Assam 786002, India
| | - Subhash Khanna
- Department of Minimal Access, Gastro Intestinal, Bariatric and Robotic Surgery, Swagat Super Speciality, and Surgical Hospital, Guwahati 781011, India
| | - Roshan M Borkar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Guwahati, Assam 781101, India
| | - Akshai Kumar
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
- Jyoti and Bhupat Mehta School of Health Science and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Pankaj Barah
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam 784028, India
| |
Collapse
|
9
|
Li YT, Shao WQ, Chen ZM, Lin J, Chen JH. GOLM1 and bile acid synthesis: Correspondence to editorial on "GOLM1 promotes cholesterol gallstone formation via ABCG5-mediated cholesterol efflux in MASH livers". Clin Mol Hepatol 2025; 31:e189-e191. [PMID: 39947708 PMCID: PMC12016590 DOI: 10.3350/cmh.2025.0120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 02/12/2025] [Indexed: 04/18/2025] Open
Affiliation(s)
- Yi-Tong Li
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei-Qing Shao
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhen-Mei Chen
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Lin
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jin-Hong Chen
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Xu T, Fang D, Xu T, Tao X, Wang Z, Liu Y. Exercise-driven gut microbiota alterations enhance colonization resistance against methicillin-resistant Staphylococcus aureus. Cell Rep 2025; 44:115424. [PMID: 40080501 DOI: 10.1016/j.celrep.2025.115424] [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: 09/05/2024] [Revised: 12/15/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Gut microbiota plays a crucial role in resisting the invasion of pathogens, particularly multidrug-resistant (MDR) bacteria, which pose a significant threat to public health. While exercise offers numerous health benefits, its impact on host colonization resistance remains largely unclear. In this study, we demonstrate that moderate exercise significantly reduces gut colonization by methicillin-resistant Staphylococcus aureus (MRSA), a clinically important MDR pathogen. Moreover, we identify an understudied strain of the intestinal probiotic Dubosiella newyorkensis (L8) as a critical factor in mediating exercise-induced colonization resistance against MRSA. Mechanistically, L8 enhances the deprivation of fucose, a crucial carbon source essential for MRSA growth and pathogenicity. This process relies on the high binding affinity of pyruvate to the ILE257 site of the lactate dehydrogenase in L8. Overall, our work highlights the importance of moderate exercise in maintaining host colonization resistance and demonstrates L8 as a probiotic in protecting against MRSA colonization.
Collapse
Affiliation(s)
- Tingting Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Dan Fang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Tianqi Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiuying Tao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhiqiang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China.
| | - Yuan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
11
|
Zhang R, Chen C, Zheng S, Zhang J, Chen W, Chen Z. Preliminary study of biliary microbiota and identification of bacterial species associated with pigmented gallstone formation. Front Cell Infect Microbiol 2025; 15:1532512. [PMID: 40182770 PMCID: PMC11966056 DOI: 10.3389/fcimb.2025.1532512] [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: 11/22/2024] [Accepted: 02/10/2025] [Indexed: 04/05/2025] Open
Abstract
Background Pigmented gallstone disease (PGS) is prevalent in China. Biliary microbiota is certified to be related to the PGS formation. Methods We performed 16S sequencing on both bile and gallstone samples in 16 patients with gallstone disease. We analyzed the microbial composition of the biliary tract and identified crucial bacteria related to the formation of PGS. Results Biliary tract bacterial composition analysis showed heterogeneity of dominated genus among individuals and correlation in bacterial composition between bile and gallstones. We screened 10 prevalent genera with significant abundance in the bile and gallstones. Actinomyces, Streptococcus, and Achromobacter had a significantly higher abundance in gallstones than in bile (P < 0.05). Furthermore, we identified 32 species that harbored uidA, pldA, and plc genes that encoded β-glucuronidase or phospholipase. Finally, we observed an enriched membrane transport for bile resistance through biliary microbiota. Conclusion β-glucuronidase-producing Streptococcus spp., including Streptococcus dysgalactiae and Streptococcus agalactiae, and Parabacteroides merdae, harbored both uidA and pldA genes and were found to be crucial bacterial species in PGS formation.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhimin Chen
- Department of Hepatobiliary and Pancreatic Surgery, Pingyang Hospital of Wenzhou
Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
12
|
Yao W, He Y, Xie Z, Wang Q, Chen Y, Yu J, Liu X, Xue DX, Liyi W, Hao C. Clostridium scindens promotes gallstone formation by inducing intrahepatic neutrophil extracellular traps through CXCL1 produced by colonic epithelial cells. MICROBIAL CELL (GRAZ, AUSTRIA) 2025; 12:37-52. [PMID: 40309357 PMCID: PMC12041793 DOI: 10.15698/mic2025.03.844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/11/2025] [Accepted: 02/20/2025] [Indexed: 05/02/2025]
Abstract
Cholelithiasis is one of the most common diseases of the biliary system. Neutrophil extracellular traps (NETs) in the liver play an important role in accelerating the formation of gallstones. The upstream mechanism of NETs formation remains unclear. In this study, 16S rRNA sequencing was used to screen the differential gut microbiota in mice with gallstones. Transcriptome sequencing was used to screen the differentially expressed core genes and signalling pathways of Clostridium scindens that acted on human colonic epithelial cells. Western blotting was used to verify the protein expression of TLR2 and the NF-κB pathway. RT-PCR was used to verify the mRNA expression of TLR2, CXCL1 and the NF-κB pathway. ELISA was used to verify CXCL1 expression in the supernatant or portal vein blood of mice. Immunofluorescence was used to detect NETs formation in cocultured neutrophils in vitro or in mouse livers. Clostridium scindens was the key differential strain in the formation of gallstones in mice. After treatment with Clostridium scindens, both in vitro and in vivo, the expression of TLR2 was upregulated, the secretion of CXCL1 was increased by regulating the NF-κB pathway. Finally, the formation of NETs and stones was significantly increased. This study reveals a new mechanism of the gut-liver immune axis in the formation of gallstones. Clostridium scindens acts on colonic epithelial cells through TLR2 to regulate the NF-κB pathway and increase the secretion of CXCL1. CXCL1 enters the liver via the portal vein and increases the formation of NETs in the liver, thereby accelerating gallstone formation.
Collapse
Affiliation(s)
- Wenchao Yao
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
- Equal contribution as a first author
| | - Yuanhang He
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Equal contribution as a first author
| | - Zhihong Xie
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiang Wang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Chen
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Gastrointestinal Surgery Ward I, Yantai Yuhuangding Hospital, the Affiliated Hospital of Qingdao University, Yantai 264000, China
| | - Jingjing Yu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuxu Liu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongbo Xue Xue
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wang Liyi
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chenjun Hao
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
13
|
Liang F, Sun Y, Yang J, Shen Z, Wang G, Zhu J, Zhou C, Xia Y. Gut microbiome is associated with radiotherapy response in lung cancer patients with brain metastases. Front Cell Infect Microbiol 2025; 15:1562831. [PMID: 40129929 PMCID: PMC11931136 DOI: 10.3389/fcimb.2025.1562831] [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: 01/18/2025] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Purpose To investigate the gut microbiome of lung cancer patients with brain metastases undergoing radiotherapy, identify key microorganisms associated with radiotherapy response, and evaluate their potential as biomarkers. Methods and materials This study enrolled 55 newly diagnosed lung cancer patients with brain metastases. Fecal samples were collected before radiotherapy and analyzed by 16S rRNA sequencing to assess the gut microbiome's composition and function. Patients were categorized into response (n=28) and non-response (n=27) groups based on treatment efficacy, and α-diversity, β-diversity, and functional pathways were compared between them. Linear Discriminant Analysis Effect Size was used to identify microbial features associated with treatment efficacy. Logistic regression analyses were performed to evaluate the predictive capacity of clinical and microbial factors for treatment outcomes. Results No significant difference in α-diversity was observed between the groups (P > 0.05), but β-diversity differed significantly (P = 0.036). Twelve characteristic microorganisms were identified in the response group, including g_ Oscillibacter and g_ Blautia, and nine in the non-response group, such as f_ Desulfovibrionaceae and g_ Megamonas. Metabolic pathways associated with treatment response included ketone body metabolism and pathways related to amyotrophic lateral sclerosis. Multivariate analysis identified g_Flavonifractor (odds ratio [OR] = 6.680, P = 0.004), g_Negativibacillus (OR = 3.862, P = 0.014), C-reactive protein (OR = 1.054, P = 0.017), and systemic inflammation response index (OR = 1.367, P = 0.043) as independent predictors of radiotherapy response. The nomogram and microbiome models achieved area under the curve (AUC) values of 0.935 and 0.866, respectively, demonstrating excellent predictive performance. Decision curve analysis further confirmed these models provided significant net benefits across risk thresholds. Conclusions The composition and functional characteristics of the gut microbiome in lung cancer patients with brain metastases prior to radiotherapy are associated with therapeutic response and possess potential as predictive biomarkers. Further studies are warranted to validate these findings.
Collapse
Affiliation(s)
- Fei Liang
- Department of Radiation Oncology, the First People's Hospital of Lianyungang/ Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Yichu Sun
- Department of Radiation Oncology, the First People's Hospital of Lianyungang/ Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Jing Yang
- Department of Radiation Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University/ The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Ziqiang Shen
- Department of Radiation Oncology, the First People's Hospital of Lianyungang/ Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Guangfeng Wang
- Department of Radiation Oncology, the First People's Hospital of Lianyungang/ Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Jiangrui Zhu
- Department of Radiation Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University/ The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Chong Zhou
- Department of Radiation Oncology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Youyou Xia
- Department of Radiation Oncology, the First People's Hospital of Lianyungang/ Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Radiation Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University/ The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| |
Collapse
|
14
|
Peng S, Wei Y, Huang H, Lan C, Zeng Z, Zhu G, Peng T. The mediating role of circulating inflammatory cytokines in causal associations between plasma metabolites and asymptomatic bile duct and cholecyst calculus: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41745. [PMID: 40068083 PMCID: PMC11902928 DOI: 10.1097/md.0000000000041745] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/14/2024] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
Asymptomatic gallbladder and biliary tract calculus may make into symptomatic disease or bring anxiety for patients. The formation of gallstones was associated with genetic risk factors and metabolic abnormalities. Genome-wide association studies (GWAS) data of 1400 plasma metabolites (PMs) and 91 circulating inflammatory cytokines (CICs) were obtained from the GWAS catalog, while the GWAS data of calculus of gallbladder without cholecystitis and calculus of bile duct without cholangitis or cholecystitis were retrieved from the IEU OpenGWAS project. The causalities from PMs or CICs to asymptomatic bile duct or cholecyst calculus were explored by 2-sample Mendelian randomization (MR) analysis. Furthermore, the MR analyses were implemented from the identified PMs to CICs. Following the false discovery rate adjustment, the significant causalities, including 6 CICs and 5 PMs on asymptomatic biliary stone and 5 CICs and 48 PMs on asymptomatic gallstone, were identified. Fibroblast growth factor 19 (FGF-19) and aspartate/mannose ratio were the common protective factors of asymptomatic biliary tract calculus, while Monocyte chemoattractant protein 2 (CCL-2) may serve as a disease-promoting agent. Moreover, Bilirubin degradation product, C17H18N2O4 (1) levels, and Bilirubin (Z,Z)/etiocholanolone glucuronide ratio were associated with FGF-19 level, while aspartate/mannose ratio was related to TNF-related apoptosis-inducing ligand level. Based on MR analysis, we identified the multiple PMs and CICs, especially FGF-19, which may affect the formation of gallbladder and biliary tract calculus. Moreover, the partial CICs could be the downstream mediator of PMs related to asymptomatic gallbladder and biliary tract calculus. These results contributed to supporting previous studies and provided evidence for disease prevention or management.
