1
|
Hou C, Chen Y, Zhang W, Yu J, Ji M, Cai S, Guo W, Ji X, Sun L, Liu X, Wang Y. An insight into the full aspects of bound polyphenols in dietary fiber: Interaction, composition, function and foundation as well as alteration in food processing. Food Chem 2025; 485:144553. [PMID: 40318329 DOI: 10.1016/j.foodchem.2025.144553] [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: 01/15/2025] [Revised: 04/08/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Dietary fiber (DF) and polyphenols are both bioactive compounds with various health-promoting effects while close relationship between them aroused wide concern in recent years. Abundant polyphenols combine with DF and contribute greatly to its beneficial effects. Although efforts made to uncover such bound polyphenols (BPs) from different angles before, systematic overview of full aspects is deficient. Here, more details about polyphenols conjugated in DF reported recently were summarized systematically. Meanwhile, the disposition of BPs in gastrointestinal tract and their interaction with microbiome were introduced to clarify the foundation of their functions. Moreover, considering the great impacts of food processing on polyphenols, different technics used in food handling were introduced with their effects on BPs emphatically discussed to provide guideline for reasonable application of specific technics for given materials. Our work is supposed to promote the understanding of BPs in DF and facilitate their future exploitation and application as a whole.
Collapse
Affiliation(s)
- Chunyan Hou
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Youkang Chen
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Wanting Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Jingjing Yu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Muhua Ji
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Shuo Cai
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Wenhao Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Xiaolong Ji
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou, 450001, China
| | - Lijun Sun
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Yutang Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
2
|
Xu Y, Zhang X, Zhang Y, Ma H, Zhou Z, Qin H, Liu H, Han X. Integrated multi-omics insight into the molecular networks of oxidative stress in triggering multiple sclerosis. Neurobiol Dis 2025; 210:106929. [PMID: 40280189 DOI: 10.1016/j.nbd.2025.106929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025] Open
Abstract
Oxidative stress (OS) is a key pathophysiological mechanism in multiple sclerosis (MS). However, the underlying mechanisms by which OS triggered MS remain unknown. To identify potential causal targets of 1216 OS-related genes for MS, a summary-data-based Mendelian randomization (SMR) method was applied. Given that genes can exert their biological functions through different omics levels, the multi-omics SMR integrating expression, methylation, and protein quantitative trait loci (eQTL, mQTL, and pQTL) of OS-related genes from blood and brain tissues was utilized. Bayesian colocalization test was conducted to examine potential regulatory mechanisms of QTL risk variation in MS. To verify the robustness of our results, we validated these findings in FinnGen cohort. Furthermore, the QTL evidence levels, colocalization findings, and replication cohort results were integrated and potential target genes were categorized into three levels. Consequently, three genes (BACH2, TRAF3, and MAPK3) were identified as potential contributors to MS in blood, and four genes (HMGCL, TSFM, TRAF3 and HLA-B) were identified as potential contributors to MS in brain tissue. Additionally, HMGCL and TSFM from brain tissue were supported by first-level evidence related to MS and were validated via in vitro experiments. This research not only contributed to fundamental research of OS in MS but also supported the identification of potential targets for clinical interventions in MS.
Collapse
Affiliation(s)
- Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaowei Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongxuan Ma
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhaokai Zhou
- Department of Urology Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongzhuo Qin
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huimin Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China.