Collapse
Affiliation(s)
- Shayong Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, P.R. China
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, P.R. China
| | - Yongguang Wei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, P.R. China
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, P.R. China
| | - Huasheng Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, P.R. China
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, P.R. China
| | - Chenlu Lan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, P.R. China
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, P.R. China
| | - Zhiming Zeng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, P.R. China
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, P.R. China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, P.R. China
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, P.R. China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, P.R. China
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, P.R. China
| |
Collapse
|
15
|
Ang TL, Koo SH, Ang D, Tan CK, Wang LM, Wong SH, Chow PKH. Postcholecystectomy Gut Microbiome Changes and the Clinical Impact: A Systematic Review With Narrative Synthesis. J Gastroenterol Hepatol 2025; 40:574-583. [PMID: 39675817 DOI: 10.1111/jgh.16846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Cholecystectomy (CCE) can affect the enterohepatic circulation of bile acids and result in gut microbiome changes. This systematic review aimed to clarify the effect of CCE on gut microbiome composition and its clinical impact. METHOD A systematic search was conducted in PubMed, Web of Science, and Scopus, combining keywords such as "cholecystectomy" or "post-cholecystectomy" with "gut microbiome," "stool microbiome," or "gut dysbiosis." Data were extracted and synthesized using narrative review. Study quality was assessed using the Newcastle-Ottawa scale. RESULTS A total of 1373 articles were screened and 14 studies were selected. Significant but inconsistent microbiome changes were reported. Changes were observed in alpha and beta diversity. At phylum level, an increase in Bacteroides and Ascomycota, decrease in Firmicutes, Actinomycetes, and Basidiomycota, and both increase and decrease in Fusobacteria were reported. At genus level, an increase in Prevotella and a decrease in Faecalibacterium were reported. In post-CCE diarrhea, decreased beta diversity, a decreased F/B ratio, an increase in Prevotella, an increase in Phocaeicola vulgatus, and a decrease in Prevotella copri were noted. For post-CCE syndrome, a higher abundance of Proteobacteria and decreased Firmicutes/Bacteroides (F/B) ratio were reported. A decreased relative abundance of Bifidobacterium longum subsp. longum from controls to CCE without colonic neoplasia to CCE with colonic neoplasia, and an increased abundance of Candida glabrata from controls, to CCE without colonic neoplasia and CCE with colonic neoplasia, were reported. CONCLUSION Patients who underwent CCE had significant gut dysbiosis. However, current studies could not clarify the detailed gut microbial structural and functional changes associated with CCE.
Collapse
Affiliation(s)
- Tiing Leong Ang
- Department of Gastroenterology and Hepatology, Changi General Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Duke-NUS Medical School, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Seok Hwee Koo
- Research Laboratory, Clinical Trials and Research Unit, Changi General Hospital, Singapore
| | - Daphne Ang
- Department of Gastroenterology and Hepatology, Changi General Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Duke-NUS Medical School, Singapore
| | - Chin Kimg Tan
- Department of Gastroenterology and Hepatology, Changi General Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lai Mun Wang
- Department of Gastroenterology and Hepatology, Changi General Hospital, Singapore
- Department of Pathology, Parkway Laboratory Services Ltd, Singapore
| | - Sunny Hei Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Department of Gastroenterology and Hepatology, Tan Tock Seng Hospital, Singapore
| | - Pierce K H Chow
- Duke-NUS Medical School, Singapore
- Department of Hepato-Pancreato-Biliary and Transplant Surgery, National Cancer Center and Singapore General Hospital, Singapore
- Program in Translational and Clinical Research in Liver Cancer, National Cancer Centre Singapore, Singapore
| |
Collapse
|
16
|
Tang J, Xu W, Yu Y, Yin S, Ye BC, Zhou Y. The role of the gut microbial metabolism of sterols and bile acids in human health. Biochimie 2025; 230:43-54. [PMID: 39542125 DOI: 10.1016/j.biochi.2024.11.003] [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: 04/22/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Sterols and bile acids are vital signaling molecules that play key roles in systemic functions, influencing the composition of the human gut microbiota, which maintains a symbiotic relationship with the host. Additionally, gut microbiota-encoded enzymes catalyze the conversion of sterols and bile acids into various metabolites, significantly enhancing their diversity and biological activities. In this review, we focus on the microbial transformations of sterols and bile acids in the gut, summarize the relevant bacteria, genes, and enzymes, and review the relationship between the sterols and bile acids metabolism of gut microbiota and human health. This review contributes to a deeper understanding of the crucial roles of sterols and bile acids metabolism by gut microbiota in human health, offering insights for further investigation into the interactions between gut microbiota and the host.
Collapse
Affiliation(s)
- Jiahui Tang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Wenwu Xu
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yangfan Yu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Shengxiang Yin
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Bang-Ce Ye
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Yunyan Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.
| |
Collapse
|
17
|
Ma M, Xie K, Jin T, Xu F. Moderate and severe depression increase the incidence of cholelithiasis: Results from Mendelian randomization study and the NHANES 2017-March 2020. Soc Psychiatry Psychiatr Epidemiol 2025:10.1007/s00127-025-02843-1. [PMID: 39982470 DOI: 10.1007/s00127-025-02843-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Depression may be a contributing factor to cholelithiasis. However, the exact correlation between cholelithiasis and depression severity remains unclear. METHODS First, a two-sample Mendelian randomization (MR) analysis was performed to validate previous research findings, utilizing separate datasets for major depressive disorder (MDD) and cholelithiasis. The MDD dataset (135,458 cases, 344,901 controls) came from a published GWAS, and cholelithiasis data (19,023 cases, 195,144 controls) were sourced from FinnGen. The primary analytical approach for the MR study was the inverse variance weighting (IVW) method. Second, an observational study based on the National Health and Nutrition Examination Survey (NHANES) was conducted to explore the relationship between the severity of depression and cholelithiasis. 7071 participants were included in the observational study in total. Depression severity (no, mild, moderate, severe) was measured by Patient Health Questionnaire-9 (PHQ-9). Weighted multivariable-adjusted logistic regression was employed to assess the association between depression severity and cholelithiasis. RESULTS In the MR study, the IVW analysis revealed that MDD may increase the risk of cholelithiasis (OR 1.25, 95% CI 1.07-1.45, P = 0.004). The observational study showed that moderate (OR 1.06, 95% CI 1.00-1.11, p = 0.037) and severe (OR 1.07, 95% CI 1.00-1.15, p = 0.044) depression rises the incidence of cholelithiasis. However, no significant association was found between mild depression and cholelithiasis (p = 0.275). CONCLUSIONS Moderate and severe depression might rise the incidence of cholelithiasis, while mild depression may not. Further validation through prospective studies is necessary.
Collapse
Affiliation(s)
- Mingxiu Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning, 110004, China
- Department of General Surgery, The Peoples' Hospital of Liaoning Province, Shenyang, Liaoning, China
| | - Kailing Xie
- Department of General Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning, 110004, China
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Tianqiang Jin
- Department of General Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning, 110004, China.
| | - Feng Xu
- Department of General Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
18
|
Wang C, Jiang T, Ye L, Du M, Liu F. Optimization of the cholesterol gallstone model in C57BL/6 mice and evaluation of Lactobacillus intervention effects. BMC Gastroenterol 2025; 25:56. [PMID: 39910449 PMCID: PMC11800605 DOI: 10.1186/s12876-025-03653-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/29/2025] [Indexed: 02/07/2025] Open
Abstract
Gallstone disease is a common and complex condition, strongly associated with abnormal cholesterol metabolism, changes in bile composition, and impaired gallbladder motility. Recent studies have suggested that the gut microbiota, particularly probiotics like lactic acid bacteria, may play a significant role in the prevention and treatment of cholesterol gallstones. This study aims to optimize the cholesterol gallstone model in C57BL/6 mice and evaluate the effects of Lactobacillus intervention on gallstone formation induced by a high-fat diet. In this study, 8-week-old male C57BL/6 mice were randomly divided into four groups: a high-fat diet + saline group (HF-S), a high-fat diet + probiotic group (HF-P), a normal diet + saline group (ND-S), and a normal diet + probiotic group (ND-P), to assess the effect of probiotics on gallstone formation. The results showed significant differences among the four groups in body weight gain, liver weight, gallstone formation, and histopathology. Based on these preliminary findings, we added two more experimental groups: a 2-week probiotic pretreatment + high-fat diet group (Pre2w-HF) and a 4-week probiotic pretreatment + high-fat diet group (Pre4w-HF), to further investigate the dose-dependence and efficacy of probiotic pretreatment. The results indicated that probiotic intervention significantly reduced the incidence and severity of gallstones induced by a high-fat diet, with the pretreatment groups showing more pronounced effects. Histological analysis also revealed that probiotic intervention reduced inflammation and pathological changes in the liver and gallbladder. This study suggests that probiotics have potential therapeutic value in the prevention and treatment of cholesterol gallstones. Future research should explore the effects of different strains and doses, as well as the underlying mechanisms involved.
Collapse
Affiliation(s)
- Cong Wang
- Department of Gastroenterology, The First Affiliated Hospital of Shihezi University, No. 107 North Second Road, Hongshan Street, Shihezi, 832008, China
| | - Ting Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Shihezi University, No. 107 North Second Road, Hongshan Street, Shihezi, 832008, China
| | - Lvwang Ye
- Department of Gastroenterology, The First Affiliated Hospital of Shihezi University, No. 107 North Second Road, Hongshan Street, Shihezi, 832008, China
| | - Mengyan Du
- Department of Gastroenterology, The First Affiliated Hospital of Shihezi University, No. 107 North Second Road, Hongshan Street, Shihezi, 832008, China
| | - Fang Liu
- Department of Gastroenterology, The First Affiliated Hospital of Shihezi University, No. 107 North Second Road, Hongshan Street, Shihezi, 832008, China.
| |
Collapse
|
19
|
Kazakos E, Kountouras J. Helicobacter pylori Infection and Cholelithiasis. J Gastroenterol Hepatol 2025; 40:548-550. [PMID: 39558474 DOI: 10.1111/jgh.16820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024]
Affiliation(s)
- Evangelos Kazakos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| |
Collapse
|
20
|
Chen P, Hao L, Li Q, Wang H, Chen H, Zhang M, Jia A, Jiang H, Dai H, Zhang R. Identification of targets and comparative study of administration methods for the lipid-lowering effects of fucoidan from Saccharina japonica. Int J Biol Macromol 2025; 290:139102. [PMID: 39716710 DOI: 10.1016/j.ijbiomac.2024.139102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/21/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
The lipid-lowering activity of fucoidan has been widely reported, but the exploration of its mechanisms is relatively limited, and studies on its direct targets are even scarcer. Additionally, it is unclear whether different administration methods affect the lipid-lowering activity of fucoidan. In current study, we used fucoidan derived from Saccharina japonica (SJF) to investigate its targets. The results showed that not only did SJF directly inhibit the Niemann-Pick C1-like 1 (NPC1L1)-mediated cholesterol transport, but it also reduced the solubility of cholesterol in mixed micelles, thereby interfering with the cholesterol uptake. Furthermore, SJF not only directly inhibited the activity of pancreatic triglyceride lipase (PTL), but also interfered with the overall catalytic process facilitated by colipase, thereby reducing the absorption of triglycerides. Moreover, comparative studies on the lipid-lowering activity of SJF administered via different methods demonstrated that dietary supplementation with SJF provided better lipid-lowering effects compared with gavage administration. Our research not only elucidates the targets and mechanisms of SJF but also provides theoretical basis for the selection of administration methods for fucoidan in lipid-lowering therapy.
Collapse
Affiliation(s)
- Pengwei Chen
- Hainan Key Laboratory for Research and Development of Natural Products from Li Folk Medicine, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Li Hao
- Department of Gerontology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 528051, China
| | - Quancai Li
- Marine Biomedical Research Institute of Qingdao, Ocean University of China, Qingdao 266003, China
| | - Hao Wang
- Hainan Key Laboratory for Research and Development of Natural Products from Li Folk Medicine, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Huiqin Chen
- Hainan Key Laboratory for Research and Development of Natural Products from Li Folk Medicine, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Miao Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Ang Jia
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China.
| | - Haofu Dai
- Hainan Key Laboratory for Research and Development of Natural Products from Li Folk Medicine, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China.
| | - Renshuai Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
21
|
Liu Y, Li H, Sun T, Sun G, Jiang B, Liu M, Wang Q, Li T, Cao J, Zhao L, Xiao F, Zhao F, Cui H. Gut microbiome and metabolome characteristics of patients with cholesterol gallstones suggest the preventive potential of prebiotics. IMETA 2025; 4:e70000. [PMID: 40027485 PMCID: PMC11865347 DOI: 10.1002/imt2.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 03/05/2025]
Abstract
Cholesterol gallstones (CGS) still lack effective noninvasive treatment. The etiology of experimentally proven cholesterol stones remains underexplored. This cross-sectional study aims to comprehensively evaluate potential biomarkers in patients with gallstones and assess the effects of microbiome-targeted interventions in mice. Microbiome taxonomic profiling was conducted on 191 samples via V3-V4 16S rRNA sequencing. Next, 60 samples (30 age- and sex-matched CGS patients and 30 controls) were selected for metagenomic sequencing and fecal metabolite profiling via liquid chromatography-mass spectrometry. Microbiome and metabolite characterizations were performed to identify potential biomarkers for CGS. Eight-week-old male C57BL/6J mice were given a lithogenic diet for 8 weeks to promote gallstone development. The causal relationship was examined through monocolonization in antibiotics-treated mice. The effects of short-chain fatty acids such as sodium butyrate, sodium acetate (NaA), sodium propionate, and fructooligosaccharides (FOS) on lithogenic diet-induced gallstones were investigated in mice. Gut microbiota and metabolites exhibited distinct characteristics, and selected biomarkers demonstrated good diagnostic performance in distinguishing CGS patients from healthy controls. Multi-omics data indicated associations between CGS and pathways involving butanoate and propanoate metabolism, fatty acid biosynthesis and degradation pathways, taurine and hypotaurine metabolism, and glyoxylate and dicarboxylate metabolism. The incidence of gallstones was significantly higher in the Clostridium glycyrrhizinilyticum group compared to the control group in mice. The grade of experimental gallstones in control mice was significantly higher than in mice treated with NaA and FOS. FOS could completely inhibit the formation of gallstones in mice. This study characterized gut microbiome and metabolome alterations in CGS. C. glycyrrhizinilyticum contributed to gallstone formation in mice. Supplementing with FOS could serve as a potential approach for managing CGS by altering the composition and functionality of gut microbiota.
Collapse
Affiliation(s)
- Ye Liu
- Peking University Fifth School of Clinical MedicineBeijing Hospital, National Center of GerontologyBeijingChina
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Hexin Li
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Tianhan Sun
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Gaoyuan Sun
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Boyue Jiang
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Meilan Liu
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Qing Wang
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Tong Li
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Jianfu Cao
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Li Zhao
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Fei Xiao
- Peking University Fifth School of Clinical MedicineBeijing Hospital, National Center of GerontologyBeijingChina
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Fangqing Zhao
- Institute of Zoology, Chinese Academy of SciencesBeijingChina
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, Beijing Institutes of Life ScienceChinese Academy of SciencesBeijingChina
| | - Hongyuan Cui
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
22
|
Ren G, Fan Y, Zhong R, Zou G, Huang X, Zhang Y. Relationship between mucin gene polymorphisms and different types of gallbladder stones. BMC Med Genomics 2025; 18:22. [PMID: 39885433 PMCID: PMC11783967 DOI: 10.1186/s12920-025-02090-y] [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: 06/22/2024] [Accepted: 01/21/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Gallstones, a common surgical condition globally, affect around 20% of patients. The development of gallstones is linked to abnormal cholesterol and bilirubin metabolism, reduced gallbladder function, insulin resistance, biliary infections, and genetic factors. In addition to these factors, research has shown that mucins play a role in gallstone formation. This study aims to explore the connection between different types of gallstones and mucin gene polymorphisms. METHODS For this purpose, a total of 121 patients with gallbladder stones PNS and 107 patients with healthy controls PNS were enrolled in this case-control study. One SNPs (rs4072037) of MUC1 gene、 three SNPs (rs2856111、rs41532344、rs41349846) of MUC2 gene、four SNPs (rs712005、rs2246980、rs2258447、rs2259292) of MUC4 gene、seven SNPs (rs28415193、rs56047977、rs2037089、rs2075854、rs3829224、rs2672785、rs2735709) of MUC5 gene、eight SNPs (rs10902268、rs61869016、rs573849895、rs59257210、rs7396383、rs74644072、rs7481521、rs9704308) of MUC6 gene、five SNPs (rs10229731、rs73168398、rs4729655、rs55903219、rs74974199) of MUC17 gene. We amplified SNP sites by polymerase chain reaction (PCR) using specific primer sets followed by DNA sequencing. RESULTS The frequencies of MUC2 rs2856111 C/T genotype (OR = 3.81, 95%CI: 1.06-13.68) was higher than the control group. MUC17 rs10229731 A/C genotype (OR = 0.33, 95%CI: 0.12-0.95), rs73168398 G/A genotype (OR = 0.26, 95%CI: 0.07-0.98), MUC6 rs10902268 G/A genotype (OR = 0.40, 95%CI: 0.17-0.95) at lower frequencies than controls. The frequencies of MUC2 rs41532344 T allele (OR = 2.55, 95%CI: 1.06-6.13), MUC4 rs712005 G allele (OR = 2.51, 95%CI: 1.20-5.22), MUC5B rs2037089 C allele (OR = 3.54, 95%CI: 1.14-11.01) and MUC5AC rs28415193 G allele (OR = 1.77, 95%CI: 1.02-3.07) were higher than the control group. MUC6 rs10902268 A allele (OR = 0.004, 95%CI: 0.00-0.27), rs61869016 C allele (OR = 0.07, 95%CI: 0.01-0.63) at lower frequencies than controls. CONCLUSIONS Polymorphisms in the mucin gene were linked to the formation of gallbladder stones. The MUC4 rs712005 G allele, MUC5B rs2037089 C allele, MUC2 rs41532344 T allele and MUC5AC rs28415193 G allele were found to predispose individuals to the development of the disease. MUC6 rs10902268 A allele and rs61869016 C allele were identified as protective factors. Meanwhile, MUC2 rs2856111 CT genotype was found to predispose individuals to the development of the disease. MUC17 rs10229731 AC genotype, rs73168398 GA genotype and MUC6 rs10902268 GA genotype were identified as protective factors.
Collapse
Affiliation(s)
- Gongqing Ren
- The Second Clinical Medical College of Jinan University, Department of Hepatobiliary Pancreatic Surgery, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Yongmao Fan
- Department of Thoracic Surgery, Shenzhen Luohu Hospital Group Luohu People's Hospital, Shenzhen, China
| | - Ruizi Zhong
- Breast Surgery, Shenzhen Futian District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Gang Zou
- Department of Burns and Plastic Surgery, Shenzhen People's Hospital, Shenzhen, China
| | - Xiaojun Huang
- Department of Hepatobiliary Pancreatic Surgery, Shenzhen People's Hospital, No.1017 Dongmen North Road, Shenzhen, 518020, Guangdong Province, China
| | - Yue Zhang
- Department of Hepatobiliary Pancreatic Surgery, Shenzhen People's Hospital, No.1017 Dongmen North Road, Shenzhen, 518020, Guangdong Province, China.
| |
Collapse
|
23
|
Li X, Yin X, Xu J, Geng L, Liu Z. Relationship between Abnormal Lipid Metabolism and Gallstone Formation. THE KOREAN JOURNAL OF GASTROENTEROLOGY = TAEHAN SOHWAGI HAKHOE CHI 2025; 85:11-21. [PMID: 39849808 DOI: 10.4166/kjg.2024.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 01/25/2025]
Abstract
Cholelithiasis is a common biliary system disease with a high incidence worldwide. Abnormal lipid metabolism has been shown to play a key role in the mechanism of gallstones. Therefore, recent research literature on the genes, proteins, and molecular substances involved in lipid metabolism during the pathogenesis of gallstones has been conducted. This study aimed to determine the role of lipid metabolism in the pathogenesis of gallstones and provide insights for future studies using previous research in genomics, metabolomics, transcriptomics, and other fields.
Collapse
Affiliation(s)
- Xiang Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaodan Yin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lei Geng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhengtao Liu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Chen J, Pan Q, Lu L, Huang X, Wang S, Liu X, Lun J, Xu X, Su H, Guo F, Yang L, You L, Xiao H, Luo W, Liu HF, Pan Q. Atg5 deficiency in basophils improves metabolism in lupus mice by regulating gut microbiota dysbiosis. Cell Commun Signal 2025; 23:40. [PMID: 39844180 PMCID: PMC11756211 DOI: 10.1186/s12964-025-02041-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/12/2025] [Indexed: 01/24/2025] Open
Abstract
Autophagic activation in immune cells, gut microbiota dysbiosis, and metabolic abnormalities have been reported separately as characteristics of systemic lupus erythematosus (SLE). Elucidating the crosstalk among the immune system, commensal microbiota, and metabolites is crucial to understanding the pathogenesis of autoimmune diseases. Emerging evidence shows that basophil activation plays a critical role in the pathogenesis of SLE; however, the underlying mechanisms remain largely unknown. Here, we investigated the effects of autophagic inhibition on the pathogenesis of basophils in SLE using Autophagy-related gene 5 (Atg5) knockout (Atg5-/-) as an autophagic inhibitor. Specifically, we knocked out basophilic Atg5 in vivo to investigate its impact on lupus metabolism. Furthermore, Atg5-/- basophils were transferred to basophil-depleted MRL/MpJ-Faslpr (MRL/lpr) mice to study their effect on disease metabolism. Metagenomic and targeted metabolomic sequencing results indicated considerable reduction in the levels of plasma autoantibodies and inflammatory cytokines in the Atg5-/- basophil transfer group compared with that in the control group. Transplanting Atg5-/- basophils improved the gut microbiota balance in MRL/lpr mice, increasing the abundance of beneficial bacteria, such as Ligilactobacillus murinus and Faecalitalea rodentium, and reducing that of potentially pathogenic bacteria such as Phocaeicola salanitronis. The transplantation of Atg5-deficient basophils improved lupus symptoms by modulating lipid and amino acid metabolism. This improvement was linked to changes in the gut microbiota, particularly an increase in Ligilactobacillus murinus and Faecalitalea rodentium populations. These microbial shifts are believed to promote the production of beneficial metabolites, such as γ-linolenic acid and oleoyl-1-palmitoyl-sn-glycero-3-phosphocholine, while reducing the levels of harmful metabolites such as arginine. These alterations in the metabolic profile contribute to the alleviation of lupus symptoms. Collectively, these findings reveal a novel role of basophil autophagy in SLE, highlighting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Jiaxuan Chen
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Quanren Pan
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lu Lu
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaorong Huang
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shuting Wang
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaoxian Liu
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaqi Lun
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaowei Xu
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hongyong Su
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fengbiao Guo
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lawei Yang
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Liuyong You
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Wenying Luo
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua-Feng Liu
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Qingjun Pan
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
25
|
Yu Y, Fu R, Jin C, Han L, Gao H, Fu B, Qi M, Li Q, Leng J. Multi-Omics Insights into Rumen Microbiota and Metabolite Interactions Regulating Milk Fat Synthesis in Buffaloes. Animals (Basel) 2025; 15:248. [PMID: 39858248 PMCID: PMC11758634 DOI: 10.3390/ani15020248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
The present study was conducted to analyze the correlation between the milk fat content of Binglangjiang buffaloes and their microbial and host metabolites. The 10 buffaloes with the highest milk fat content (HF, 5.60 ± 0.61%) and the 10 with the lowest milk fat content (LF, 1.49 ± 0.13%) were selected. Their rumen fluid and plasma were collected for rumen microbiota and metabolome analysis. The results showed that the rumen bacteria abundance of Synergistota, Quinella, Selenomonas, and Fretibacterium was significantly higher in the HF buffaloes. The abundance of 14 rumen fungi, including Candida, Talaromyces, Cyrenella, and Stilbella, was significantly higher in the HF buffaloes. The analysis of the metabolites in the rumen and plasma showed that several metabolites differed between the HF and LF buffaloes. A total of 68 and 42 differential metabolites were identified in the rumen and plasma, respectively. By clustering these differential metabolites, most of those clustered in the HF group were lipid and lipid-like molecules such as secoeremopetasitolide B, lucidenic acid J LysoPE (0:0/18:2 (9Z, 12Z)), and 5-tetradecenoic acid. Spearman's rank correlations showed that Quinella, Fretibacterium, Selenomonas, Cyrenella, and Stilbella were significantly positively correlated with the metabolites of the lipids and lipid-like molecules in the rumen and plasma. The results suggest that rumen microbiota such as Quinella, Fretibacterium, Selenomonas, and Cyrenella may regulate milk fat synthesis by influencing the lipid metabolites in the rumen and plasma. In addition, the combined analysis of the rumen microbiota and host metabolites may provide a fundamental understanding of the role of the microbiota and host in regulating milk fat synthesis.