| |
Collapse
|
3
|
Ni T, Shen Z, Shi H, Han Z. Uncovering the gut microbiota's role in temporomandibular joint disorders: A bidirectional Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42590. [PMID: 40489886 DOI: 10.1097/md.0000000000042590] [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] [Indexed: 06/11/2025] Open
Abstract
Previous studies have revealed the association between gut microbiota (GM) and temporomandibular joint inflammation, which leads to temporomandibular joint disorders (TMD). However, the causality of the associations remains unknown. This Mendelian randomization (MR) study aims to clarify the causal relationships between GM and TMD. We employed a two-sample MR approach to analyze GM data from the MiBioGen consortium, including 18,340 participants and 211 taxa, and TMD data from the FinnGen consortium R10 release, including 6314 cases and 222,498 controls. Various MR methods, including inverse-variance weighted (IVW), MR-Egger, and weighted median analyses, alongside comprehensive sensitivity analyses, were used to assess causality. After comprehensive sensitivity analyses, the study found causal links between 6 certain GM at the genus level and TMD, including Eubacterium fissicatena group (IVW's odds ratio [OR] = 1.18; 95% confidence interval [CI]: 1.02-1.37; P = .027), Catenibacterium (IVW's OR = 1.33; 95% CI: 1.11-1.59; P = .002), Oxalobacter (IVW's OR = 0.85; 95% CI: 0.75-0.97; P = .013), Ruminococcaceae NK4A214 (IVW's OR = 0.80; 95% CI: 0.66-0.99; P = .036), and Senegalimassilia (IVW's OR = 0.75; 95% CI: 0.59-0.96; P = .024). Following replication verification analysis, Catenibacterium (FDR = .04) at the genus level were positively correlated to the risk of TMD. The reverse MR analysis revealed that TMD had no significant effect on the 6 GMs. The findings of this MR study support a strong and negative causal association between genus Catenibacterium and TMD, highlighting potential targets for the prevention and treatment of TMD.
Collapse
Affiliation(s)
- Tianyi Ni
- Department of Otorhinolaryngology Head & Neck Surgery, Huadong Hospital Affiliated Fudan University, Shanghai, China
| | - Ziyu Shen
- Department of Algology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Hekai Shi
- Department of General Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zhao Han
- Department of Otorhinolaryngology Head & Neck Surgery, Huadong Hospital Affiliated Fudan University, Shanghai, China
| |
Collapse
|
4
|
Pan J, Zhang B, Ren W. A preliminary exploration of a predictive model and nomogram for the efficacy of compound digestive enzyme therapy based on serum (PGI, PGII, VIP, and PRDX1) in patients with functional dyspepsia. BMC Gastroenterol 2025; 25:413. [PMID: 40437392 PMCID: PMC12121025 DOI: 10.1186/s12876-025-04024-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 05/22/2025] [Indexed: 06/01/2025] Open
Abstract
OBJECTIVE This study aimed to explore the feasibility of constructing compound digestive enzyme therapeutic effect prediction model based on serum pepsinogen I (PGI), pepsinogen II (PGII), vasoactive intestinal peptide (VIP), and peroxidase 1 (PRDX1) in patients with functional dyspepsia (FD), and draw nomograms, to provide reference for the selection of clinical treatment. METHODS A total of 249 FD patients who visited the Department of Gastroenterology in our hospital from January 2021 to December 2024 were selected, and the preoperative clinical and laboratory indicators were collected. the patient cohort was split in a 7:3 ratio into a training set (n = 174) and a validation set (n = 75). The risk factors were screened by univariate and multivariate logistic regression in the training set, and the nomogram model was constructed. The receiver operating characteristic curve (ROC) was drawn and the calibration curve was used to evaluate the effectiveness of the model. The model was verified in the verification set, and the clinical value was evaluated by decision curve analysis (DCA). RESULTS The results of multivariate logistic regression showed that PGI, PGII, VIP, PRDX1, white blood cell count, aspartate aminotransferase and high density lipoprotein cholesterol were the independent risk factors for poor efficacy of compound digestive enzymes in the treatment of FD. The C-index was 0.830 and 0.827, respectively, the area under the ROC curve (AUC) was 0.835 (95% CI: 0.792-0.941) and 0.835 (95% CI: 0.687-0.983), and the sensitivity and specificity were 0.768, 0.857, and 0.778, 0.780, respectively. CONCLUSION The therapeutic effect prediction model of compound digestive enzyme base on serum PGI, PGII, VIP, PRDX1 in patients with FD has some clinical value, but it still need to be further verified by large sample size and multi-center study.