Collapse
Affiliation(s)
- Ye Yu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (Y.Y.); (R.F.); (C.J.); (L.H.); (H.G.); (B.F.); (M.Q.); (Q.L.)
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Runqi Fu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (Y.Y.); (R.F.); (C.J.); (L.H.); (H.G.); (B.F.); (M.Q.); (Q.L.)
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Chunjia Jin
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (Y.Y.); (R.F.); (C.J.); (L.H.); (H.G.); (B.F.); (M.Q.); (Q.L.)
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Lin Han
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (Y.Y.); (R.F.); (C.J.); (L.H.); (H.G.); (B.F.); (M.Q.); (Q.L.)
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Huan Gao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (Y.Y.); (R.F.); (C.J.); (L.H.); (H.G.); (B.F.); (M.Q.); (Q.L.)
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Binlong Fu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (Y.Y.); (R.F.); (C.J.); (L.H.); (H.G.); (B.F.); (M.Q.); (Q.L.)
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Min Qi
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (Y.Y.); (R.F.); (C.J.); (L.H.); (H.G.); (B.F.); (M.Q.); (Q.L.)
- Yunnan Animal Husbandry Station, Kunming 650224, China
| | - Qian Li
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (Y.Y.); (R.F.); (C.J.); (L.H.); (H.G.); (B.F.); (M.Q.); (Q.L.)
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Jing Leng
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (Y.Y.); (R.F.); (C.J.); (L.H.); (H.G.); (B.F.); (M.Q.); (Q.L.)
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
26
|
Zhang F, Wang Y, Wang M, Tan C, Huang S, Mou H, Wu K, Peng L, Fang Z, Tian Y, Sheng J, Zhao C. Structural characteristics and nonvolatile metabolites of theabrownins and their impact on intestinal microbiota in high-fat-diet-fed mice. Food Chem 2025; 463:141317. [PMID: 39332361 DOI: 10.1016/j.foodchem.2024.141317] [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: 03/15/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/29/2024]
Abstract
This study prepared enzymatic theabrownins (TBs-e), alkaline theabrownins (TBs-a), and Pu-erh tea theabrownins (TBs-f), and investigated whether different preparation processes affected the structures, nonvolatile metabolites, and biofunctional activities of TBs. Structural characterization revealed that TBs were polymeric phenolic compounds rich in hydroxyl and carboxyl groups. Nontargeted metabolomics revealed that amino acids were the primary nonvolatile metabolites in TBs-e and TBs-a, accounting for over 70 % of the total nonvolatile content. TBs-f contained more polyphenols, caffeine, and flavonoids, accounting for 14.2 %, 3.9 %, and 0.8 % of total nonvolatile content, respectively. In vivo, at 560 mg/kg body weight, TBs-f were associated with regulation of blood glucose and lipid concentrations in mice. Moreover, 16S rRNA indicated that at 1120 mg/kg body weight, TBs-a were associated with increased numbers of microbiota linked with hypolipidemic activity. This study explores the impacts of different preparation processes on TBs and provides a theoretical foundation for the understanding of TBs.
Collapse
Affiliation(s)
- Feng Zhang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Ya Wang
- College of Science, Yunnan Agricultural University, Kunming 650201, China
| | - Mingming Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Chunlei Tan
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Si Huang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Hongyu Mou
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Kuan Wu
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Lei Peng
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Zhongqi Fang
- Boao Yiling Life Care Center, Qionghai 571400, China
| | - Yang Tian
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China; Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China; PuEr University, PuEr 665000, China
| | - Jun Sheng
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China; Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China
| | - Cunchao Zhao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Yunnan Plateau Characteristic Agricultural Industry Research Institute, Kunming 650201, China; Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China; Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
27
|
Chen L, Ye Z, Li J, Wang L, Chen Y, Yu M, Han J, Huang J, Li D, Lv Y, Xiong K, Tian D, Liao J, Seidler U, Xiao F. Gut bacteria Prevotellaceae related lithocholic acid metabolism promotes colonic inflammation. J Transl Med 2025; 23:55. [PMID: 39806416 PMCID: PMC11727794 DOI: 10.1186/s12967-024-05873-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The conversion of primary bile acids to secondary bile acids by the gut microbiota has been implicated in colonic inflammation. This study investigated the role of gut microbiota related bile acid metabolism in colonic inflammation in both patients with inflammatory bowel disease (IBD) and a murine model of dextran sulfate sodium (DSS)-induced colitis. METHODS Bile acids in fecal samples from patients with IBD and DSS-induced colitis mice, with and without antibiotic treatment, were analyzed using ultraperformance liquid chromatography-mass spectrometry (UPLC-MS). The composition of the microbiota in fecal samples from IBD patients and DSS-colitis mice was characterized via Illumina MiSeq sequencing of the bacterial 16S rRNA gene V3-V4 region. Metagenomic profiling further identified metabolism-related gene signatures in stool samples from DSS-colitis mice. Histological analysis, quantitative PCR (qPCR) and Western Blotting were conducted on colonic samples from DSS-induced colitis mice to assess colonic inflammation, mucosal barrier integrity, and associated signaling pathways. The multivariate analysis of bile acids was conducted using Soft Independent Modelling of Class Analogy (SIMCA, Umetrics, Sweden). The relation between the relative abundance of specific phyla/genera and bile acid concentration was assess through Spearman's correlation analyses. Finally, lithocholic acid (LCA), the key bile acid, was administered via gavage to evaluate its effect on colonic inflammation and mucosal barrier integrity. RESULTS In patients with IBD, the composition of colonic bile acids and gut microbiota was altered. Moreover, changes in the gut microbiota further modulate the composition of bile acids in the intestine. As the gut microbiota continues to shift, the bile acid profile undergoes additional alterations. The aforementioned alterations were also observed in mice with DSS-induced colitis. The study revealed a correlation between dysbiosis of the gut microbiota and modifications in the profile of colonic bile acids, notably LCA observed in both patients with IBD and mice with DSS-induced colitis. Through multivariate analysis, LCA was identified as the key bile acid that significantly affects colonic inflammation and the integrity of mucosal barrier. Subsequent experiments confirmed that LCA supplementation effectively mitigated the inhibitory effects of gut microbiota on colitis progression in mice, primarily through the activation of the sphingosine-1-phosphate receptor 2 (S1PR2)/NF-κB p65 signaling pathway. Analysis of the microbiome and metagenomic data revealed changes in the gut microbiota, notably an increased abundance of an unclassified genus within the family Prevotellaceae in DSS-induced colitis mice. Furthermore, a positive correlation was observed between the relative abundance of Prevotellaceae and bile acid biosynthesis pathways, as well as colonic LCA level. CONCLUSIONS These findings suggest that LCA and its positively correlated gut bacteria, Prevotellaceae, are closely associated with intestinal inflammation. Targeting colonic inflammation may involve inhibiting LCA and members of the Prevotellaceae family as potential therapeutic strategies.
Collapse
Affiliation(s)
- Liping Chen
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Zhenghao Ye
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Junhua Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijia Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Yu Chen
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Meiping Yu
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Jian Han
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Jiangeng Huang
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongyan Li
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongling Lv
- Meiyitian Biopharmaceutical (Wuhan) Ltd., Wuhan, China
| | - Kai Xiong
- Meiyitian Biopharmaceutical (Wuhan) Ltd., Wuhan, China
| | - De'an Tian
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Jiazhi Liao
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China
| | - Ursula Seidler
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Fang Xiao
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China.
| |
Collapse
|
28
|
Wei Y, Mao H, Liu Q, Fang W, Zhang T, Xu Y, Zhang W, Chen B, Zheng Y, Hu X. Lipid metabolism and microbial regulation analyses provide insights into the energy-saving strategies of hibernating snakes. Commun Biol 2025; 8:45. [PMID: 39800781 PMCID: PMC11725596 DOI: 10.1038/s42003-025-07493-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Hibernation is a necessary means for animals to maintain survival while coping with low temperatures and food shortages. While most studies have largely focused on mammalian hibernation, its reptilian equivalent has been less studied. In order to provide insights into the energy metabolism and potential microbial regulatory mechanisms in hibernating snakes, the serum, liver, gut content samples were measured by multi-omic methods. Here we show the active snakes have more vigorous lipid metabolism, whereas snakes in hibernation groups have higher sphingolipid metabolism. Furthermore, the results indicate that the potential energy supply pathway was gluconeogenesis. Microbial analysis reveals that Proteobacteria and Firmicutes showed dynamic changes with the transformation among active, pre-hibernation and hibernation periods. The correlation analysis reveals the potential role of Romboutsia, Providencia and Vagococcus in regulating above metabolism by producing certain metabolites. The results advance the understanding of the complex energy-saving strategy in hibernating poikilotherms.
Collapse
Affiliation(s)
- Yuting Wei
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Huirong Mao
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Qiuhong Liu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Wenjie Fang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Tianxiang Zhang
- Institute of Wildlife Conservation, Jiangxi Academy of Forestry, Nanchang, China
| | - Yongtao Xu
- Jiangxi Provincial Key Laboratory of Conservation Biology, Jiangxi Agricultural University, Nanchang, China
- College of Forestry, Jiangxi Agricultural University, Nanchang, China
| | - Weiwei Zhang
- Jiangxi Provincial Key Laboratory of Conservation Biology, Jiangxi Agricultural University, Nanchang, China
- College of Forestry, Jiangxi Agricultural University, Nanchang, China
| | - Biao Chen
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Yunlin Zheng
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Xiaolong Hu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China.
- Jiangxi Provincial Key Laboratory of Conservation Biology, Jiangxi Agricultural University, Nanchang, China.
| |
Collapse
|
29
|
Zheng L, Ye ZY, Ma JJ. Effect of cholesterol metabolism on hepatolithiasis. World J Gastroenterol 2025; 31:99960. [PMID: 39777239 PMCID: PMC11684189 DOI: 10.3748/wjg.v31.i1.99960] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/03/2024] [Accepted: 11/05/2024] [Indexed: 12/09/2024] Open
Abstract
Surgical intervention is currently the primary treatment for hepatolithiasis; however, some patients still experience residual stones and high recurrence rates after surgery. Cholesterol metabolism seems to play an important role in hepatolithiasis pathogenesis. A high cholesterol diet is one of the significant reasons for the increasing incidence of hepatolithiasis. Therefore, regular diet and appropriate medical intervention are crucial measures to prevent hepatolithiasis and reduce recurrence rate after surgery. Reducing dietary cholesterol and drugs that increase cholesterol stone solubility are key therapeutic approaches in treating hepatolithiasis. This article discusses the cholesterol metabolic pathways related to the pathogenesis of hepatolithiasis, as well as food intake and targeted therapeutic drugs.
Collapse
Affiliation(s)
- Lin Zheng
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215 Heping West Road, Shijiazhuang 050000, Hebei Province, China
| | - Zi-Yu Ye
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215 Heping West Road, Shijiazhuang 050000, Hebei Province, China
| | - Jun-Ji Ma
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215 Heping West Road, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
30
|
Mi N, Yang M, Wei L, Nie P, Zhan S, Nguyen LH, Smith FG, Acharjee A, Liu X, Huang J, Xia B, Yuan J, Meng W. Gallstone Disease Is Associated With an Increased Risk of Inflammatory Bowel Disease: Results From 3 Prospective Cohort Studies. Am J Gastroenterol 2025; 120:204-212. [PMID: 39364876 DOI: 10.14309/ajg.0000000000003111] [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: 06/27/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION Gallstone diseases affect intestinal inflammation, bile flow, and gut microbiota, which in turn may increase the risk of inflammatory bowel disease (IBD). However, epidemiological studies exploring the associations between gallstone diseases and subsequent IBD risk have been limited. METHODS This is a combined analysis of 3 prospective cohort studies (Nurses' Health Study, Nurses' Health Study II, and UK Biobank) and replicated in a case-control study (Chinese IBD Etiology Study). We evaluated the hazard ratios (HRs)/odds ratios (ORs) between gallstone diseases with IBD risk by Cox logistic regression or conditional logistic regression, adjusting for demographic characteristics, lifestyles, comorbidities, and medication usage. RESULTS We identified 3,480 cases of IBD over 2,127,471 person-years of follow-up in the 3 cohort studies. The participants with gallstone disease had a 38% increase in the risk of IBD (HR 1.38, 95% confidence intervals [CI] 1.21-1.59), 68% increase in Crohn's disease (HR 1.68, 95% CI 1.38-2.06), and 24% increase in ulcerative colitis (HR 1.24, 95% CI 1.03-1.49). In Chinese IBD Etiology Study, we found even larger magnitude of effects between gallstone diseases and IBD risk (IBD: OR 3.03, 95% CI 2.32-3.97; Crohn's disease: OR 5.31; 95% CI 3.71-7.60; ulcerative colitis: OR 1.49; 95% CI 1.07-2.06). There were no major differences in the estimated associations between the presence of unremoved gallstones and prior cholecystectomy with IBD risk. DISCUSSION Gallstone disease was linked to an increased risk of IBD and its subtypes, independent of traditional risk factors. Further research is needed to confirm these associations and clarify the underlying biological mechanisms.
Collapse
Affiliation(s)
- Ningning Mi
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
- Department of Epidemiology and Biostatistics, Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Man Yang
- Guangdong Provincial Key Laboratory of Gastroenterology, Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Chinese Health RIsk MAnagement Collaboration (CHRIMAC), Shenzhen, Guangdong, China
| | - Lina Wei
- Digestive System Department, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Peng Nie
- Department of General Surgery, Gansu Wuwei Tumour Hospital, Wuwei, Gansu, China
| | - Shukai Zhan
- Department of Gastroenterology, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Long H Nguyen
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Fang Gao Smith
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- University Hospitals Birmingham NHS Foundation Trust and Health Data Research UK Midlands, Birmingham, UK
| | - Animesh Acharjee
- Institute of Translational Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- NIHR Surgical Reconstruction and Microbiology Research Centre, Birmingham, UK
| | - Xudong Liu
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Junjie Huang
- Faculty of Medicine, The Jockey Club School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bin Xia
- Department of Epidemiology and Biostatistics, Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Chinese Health RIsk MAnagement Collaboration (CHRIMAC), Shenzhen, Guangdong, China
| | - Jinqiu Yuan
- Department of Epidemiology and Biostatistics, Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Chinese Health RIsk MAnagement Collaboration (CHRIMAC), Shenzhen, Guangdong, China
| | - Wenbo Meng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
31
|
Qi Y, Ma Y, Duan G. Pharmacological Mechanisms of Bile Acids Targeting the Farnesoid X Receptor. Int J Mol Sci 2024; 25:13656. [PMID: 39769418 PMCID: PMC11727972 DOI: 10.3390/ijms252413656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025] Open
Abstract
Bile acids (BAs), a category of amphiphilic metabolites synthesized by liver cells and released into the intestine via the bile duct, serve a vital role in the emulsification of ingested fats during the digestive process. Beyond their conventional emulsifying function, BAs, with their diverse structures, also act as significant hormones within the body. They are pivotal in facilitating nutrient absorption by interacting with the farnesoid X receptor (FXR), and they serve as key regulators of lipid and glucose metabolism, as well as immune system balance. Consequently, BAs contribute to the metabolism of glucose and lipids, enhance the digestion and absorption of lipids, and maintain the equilibrium of the bile pool. Their actions are instrumental in addressing obesity, managing cholestasis, and treating diabetes, and are involved in the onset and progression of cancer. This paper presents an updated systematic review of the pharmacological mechanisms by which BAs target the FXR, incorporating recent findings and discussing their signaling pathways in the context of novel research, including their distinct roles in various disease states and populations. The aim is to provide a theoretical foundation for the continued research and clinical application of BAs.
Collapse
Affiliation(s)
- Youchao Qi
- College of Agriculture and Animal Husbandry, Qinghai University, Xining 810016, China;
- Academy of Agriculture and Forestry Sciences, Qinghai University, Xining 810016, China
- Qinghai Plateau Key Laboratory of Tree Genetics and Breeding, Xining 810016, China
- Key Laboratory of Medicinal Animal and Plant Resources of Qinghai Tibetan Plateau, Qinghai Normal University, Xining 810008, China;
- Academy of Plateau Science and Sustainability, Qinghai Normal University, Xining 810008, China
| | - Yonggui Ma
- Key Laboratory of Medicinal Animal and Plant Resources of Qinghai Tibetan Plateau, Qinghai Normal University, Xining 810008, China;
- Academy of Plateau Science and Sustainability, Qinghai Normal University, Xining 810008, China
| | - Guozhen Duan
- Academy of Agriculture and Forestry Sciences, Qinghai University, Xining 810016, China
- Qinghai Plateau Key Laboratory of Tree Genetics and Breeding, Xining 810016, China
| |
Collapse
|
32
|
Zheng X, Yan Y, Li X, Liu M, Zhao X, He J, Zhuang X. Microbial characteristics of bile in gallstone patients: a comprehensive analysis of 9,939 cases. Front Microbiol 2024; 15:1481112. [PMID: 39749136 PMCID: PMC11693992 DOI: 10.3389/fmicb.2024.1481112] [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: 08/26/2024] [Accepted: 12/04/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction The exact triggers of gallstone formation remain incompletely understood, but research indicates that microbial infection is a significant factor and can interfere with treatment. There is no consensus on the bile microbial culture profiles in previous studies, and determining the microbial profile could aid in targeted prevention and treatment. The primary aim of this study is to investigate the differences in microbial communities cultured from bile specimens of patients with gallstones. Methods We collected the clinical characteristics and bile microbial status of 9,939 gallstone patients. Statistical analysis was employed to assess the relationship between microbes and clinical features, and a random forest model was utilized to predict recurrence. Results Results showed a higher proportion of females among patients, with the age group of 60-74 years being the most prevalent. The most common type of gallstone was solitary gallbladder stones. A total of 76 microbes were cultured from 5,153 patients, with Escherichia coli, Klebsiella pneumoniae, and Enterococcus faecalis being the most frequently identified. Significant differences in microbial diversity and positive detection rates were observed across different age groups, types of gallstones, and recurrence status. Positive frequencies of E. coli, Enterococcus faecium, and K. pneumoniae varied significantly by age group and gallstone type. The microbial diversity in the recurrence group was significantly lower compared to the non-recurrence group. The recurrence rate was significantly higher in the group with single microbial species compared to those with no microbes or multiple microbes. For the recurrence group, there were significant differences in the frequencies of seven microbes (Aeromonas hydrophila, Enterococcus casseliflavus, Enterococcus faecium, E. coli, K. pneumoniae, Proteus mirabilis, Pseudomonas aeruginosa) before and after recurrence, with these microbes appearing in a higher number of patients after recurrence. Regression analysis identified patient age, stone size, diabetes, venous thrombosis, liver cirrhosis, malignancy, coronary heart disease, and the number of microbial species as important predictors of recurrence. A random forest model constructed using these variables demonstrated good performance and high predictive ability (ROC-AUC = 0.862). Discussion These findings highlight the significant role of microbial communities in gallstone formation and recurrence. Furthermore, the identified predictors of recurrence, including clinical factors and microbial diversity, may help develop personalized prevention and recurrence strategies for gallstone patients.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Shandong University, Jinan, Shandong, China
| | - Yunjun Yan
- Jinan Dian Medical Laboratory CO., LTD, Jinan, China
| | - Xin Li
- Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Mimin Liu
- Jinan Dian Medical Laboratory CO., LTD, Jinan, China
| | - Xiaoyue Zhao
- Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jing He
- Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xuewei Zhuang
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Shandong University, Jinan, Shandong, China
- Jinan Key Laboratory for Precision Medicine, Jinan, Shandong, China
| |
Collapse
|
33
|
Zhang T, Hasegawa Y, Waldor MK. Enteric bacterial infection stimulates remodelling of bile metabolites to promote intestinal homeostasis. Nat Microbiol 2024; 9:3376-3390. [PMID: 39567665 PMCID: PMC11602723 DOI: 10.1038/s41564-024-01862-z] [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: 10/26/2023] [Accepted: 10/15/2024] [Indexed: 11/22/2024]
Abstract
The liver makes bile, an aqueous solution critical for fat absorption, which is secreted into the duodenum. Despite extensive studies on bile salts, other components of bile are less well characterized. Here we used global metabolomic analysis on bile from specific-pathogen-free, germ-free, Citrobacter rodentium-infected or Listeria monocytogenes-infected mice and identified a metabolome of 812 metabolites that were altered by both microbiota and enteric infection. Hepatic transcriptomics identified enteric-infection-triggered pathways that probably underlie bile remodelling. Enteric infection increased levels of four dicarboxylates in bile, including itaconate. Analysis of Acod1-/- mice indicated that increased itaconate also increased tuft cell abundance, altered microbiota composition and function as detected by metagenomic analysis, and modulated host defence, leading to reduced Vibrio cholerae colonization. Our data suggest that enteric-infection-associated signals are relayed between the intestine and liver and induce transcriptional programmes that shape the bile metabolome, modifying the immunomodulatory and host defence functions of bile.
Collapse
Affiliation(s)
- Ting Zhang
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Yuko Hasegawa
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
34
|
Liu X, Feng Z, Zhang F, Wang B, Wei Z, Liao N, Zhang M, Liang J, Wang L. Causal effects of gut microbiota on gout and hyperuricemia: insights from genome-wide Mendelian randomization, RNA-sequencing, 16S rRNA sequencing, and metabolomes. Biosci Rep 2024; 44:BSR20240595. [PMID: 39492788 PMCID: PMC11598824 DOI: 10.1042/bsr20240595] [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: 05/08/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND This study investigated the causal relationship between gut microbiota (GM), serum metabolome, and host transcriptome in the development of gout and hyperuricemia (HUA) using genome-wide association studies (GWAS) data and HUA mouse model experiments. METHODS Mendelian randomization (MR) analysis of GWAS summary statistics was performed using an inverse variance weighted (IVW) approach to determine or predict the causal role of the GM on gout. The HUA mouse model was used to characterize changes in the gut microbiome, host metabolome, and host kidney transcriptome by integrating cecal 16S rRNA sequencing, untargeted serum metabolomics, and host mRNA sequencing. RESULTS Our analysis demonstrated causal effects of seven GM taxa on gout, including genera of Ruminococcus, Odoribacter, and Bacteroides. Thirty eight immune cell traits were associated with gout. Dysbiosis of Dubosiella, Lactobacillus, Bacteroides, Alloprevotella, and Lachnospiraceae_NK4A136_group genera were associated with changes in the serum metabolites and kidney transcriptome of the HUA model mice. The changes in the gut microbiome of the HUA model mice correlated significantly with alterations in the levels of serum metabolites such as taurodeoxycholic acid, phenylacetylglycine, vanylglycol, methyl hexadecanoic acid, carnosol, 6-aminopenicillanic acid, sphinganine, p-hydroxyphenylacetic acid, pyridoxamine, and de-o-methylsterigmatocystin, and expression of kidney genes such as CNDP2, SELENOP, TTR, CAR3, SLC12A3, SCD1, PIGR, CD74, MFSD4B5, and NAPSA. CONCLUSION Our study demonstrated a causal relationship between GM, immune cells, and gout. HUA development involved alterations in the vitamin B6 metabolism because of GM dysbiosis that resulted in altered pyridoxamine and pyridoxal levels, dysregulated sphingolipid metabolism, and excessive inflammation.