Collapse
Affiliation(s)
- Jiachao Pan
- Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, No. 105 Jiefang Road, Lixia District, Jinan City, Shandong Province, 250013, China
| | - Bo Zhang
- Department of Internal Medicine Second Ward, Jinan Hospital, Jinan, 250013, China
| | - Wenqiang Ren
- Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, No. 105 Jiefang Road, Lixia District, Jinan City, Shandong Province, 250013, China.
| |
Collapse
|
5
|
Cheng J, Cao J, Yang Y, Wang Y, Hu X, Liu Z, Huang Q, Ye Z, Xian W, Sun K, Xie M, Zheng J, Zhao Y, Zheng R, Tan H, Wang X, Zhang X, Wang C, Li C. Multi-omics analysis unraveling stemness features associated with oncogenic dedifferentiation in 12 cancers. Cancer Lett 2025; 625:217816. [PMID: 40412796 DOI: 10.1016/j.canlet.2025.217816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 05/16/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Tumorigenesis is typically accompanied by cellular dedifferentiation and the acquisition of stem cell-like attributes. However, few studies have comprehensively evaluated the putative relationships between these characteristics and various cancers. Here, we integrated gene expression and DNA methylation quantitative trait loci (cis-eQTL and cis-mQTL) data from the blood to perform multi-omics Mendelian randomization analysis. Our analyses revealed 967 stem cell-associated genes (P<0.05) and 11,262 methylation sites (P<0.01) significantly related to 12 cancers. SMAD7 (cg14321542) in colon cancer, IGF2 (cg13508136) in prostate cancer, and FADS1 (cg07005513) in rectal cancer were prioritized as candidate causal genes and regulatory elements. Notably, using cis-eQTL data from the corresponding tissue sites, we detected 16 stem cell-associated genes dramatically causally associated with six cancers (FDR<0.2). The gene THBS3 was particularly common in both blood and stomach tissues and exhibited prognostic significance. Furthermore, it was markedly associated with one microbial metabolic pathway and four immunophenotypes. Functional validation using the ECC12 gastric cancer cell line revealed that the inhibition of its expression could accelerate oxidative phosphorylation and reactive oxygen species production, reduce clonal proliferation ability, and promote the apoptosis of stomach tumor cells. Additionally, based on spatial transcriptomic data from gastrointestinal cancers, the results demonstrated the clusters enriched with the most stem cell-associated genes exhibited significantly enhanced tumor-promoting potency, and the THBS3-expressing cells displayed suppressed oxidative phosphorylation. Overall, this study enhances our understanding of tumorigenic mechanisms and aids in the identification of therapeutic targets.
Collapse
Affiliation(s)
- Jun Cheng
- Department of Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker Detection, The Second Hospital of Shandong University, Shandong, Jinan 250033, China
| | - Jiafan Cao
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Yalan Yang
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Ying Wang
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Xianzhe Hu
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Zhuoyuan Liu
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qiuyin Huang
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhitao Ye
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Xian
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kexin Sun
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Mengyun Xie
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Jiayin Zheng
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Yijun Zhao
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Runhui Zheng
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huo Tan
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoqi Wang
- Medical center of hematology, Xinqiao Hospital of Army Medical University. Chongqing China.
| | - Xi Zhang
- Medical center of hematology, Xinqiao Hospital of Army Medical University. Chongqing China.
| | - Chuanxin Wang
- Department of Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker Detection, The Second Hospital of Shandong University, Shandong, Jinan 250033, China.
| | - Changzheng Li
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510000, China.