Collapse
Affiliation(s)
- Xia Liu
- Medical College, Guangxi University, Nanning 530004, China
- HIV/AIDS Clinical Treatment Center of Guangxi (Nanning) and The Fourth People’s Hospital of Nanning, Nanning 530023, China
| | - Zhe Feng
- Department of Joint and Sports Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530011, China
| | - Fenglian Zhang
- Medical College, Guangxi University, Nanning 530004, China
| | - Bo Wang
- Medical College, Guangxi University, Nanning 530004, China
| | - Zhijuan Wei
- Medical College, Guangxi University, Nanning 530004, China
| | - Nanqing Liao
- Medical College, Guangxi University, Nanning 530004, China
| | - Min Zhang
- Department of Gerontology, Nanning Social Welfare Hospital, Nanning 530004, China
| | - Jian Liang
- Medical College, Guangxi University, Nanning 530004, China
| | - Lisheng Wang
- Medical College, Guangxi University, Nanning 530004, China
| |
Collapse
|
35
|
Zhong X, Wu F, Gao W, Hu J, Shen B, Zhong K, Peng J, Zhang C, Zhang C. Effects of Extracellular Matrix Changes Induced by a High-Fat Diet on Gallbladder Smooth Muscle Dysfunction. FRONT BIOSCI-LANDMRK 2024; 29:401. [PMID: 39735983 DOI: 10.31083/j.fbl2912401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Gallstone formation is a common digestive ailment, with unclear mechanisms underlying its development. Dysfunction of the gallbladder smooth muscle (GSM) may play a crucial role, particularly with a high-fat diet (HFD). This study aimed to investigate the effects of an HFD on GSM and assess how it alters contractility through changes in the extracellular matrix (ECM). METHODS Guinea pigs and C57BL/6 mice were fed either an HFD or normal diet (ND). Primary cultures of their (guinea pigs) gallbladder smooth muscle cells (GSMCs) were used for in vitro experiments. Histological stains, RNA-sequencing, bioinformatics analysis, three-dimensional tissue culture, real-time polymerase chain reaction (PCR), Western blot, atomic force microscopy, and muscle tension measurements were performed. RESULTS Histological evidence indicated structural changes in the gallbladder muscle layer and ECM collagen deposition in the HFD group. The HFD group also showed increased expression of collagen, integrin family, and matrix metalloproteinase (MMP) and the phosphoinositide 3-kinase (PI3K)-protein kinase B (PKB/Akt) signaling pathway. Compared with GSMCs cultured on Matrigel containing 1 mg/mL of collagen I, those cultured with 2 mg/mL showed a phenotype change from contractile to synthetic cells. Consistent with these findings, the HFD group also demonstrated increased ECM stiffness and decreased smooth muscle contractility. CONCLUSIONS Our findings reveal a mechanism by which an HFD alters the ECM composition of the gallbladder muscle, activating the integrin/PI3K-Akt/MMP signaling pathway, thereby impacting GSMC phenotype and contractility. These insights enhance the understanding of gallstone formation mechanism and provide potential therapeutic targets to treat gallbladder dysfunction.
Collapse
Affiliation(s)
- Xingguo Zhong
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, 230041 Hefei, Anhui, China
| | - Feiyang Wu
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, 230041 Hefei, Anhui, China
- Graduate School, Bengbu Medical University, 233030 Bengbu, Anhui, China
| | - Weicheng Gao
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, 230041 Hefei, Anhui, China
- Graduate School, Bengbu Medical University, 233030 Bengbu, Anhui, China
| | - Jinlong Hu
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, 230041 Hefei, Anhui, China
| | - Bing Shen
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, 999078 Taipa, Macao SAR, China
| | - Kaiyuan Zhong
- College of Medical Technology, Qiqihar Medical University, 161003 Qiqihar, Heilongjiang, China
| | - Junbin Peng
- Medical School, Anhui University of Science & Technology, 232001 Huainan, Anhui, China
| | - Chong Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| | - Chao Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| |
Collapse
|
36
|
Chen P, Jiang X, Fu J, Ou C, Li Y, Jia J, Liao C. The potential mechanism of action of gut flora and bile acids through the TGR5/TRPV1 signaling pathway in diabetic peripheral neuropathic pain. Front Endocrinol (Lausanne) 2024; 15:1419160. [PMID: 39619328 PMCID: PMC11604420 DOI: 10.3389/fendo.2024.1419160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/22/2024] [Indexed: 12/13/2024] Open
Abstract
Diabetic peripheral neuropathic pain (DPNP) is a major complication of diabetes that markedly affects the quality of life and health status of patients. Recent studies have investigated the potential regulatory influence of gut flora and bile acids on DPNP via the TGR5/TRPV1 signaling pathway. Dysbiosis of the gut flora not only directly affects bile acid metabolism but also significantly correlates with diabetes-associated neuropathy through interactions with the bile acid receptor TGR5 and the ion channel TRPV1. This review describes how alterations in the gut flora and bile acid metabolism contribute to the pathogenesis of DPNP through the TGR5/TRPV1 signaling pathway, revealing potential applications for this pathway in DPNP management. Furthermore, experimental and clinical studies have demonstrated the modulation of gut flora and bile acid metabolism as well as targeting the TGR5/TRPV1 signaling pathway as an innovative therapeutic approach. Further studies are warranted to elucidate the underlying mechanism and develop treatment modalities based on gut flora regulation and signaling pathway interventions, thus providing novel insights and approaches for DPNP therapy.
Collapse
Affiliation(s)
- Peng Chen
- Department of Pediatrics, Southwest Medical University, Luzhou, Sichuan, China
| | - Xian Jiang
- Department of Anesthesiology, Luzhou People’s Hospital, Luzhou, Sichuan, China
| | - Jia Fu
- Department of Pain Management, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Cehua Ou
- Department of Pain Management, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yao Li
- Department of Science and Technology, Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Changli Liao
- Department of Science and Technology, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
37
|
Aydin BN, Stinson EJ, Hanson RL, Looker HC, Cabeza De Baca T, Krakoff J, Chang DC. Hepatic Insulin Resistance Increases Risk of Gallstone Disease in Indigenous Americans in the Southwestern United States. Clin Transl Gastroenterol 2024; 15:e00763. [PMID: 39166750 PMCID: PMC11596648 DOI: 10.14309/ctg.0000000000000763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
INTRODUCTION Animal models indicate that hepatic insulin resistance (IR) promotes cholesterol gallstone disease (GSD). We sought to determine whether hepatic and whole-body IR is associated with incident GSD. METHODS At baseline, 450 Southwestern Indigenous American adults without GSD were included. Participants had a 2-step hyperinsulinemic-euglycemic clamp with glucose tracer at submaximal and maximal insulin stimulation (240 and 2,400 pmol/m 2 /min) for whole-body IR (M-low and M-high) and hepatic glucose production (HGP) before and during submaximal insulin infusion (HGP-basal and HGP-insulin). Incident GSD was identified during follow-up visits conducted at ∼2-year intervals. The associations of HGP (basal, insulin, and % suppression), M-low, and M-high with risk of GSD were assessed by Cox regression models adjusted for age, sex, body fat (%), glucose, and insulin. RESULTS Sixty participants (13%) developed GSD (median follow-up: 11.6 years). Participants who developed GSD were of similar age and whole-body IR as those who did not ( P 's > 0.07) but were more likely to be female; have higher body fat, higher HGP-basal, and HGP-insulin; and lower % suppression of HGP ( P 's < 0.02). In separate adjusted models, higher HGP-insulin and lower % suppression of HGP were associated with increased risk for GSD (hazard ratio [HR] per SD: HR 1.38, 95% CI 1.12-1.69, P = 0.002; HR 1.41, 95% CI 1.16-1.72, P = 0.0007). HGP-basal, M-low, and M-high were not associated with GSD in adjusted models ( P 's > 0.22). DISCUSSION Resistance to insulin suppression of HGP increases risk for GSD. Hepatic IR is a link between GSD and other conditions of the metabolic syndrome.
Collapse
Affiliation(s)
- Beyza N. Aydin
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Emma J. Stinson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Robert L. Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Helen C. Looker
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Tomás Cabeza De Baca
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Douglas C. Chang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| |
Collapse
|
38
|
Zhao Y, Qin C, Lin C, Li Z, Zhao B, Li T, Zhang X, Wang W. Pancreatic ductal adenocarcinoma cells reshape the immune microenvironment: Molecular mechanisms and therapeutic targets. Biochim Biophys Acta Rev Cancer 2024; 1879:189183. [PMID: 39303859 DOI: 10.1016/j.bbcan.2024.189183] [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: 06/09/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a digestive system malignancy characterized by challenging early detection, limited treatment alternatives, and generally poor prognosis. Although there have been significant advancements in immunotherapy for hematological malignancies and various solid tumors in recent decades, with impressive outcomes in recent preclinical and clinical trials, the effectiveness of these therapies in treating PDAC continues to be modest. The unique immunological microenvironment of PDAC, especially the abnormal distribution, complex composition, and variable activation states of tumor-infiltrating immune cells, greatly restricts the effectiveness of immunotherapy. Undoubtedly, integrating data from both preclinical models and human studies helps accelerate the identification of reliable molecules and pathways responsive to targeted biological therapies and immunotherapies, thereby continuously optimizing therapeutic combinations. In this review, we delve deeply into how PDAC cells regulate the immune microenvironment through complex signaling networks, affecting the quantity and functional status of immune cells to promote immune escape and tumor progression. Furthermore, we explore the multi-modal immunotherapeutic strategies currently under development, emphasizing the transformation of the immunosuppressive environment into an anti-tumor milieu by targeting specific molecular and cellular pathways, providing insights for the development of novel treatment strategies.
Collapse
Affiliation(s)
- Yutong Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Cheng Qin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Chen Lin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Zeru Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Bangbo Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Tianyu Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Xiangyu Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Weibin Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| |
Collapse
|
39
|
Provitera L, Tomaselli A, Algieri F, Tripodi M, Raffaeli G, Amodeo I, Raymo L, Bronzoni CV, Fumagalli M, Garrido F, Cavallaro G. Gut Microbiota-Derived Metabolites and Their Role in the Pathogenesis of Necrotizing Enterocolitis in Preterm Infants: A Narrative Review. Metabolites 2024; 14:570. [PMID: 39590806 PMCID: PMC11596930 DOI: 10.3390/metabo14110570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease that occurs predominantly in premature infants and is characterized by the inflammation and necrosis of the intestine, showing high morbidity and mortality rates. Despite decades of research efforts, a specific treatment is currently lacking, and preventive strategies are the mainstays of care. This review aims to help understand the complex interplay between gut microbiota and their metabolites in NEC pathogenesis. In particular, we focused on how these factors can influence gut health, immune responses, and intestinal barrier integrity. Discussion: Current research has increasingly focused on the role of the gut microbiota and their metabolites in NEC pathogenesis, thanks to their involvement in modulating gut health, immune responses, and intestinal barrier integrity. Conclusions: A deeper understanding of the interplay between gut microbiota and their metabolites is essential for developing personalized strategies to prevent NEC. By targeting these microbial interactions, new therapeutic approaches may emerge that offer improved outcomes for preterm infants at a high risk of NEC.
Collapse
Affiliation(s)
- Livia Provitera
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Andrea Tomaselli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Francesca Algieri
- Research and Development Unit, Postbiotica S.R.L., 20123 Milan, Italy;
| | - Matteo Tripodi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Genny Raffaeli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Ilaria Amodeo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Ludovica Raymo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Carolina Vittoria Bronzoni
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Monica Fumagalli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Felipe Garrido
- Department of Pediatrics, Clínica Universidad de Navarra, 28027 Madrid, Spain;
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| |
Collapse
|
40
|
Wang B, Xiong Y, Li R, Zhang S. Depression increases the risk of gallstone: A cross-sectional study and Mendelian randomization analysis. J Affect Disord 2024; 362:606-614. [PMID: 39029662 DOI: 10.1016/j.jad.2024.07.119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/28/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Gallstone, a common digestive disorder, poses a significant public health burden. Concurrently, depression is acknowledged as a health risk. However, limited information exists on depression's impact on gallstone formation. This study investigates depression's causal effect on gallstone risk. METHODS Using National Health and Nutrition Examination Survey (NHANES) data, we conducted an observational study. The severity of depression was assessed using the Patient Health Questionnaire-9 (PHQ-9). Multivariable logistic regression and subgroup analyses explored the correlation between depression and gallstone risk. Mendelian Randomization (MR) analysis, leveraging Genome-Wide Association Studies (GWAS) data, reduced observational bias and elucidated causality. Inverse Variance Weighting (IVW) was the primary method, with sensitivity analyses validating results. RESULTS In the observational study (7707 participants), gallstone risk was elevated in mild (OR: 1.58, 95 % CI 1.31-1.90, P < 0.001), moderate (OR: 2.07, 95 % CI 1.59-2.67, P < 0.001), and severe (OR: 2.41, 95 % CI 1.70-3.34, P < 0.001) depression groups (P for trend <0.001). Subgroup analyses revealed a stronger association in those under 65, females, non-Hispanic Black, individuals with obesity, smokers, and those with college education or higher. Mendelian Randomization indicated a causal link between genetically predicted depression and higher cholelithiasis risk (OR: 2.06, 95 % CI 1.34-3.17, P = 0.001), validated through sensitivity analyses and multi-cohort verification. CONCLUSION Depression independently increases gallstone risk, particularly in those under 65, females, non-Hispanic Black, individuals with obesity, smokers, and those with college education or higher. Further validation is needed through multi-center, prospective cohort studies.
Collapse
Affiliation(s)
- Bo Wang
- Department of Geriatric Digestive Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yongqiang Xiong
- Department of Geriatric Digestive Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ren Li
- Department of Geriatric Digestive Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shu Zhang
- Department of Geriatric Digestive Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Experimental Teaching Center for Clinical Skills, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
41
|
Yang Z, Song C, Lu C. Investigation of the causal relationship between cholelithiasis and Parkinson's disease: A bidirectional Mendelian randomization study. Health Sci Rep 2024; 7:e70126. [PMID: 39377023 PMCID: PMC11457209 DOI: 10.1002/hsr2.70126] [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: 04/13/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
Background Parkinson's disease (PD) and cholelithiasis are a huge public health burden. Although observational studies have suggested a potential link between PD and cholelithiasis, the causal relationship between the two remains uncertain. To address this gap, we performed a two-sample bidirectional Mendelian randomization analysis using genetic tools. Method Genome-wide association study summary statistics for all traits were obtained from publicly available databases. We used strict control steps in instrumental variable selection to screen for single nucleotide polymorphisms (SNPs) from summary-level genome-wide association studies. In addition, all F-statistics were >10, indicating no weak instrumental bias. The inverse variance weighting (IVW) method was the primary method used to assess causal associations. Four other MR methods (MR-Egger, Weighted Median, Simple mode, and Weighted mode) were also used to complement IVW. Various sensitivity tests were also performed to assess reliability: (1) Cochrane's Q test for assessing heterogeneity, (2) MR-Egger intercept test and MR-PRESSO global test for assessing horizontal multiplicity, and (3) leave-one-out sensitivity test for determining stability. Results We selected a total of 30 SNPs as instrumental variables. It was demonstrated that cholelithiasis had a causal effect on the risk of PD (OR = 1.146, 95% CI: 1.062-1.236, p < 0.001) in IVW method. Conclusion The results of our analysis revealed an increased risk effect of cholelithiasis against PD, which may give light on new approaches to PD prevention and therapy.
Collapse
Affiliation(s)
- Zijiao Yang
- Department of Geriatrics, Yangpu Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Chengfu Song
- Department of Geriatrics, Yangpu Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Chong Lu
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| |
Collapse
|
42
|
Shen S, Han G, Dong Z, Wu S, Ma S, Ding Z, Zhao Y, Wan X. Accumulation of rare earth elements in human gallstones: a perspective from dietary and human health. BMC Gastroenterol 2024; 24:324. [PMID: 39333954 PMCID: PMC11437671 DOI: 10.1186/s12876-024-03426-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Gallstone disease poses a global threat to human health and is strongly linked to environmental factors. However, there is currently no data on the presence of rare earth elements (REEs) in human gallstones. This paper investigates the concentration and distribution of REEs in gallstones for the first time, aiming to explore the environmental implications on human health. METHODS A total of 25 gallstone samples were collected in Shanghai and the content of REEs was measured by Inductively coupled plasma-Mass Spectrometry (ICP-MS) to explore the distribution of REEs in gallstones. RESULTS The concentration of REEs in gallstones ranged from 4.89 to 190.8 ng/g (mean 39.21). In most of the gallstone analyses, REEs have been detected and generally attributed to environmental exposure or food contamination. The Y/Ho ratio of gallstones was lower than that of continental rocks, similar to that in the blood, indicating limited fractionation during fluid transport processes in the gallbladder. CONCLUSIONS The upper continental crust (UCC)-normalized REEs pattern in gallstones showed depletion of light REEs, while most showed enrichment of heavy REEs. Positive Gd anomalies were found in most samples, while few samples suggested anthropogenic influence. Whether exogenous inputs or in vivo biofractionation lead to changes in REEs fractionated patterns require further analyses.
Collapse
Affiliation(s)
- Shuang Shen
- Digestive Endoscopic Center, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Central Lab, School of Medicine, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Guilin Han
- State Key Laboratory of Biogeology and Environmental Geology, China University of Geosciences (Beijing), Beijing, 100083, China.
- Frontiers Science Center for Deep-time Digital Earth, Institute of Earth Scineces, University of Geosciences (Beijing), 100083, Beijing, China.
| | - Zhixia Dong
- Digestive Endoscopic Center, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Shan Wu
- Digestive Endoscopic Center, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Shunrong Ma
- State Key Laboratory of Biogeology and Environmental Geology, China University of Geosciences (Beijing), Beijing, 100083, China
- Frontiers Science Center for Deep-time Digital Earth, Institute of Earth Scineces, University of Geosciences (Beijing), 100083, Beijing, China
| | - Ziyang Ding
- State Key Laboratory of Biogeology and Environmental Geology, China University of Geosciences (Beijing), Beijing, 100083, China
- Frontiers Science Center for Deep-time Digital Earth, Institute of Earth Scineces, University of Geosciences (Beijing), 100083, Beijing, China
| | - Ye Zhao
- Nu Instruments, Wrexham Industrial Estate, 74 Clywedog Road South, 13 9XS, Wresham, LL, UK
| | - Xinjian Wan
- Digestive Endoscopic Center, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
43
|
Datta S, Pasham S, Inavolu S, Boini KM, Koka S. Role of Gut Microbial Metabolites in Cardiovascular Diseases-Current Insights and the Road Ahead. Int J Mol Sci 2024; 25:10208. [PMID: 39337693 PMCID: PMC11432476 DOI: 10.3390/ijms251810208] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of premature morbidity and mortality globally. The identification of novel risk factors contributing to CVD onset and progression has enabled an improved understanding of CVD pathophysiology. In addition to the conventional risk factors like high blood pressure, diabetes, obesity and smoking, the role of gut microbiome and intestinal microbe-derived metabolites in maintaining cardiovascular health has gained recent attention in the field of CVD pathophysiology. The human gastrointestinal tract caters to a highly diverse spectrum of microbes recognized as the gut microbiota, which are central to several physiologically significant cascades such as metabolism, nutrient absorption, and energy balance. The manipulation of the gut microbial subtleties potentially contributes to CVD, inflammation, neurodegeneration, obesity, and diabetic onset. The existing paradigm of studies suggests that the disruption of the gut microbial dynamics contributes towards CVD incidence. However, the exact mechanistic understanding of such a correlation from a signaling perspective remains elusive. This review has focused upon an in-depth characterization of gut microbial metabolites and their role in varied pathophysiological conditions, and highlights the potential molecular and signaling mechanisms governing the gut microbial metabolites in CVDs. In addition, it summarizes the existing courses of therapy in modulating the gut microbiome and its metabolites, limitations and scientific gaps in our current understanding, as well as future directions of studies involving the modulation of the gut microbiome and its metabolites, which can be undertaken to develop CVD-associated treatment options. Clarity in the understanding of the molecular interaction(s) and associations governing the gut microbiome and CVD shall potentially enable the development of novel druggable targets to ameliorate CVD in the years to come.
Collapse
Affiliation(s)
- Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Sindhura Pasham
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Sriram Inavolu
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Krishna M Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| |
Collapse
|
44
|
Zhou C, Ye X, Liu Z, Liu T, Li S, Yang J, Wei J, Yu P, Jia R, Zhao W. Dissecting the causal links between gut microbiome, immune traits and polyp using genetic evidence. Front Immunol 2024; 15:1431990. [PMID: 39346904 PMCID: PMC11427361 DOI: 10.3389/fimmu.2024.1431990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/19/2024] [Indexed: 10/01/2024] Open
Abstract
Background Previous research has demonstrated an association between gut microbiota and immune status with the development of several diseases. However, whether these factors contribute to polyps remains unclear. This study aims to use Mendelian randomization (MR) to investigate the causal relationship between gut microbiota and 4 types of polyps (nasal, gallbladder, colon, and gastric polyps), as well as to analyze the mediating role of immune traits. Methods This study utilized large-scale GWAS meta-analyses of gut microbiota (MiBioGen Consortium), 731 immune traits, and 4 types of polyps (one from the FinnGen Consortium and three from the NBDC Human Database). Univariate MR with the inverse variance weighted (IVW) estimation method was employed as the primary analytical approach. A two-step MR analysis was performed to identify potential mediating immune traits. Additionally, multivariable MR approach based on Bayesian model averaging (MR-BMA) was employed to further prioritize gut microbiota and immune traits associated with polyp development. Results Based on IVW method in univariate MR analysis, we identified 39 gut microbial taxa and 135 immune traits significantly causally associated with at least one type of polyp. For nasal polyps, 13 microbial taxa and 61 immune traits were causally associated. After false discovery rate (FDR) correction, CD3 on Central Memory CD8+ T cells and CD3 on CD4 regulatory T cells remained significant. MR-BMA identified 4 gut microbial taxa and 4 immune traits as high priority. For gallbladder polyps, 9 microbial taxa and 30 immune traits were causally associated. MR-BMA identified 8 microbial taxa and 6 immune traits as higher importance. For colon polyps, 6 microbial taxa and 21 immune traits were causally associated. MR-BMA identified 4 microbial taxa and 3 immune traits as higher importance. For gastric polyps, 12 microbial taxa and 33 immune traits were causally associated. Actinobacteria remained significant after FDR correction, and MR-BMA identified 7 gut microbial taxa and 6 immune traits as high priority. We identified 16 causal pathways with mediator directions consistent with the direction of gut microbiome-polyp association. Of these, 6 pathways were associated with the mechanism of nasal polyps, 1 with gallbladder polyps, 2 with colon polyps, and 7 with gastric polyps. Conclusions Our findings shed light on the causal relationships between gut microbiota, immune traits, and polyp development, underscoring the crucial roles of gut microbiota and immune status in polypogenesis. Furthermore, these findings suggest potential applications in polyp prevention, early screening, and the development of effective strategies to reduce polyp risk.
Collapse
Affiliation(s)
- Cheng Zhou
- Department of Gastroenterology, Changzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Changzhou, China
| | - Xiaofeng Ye
- Department of Gastroenterology, Changzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Changzhou, China
| | - Zhinuo Liu
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tong Liu
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Shanzheng Li
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinqiu Yang
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jingjing Wei
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Peng Yu
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ran Jia
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Wenxia Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
45
|
Uche-Anya E, Ha J, Balasubramanian R, Rexrode KM, Chan AT. Metabolomic profiles of incident gallstone disease. BMJ Open Gastroenterol 2024; 11:e001417. [PMID: 39209332 PMCID: PMC11367368 DOI: 10.1136/bmjgast-2024-001417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS Gallstone disease affects ≥40 million people in the USA and accounts for health costs of ≥$4 billion a year. Risk factors such as obesity and metabolic syndrome are well established. However, data are limited on relevant metabolomic alterations that could offer mechanistic and predictive insights into gallstone disease. This study prospectively identifies and externally validates circulating prediagnostic metabolites associated with incident gallstone disease. METHODS Female participants in Nurses' Health Study (NHS) and Nurses' Health Study II (NHS II) who were free of known gallstones (N=9960) were prospectively followed up after baseline metabolomic profiling with liquid chromatography-tandem mass spectrometry. Multivariable logistic regression and enrichment analysis were used to identify metabolites and metabolite groups associated with incident gallstone disease at PFDR<0.05. Findings were validated in 1866 female participants in the Women's Health Initiative and a comparative analysis was performed with 2178 male participants in the Health Professionals Follow-up Study. RESULTS After multivariate adjustment for lifestyle and putative risk factors, we identified and externally validated 17 metabolites associated with incident gallstone disease in women-nine triacylglycerols (TAGs) and diacylglycerols (DAGs) were positively associated, while eight plasmalogens and cholesterol ester (CE) were negatively associated. Enrichment analysis in male and female cohorts revealed positive class associations with DAGs, TAGs (≤56 carbon atoms and ≤3 double bonds) and de novo TAG biosynthesis pathways, as well as inverse associations with CEs. CONCLUSIONS This study highlights several metabolites (TAGs, DAGs, plasmalogens and CE) that could be implicated in the aetiopathogenesis of gallstone disease and serve as clinically relevant markers.
Collapse
Affiliation(s)
- Eugenia Uche-Anya
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jane Ha
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Raji Balasubramanian
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Kathryn M Rexrode
- Division of Women's Health, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Li X, Shang S, Wu M, Song Q, Chen D. Gut microbial metabolites in lung cancer development and immunotherapy: Novel insights into gut-lung axis. Cancer Lett 2024; 598:217096. [PMID: 38969161 DOI: 10.1016/j.canlet.2024.217096] [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: 04/03/2024] [Revised: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Metabolic derivatives of numerous microorganisms inhabiting the human gut can participate in regulating physiological activities and immune status of the lungs through the gut-lung axis. The current well-established microbial metabolites include short-chain fatty acids (SCFAs), tryptophan and its derivatives, polyamines (PAs), secondary bile acids (SBAs), etc. As the study continues to deepen, the critical function of microbial metabolites in the occurrence and treatment of lung cancer has gradually been revealed. Microbial derivates can enter the circulation system to modulate the immune microenvironment of lung cancer. Mechanistically, oncometabolites damage host DNA and promote the occurrence of lung cancer, while tumor-suppresive metabolites directly affect the immune system to combat the malignant properties of cancer cells and even show considerable application potential in improving the efficacy of lung cancer immunotherapy. Considering the crosstalk along the gut-lung axis, in-depth exploration of microbial metabolites in patients' feces or serum will provide novel guidance for lung cancer diagnosis and treatment selection strategies. In addition, targeted therapeutics on microbial metabolites are expected to overcome the bottleneck of lung cancer immunotherapy and alleviate adverse reactions, including fecal microbiota transplantation, microecological preparations, metabolite synthesis and drugs targeting metabolic pathways. In summary, this review provides novel insights and explanations on the intricate interplay between gut microbial metabolites and lung cancer development, and immunotherapy through the lens of the gut-lung axis, which further confirms the possible translational potential of the microbiome metabolome in lung cancer treatment.
Collapse
Affiliation(s)
- Xinpei Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shijie Shang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Qian Song
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| | - Dawei Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
47
|
Pan T, Zhang C, Liang J, Wang X, Di X, Zhou Y, Bai P, Yuan H. Association between life-ever gallstones and depressive symptoms in U.S. adults: a cross-sectional study. Sci Rep 2024; 14:18845. [PMID: 39143232 PMCID: PMC11325026 DOI: 10.1038/s41598-024-69777-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 08/08/2024] [Indexed: 08/16/2024] Open
Abstract
Research on the potential association between life-ever gallstones and depressive symptoms is limited. This study aims to evaluate whether the presence of gallstone disease is associated with depressive symptoms. In this cross-sectional study, we analyzed data from the National Health and Nutrition Examination Survey (NHANES) 2017-March 2020 cycles. The presence of depressive symptoms and gallstone disease was assessed using questionnaire responses. Adjusted odds ratios (OR) were calculated using a multivariate logistic regression model, with adjustments made for age, sex, race, body mass index, history of cardiovascular disease, hypertension, arthritis, and pulmonary disease across different models. Subgroup and sensitivity analyses were conducted to ensure the stability of the results. This study included 6201 adults aged 20 years and above, with 539(8.7%) experiencing depressive symptoms. After adjusting for age, sex, race, body mass index, CVD history, hypertension, arthritis, pulmonary disease, depressive symptoms were possibly associated with life-ever gallstones (OR 1.37, 95% CI 0.91-2.08).When depressive symptoms were categorized as mild, moderate, moderately severe, and severe,life-ever gallstones was possibly associated with mild depressive symptoms (OR 1.12, 95% CI 0.81-1.56), moderate depressive symptoms (OR 1.37, 95% CI 0.89-2.12), moderately severe depressive symptoms (OR 1.93, 95% CI 0.93-3.99), and severe depressive symptoms (OR 0.67, 95% CI 0.16-2.88).As a continuous variable, life-ever gallstones was associated with the PHQ-9 score (OR 0.42, 95% CI 0.02-0.83). The results remained stable after multiple imputation for all missing data. This cross-sectional study demonstrates no significant association between life-ever gallstones and depressive symptoms in US adults.
Collapse
Affiliation(s)
- Ting Pan
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, No. 51, Xiaoguan Street, Outside Andingmen, Chaoyang District, Beijing, 100029, China
| | - Chongyang Zhang
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, No. 51, Xiaoguan Street, Outside Andingmen, Chaoyang District, Beijing, 100029, China
| | - Junjie Liang
- Capital Medical University Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| | - Xinru Wang
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, No. 51, Xiaoguan Street, Outside Andingmen, Chaoyang District, Beijing, 100029, China
| | - Xueshi Di
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, No. 51, Xiaoguan Street, Outside Andingmen, Chaoyang District, Beijing, 100029, China
| | - Yuqi Zhou
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, No. 51, Xiaoguan Street, Outside Andingmen, Chaoyang District, Beijing, 100029, China
| | - Peng Bai
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, No. 51, Xiaoguan Street, Outside Andingmen, Chaoyang District, Beijing, 100029, China.
| | - Hongwei Yuan
- Dongzhimen Hospital of Beijing University of Chinese Medicine, No. 5 Haiyun Cang, Dongcheng District, Beijing, China.
| |
Collapse
|
48
|
Liu M, Jiang X, Zeng X, Guo Y, Zhang T, Fan X, Xu J, Wu Z, Pan D. A protective mechanism of heat inactivation to enhance Levilactobacillus brevis PDD-2 against alcohol-induced chronic liver disease based on proteomic analysis. Food Funct 2024; 15:8356-8369. [PMID: 39023014 DOI: 10.1039/d4fo01051e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
A proteomics-based analysis of the effect of heat inactivation on the alleviation of alcoholic liver disease (ALD) using Levilactobacillus brevis PDD-2 is presented, aimed at exploring the potential and mechanisms of postbiotic elements prepared through heat inactivation in the treatment of ALD. It was found that L. brevis PDD-2 and its postbiotic (heat-inactivated L. brevis PDD-2) alleviate chronic ALD via the gut-liver axis. In particular, heat-inactivated L. brevis PDD-2 significantly increased the relative abundance of Erysipelotrichaceae and better facilitated the oxidative stress balance in the liver. The tandem mass tag (TMT)-based quantitative proteomics technique analyses revealed that heat-inactivated L. brevis PDD-2 was associated with up-regulated expression levels of proteins related to the redox system, cellular metabolism, amino acid and oligopeptide transport, and surface proteins with immunomodulatory capacity. These findings provide a theoretical basis for developing novel therapeutic strategies and lay a solid foundation for further revealing its exhaustive mechanisms.
Collapse
Affiliation(s)
- Mingzhen Liu
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo 315211, Zhejiang, China.
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Xiaoxiao Jiang
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, China
- Institute of Agricultural Products, Shandong Academy of Agricultural Sciences, Jinan 250100, Shandong, China
| | - Xiaoqun Zeng
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo 315211, Zhejiang, China.
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Yuxing Guo
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, China
| | - Tao Zhang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo 315211, Zhejiang, China.
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Xiankang Fan
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo 315211, Zhejiang, China.
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Jue Xu
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo 315211, Zhejiang, China.
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Zhen Wu
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo 315211, Zhejiang, China.
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Daodong Pan
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo 315211, Zhejiang, China.
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| |
Collapse
|
49
|
Yan S, He J, Yu X, Shang J, Zhang Y, Bai H, Zhu X, Xie X, Lee L. Causal relationship between gut microbiota and thyroid nodules: a bidirectional two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1417009. [PMID: 39175567 PMCID: PMC11338761 DOI: 10.3389/fendo.2024.1417009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024] Open
Abstract
Objective Emerging evidence suggests alterations in gut microbiota (GM) composition following thyroid nodules (TNs) development, yet the causal relationship remains unclear. Utilizing Mendelian Randomization (MR), this study aims to elucidate the causal dynamics between GM and TNs. Methods Employing summary statistics from the MiBioGen consortium (n=18,340) and FinnGen consortium (1,634 TNs cases, 263,704 controls), we conducted univariable and multivariable MR analyses to explore the GM-TNs association. Techniques including inverse variance weighted, MR-Egger regression, weighted median, and MR-PRESSO were utilized for causal inference. Instrumental variable heterogeneity was assessed through Cochran's Q statistic and leave-one-out analysis. Reverse MR was applied for taxa showing significant forward MR associations, with multivariate adjustments for confounders. Results Our findings suggest that certain microbiota, identified as Ruminococcaceae_NK4A214_group (OR, 1.89; 95%CI, 0.47-7.64; p = 0.040), Senegalimassilia (OR, 1.72; 95%CI, 1.03-2.87; p =0.037), Lachnospiraceae (OR,0.64; 95%CI,0.41-0.99; p =0.045), exhibit a protective influence against TNs' development, indicated by negative causal associations. In contrast, microbiota categorized as Desulfovibrionales (OR, 0.63; 95%CI, 0.41-0.95; p =0.028), Prevotella_7 (OR, 0.79; 95%CI, 0.63-1.00; p =0.049), Faecalibacterium (OR, 0.66; 95%CI, 0.44-1.00; p =0.050), Desulfovibrionaceae (OR, 0.55; 95%CI, 0.35-0.86; p =0.008), Deltaproteobacteria (OR, 0.65; 95%CI, 0.43-0.97; p =0.036) are have a positive correlation with with TNs, suggesting they may serve as risk factors. Reverse MR analyses did not establish significant causal links. After comprehensive adjustment for confounders, taxa Desulfovibrionales (Order), Desulfovibrionaceae (Family), Deltaproteobacteria (Class) remain implicated as potential contributors to TNs' risk. Discussion This study substantiates a significant causal link between GM composition and TNs development, underscoring the thyroid-gut axis's relevance. The findings advocate for the integration of GM profiles in TNs' prevention and management, offering a foundation for future research in this domain.
Collapse
Affiliation(s)
- Shaoshuai Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jiawei He
- School of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Xudong Yu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jianwei Shang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yaosheng Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Han Bai
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xingyu Zhu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoming Xie
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Leanne Lee
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
50
|
Zaparte A, Christopher CJ, Arnold C, Richey L, Castille A, Mistretta K, Taylor CM, Lin H, Nelson S, Kirwan JP, Apolzan JW, Campagna SR, Welsh DA. Effects of E-Cigarettes on the Lung and Systemic Metabolome in People with HIV. Metabolites 2024; 14:434. [PMID: 39195530 DOI: 10.3390/metabo14080434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
The popularity of e-cigarettes (vaping) has soared, creating a public health crisis among teens and young adults. Chronic vaping can induce gut inflammation and reduce intestinal barrier function through the production of the proinflammatory molecule hydrogen sulfide (H2S). This is particularly concerning for people with HIV (PWH) as they already face impaired immune function and are at a higher risk for metabolic dysregulation, diabetes, and chronic liver disease. Furthermore, PWH experience unhealthy behaviors, making it crucial to understand the systemic metabolic dysregulation and pathophysiological mechanisms associated with vaping in this population. Here, we employed liquid chromatography-mass spectrometry (LC-MS)-based metabolomics to investigate the upper respiratory, circulation, and gut metabolic profiles of PWH who vape (n = 7) and smoke combustible tobacco/marijuana (n = 6) compared to control participants who did not vape or smoke (n = 10). This hypothesis-generating exploratory study revealed systemic alterations in purine, neurotransmitter, and vitamin B metabolisms and tissue-specific changes in inflammatory pathways and cryptic sulfur cycling associated with vaping and combustible tobacco/marijuana smoking in PWH. In addition, this study provides the first link between microbial-derived metabolite 2,3-dihydroxypropane-1-sulfonate (DHPS) and vaping/smoking (tobacco and marijuana)-induced metabolic dyshomeostasis in the gut. These findings highlight the importance of identifying the full biological and clinical significance of the physiological changes and risks associated with vaping.
Collapse
Affiliation(s)
- Aline Zaparte
- Department of Internal Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Courtney J Christopher
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN 37996, USA
| | - Connie Arnold
- Department of Medicine, Louisiana State University Health Sciences, Shreveport, LA 71103, USA
| | - Lauren Richey
- Department of Internal Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Adairre Castille
- Department of Internal Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Kyle Mistretta
- Department of Internal Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Christopher M Taylor
- Department of Microbiology, Immunology, & Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Huiyi Lin
- School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Steve Nelson
- Department of Internal Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - John P Kirwan
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - John W Apolzan
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Shawn R Campagna
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN 37996, USA
- Biological and Small Molecule Mass Spectrometry Core, University of Tennessee, 1420 Circle Drive, Knoxville, TN 37996, USA
| | - David A Welsh
- Department of Internal Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|