| |
Collapse
|
6
|
Shi J, Peng B, Xu R, Chang X, Wang C, Zhou X, Zhang L. Exploration oxidative stress underlying gastroesophageal reflux disease and therapeutic targets identification: a multi-omics Mendelian randomization study. Postgrad Med J 2025; 101:517-525. [PMID: 39671389 DOI: 10.1093/postmj/qgae182] [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: 06/20/2024] [Revised: 10/12/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
INTRODUCTION Gastroesophageal reflux disease (GERD) is a chronic inflammatory gastrointestinal disease, which has no thoroughly effective or safe treatment. Elevated oxidative stress is a common consequence of chronic inflammatory conditions. METHODS We employed Summary-data based MR (SMR) analysis to assess the associations between gene molecular characteristics and GERD. Exposure data were the summary-level data on the levels of DNA methylation, gene expression, and protein expression, which obtained from related methylation, expression, and protein quantitative trait loci investigations (mQTL, eQTL, and pQTL). Outcome data, Genome-wide association study (GWAS) summary statistics of GERD, were extracted from the Ong's study (discovery), the Dönertaş's study (replication), and the FinnGen study (replication). Colocalization analysis was performed to determine if the detected signal pairs shared a causative genetic mutation. Oxidative stress related genes and druggable genes were imported to explore oxidative stress mechanism underlying GERD and therapeutic targets of GERD. The Drugbank database was utilized to conduct druggability evaluation. RESULTS After multi-omics SMR analysis and colocalization analysis, we identified seven key genes for GERD, which were SUOX and SERPING1, DUSP13, SULT1A1, LMOD1, UBE2L6, and PSCA. SUOX was screened out to be the mediator, which suggest that GERD is related to oxidative stress. SERPING1, SULT1A1, and PSCA were selected to be the druggable genes. CONCLUSIONS These findings offered strong support for the identification of GERD treatment targets in the future as well as for the study of the oxidative stress mechanism underlying GERD.
Collapse
Affiliation(s)
- Jiaxin Shi
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150081, China
| | - Bo Peng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150081, China
| | - Ran Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150081, China
| | - Xiaoyan Chang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150081, China
| | - Chenghao Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150081, China
| | - Xiang Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150081, China
| | - Linyou Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150081, China
| |
Collapse
|
7
|
Wang Z, Zhang T, Tang M. Navigating nanotoxicity: Unraveling nanomaterial-induced effects via multi-omics integration. NANOIMPACT 2025; 38:100565. [PMID: 40383513 DOI: 10.1016/j.impact.2025.100565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/14/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
The growing use of nanomaterials in industry and medicine raises significant concerns about their safety, particularly regarding their interactions with biological systems. Traditional toxicological methods, with limited throughput and mechanistic understanding, are increasingly being complemented by omics technologies. Genomics, transcriptomics, proteomics, and metabolomics provide comprehensive insights into the molecular mechanisms of nanomaterial toxicity and enable the identification of potential biomarkers. In addition, single-cell and spatial omics approaches are emerging as powerful tools to assess toxicity at the cellular and tissue levels, revealing heterogeneous responses and spatial distribution of nanomaterials. Despite their advantages, omics technologies face challenges in nanotoxicology, including large, complex data sets, integration difficulties, and a lack of standardized protocols. To address these challenges, we propose the development of new bioinformatics tools, multi-omics integration platforms, and standardized analysis processes to enhance research efficiency and accuracy. These efforts can provide a practical roadmap for integrating the application of omics technologies, including single-cell and spatial approaches, in the study of nanomaterial toxicity studies.
Collapse
Affiliation(s)
- Zhihui Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education; School of Public Health, Southeast University, Nanjing 210009, People's Republic of China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education; School of Public Health, Southeast University, Nanjing 210009, People's Republic of China.
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education; School of Public Health, Southeast University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
8
|
Li Y, Cai J, Xu Y, Zou Y, Xu S, Zheng X, Fu L, Zhang J, Ma X, He Y, Wang X, Deng K, Guo J. Macrophage-myofibroblast transition contributes to the macrophage elimination and functional regeneration in the late stage of nerve injury. Exp Neurol 2025; 387:115194. [PMID: 39993460 DOI: 10.1016/j.expneurol.2025.115194] [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: 01/17/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025]
Abstract
Massive of macrophages are recruited to the injured nerve to remove the axonal and myelin debris for creating a conducive micro-environment for nerve regeneration. However, the fate of macrophages after the debris clearing remains unclear. In this study, we demonstrated that the number of macrophages in the crush injured sciatic nerve of mice peaked at 7 days post injury (dpi) and then decreased significantly in the late stage of nerve injury. Mechanismly, the macrophage elimination was primarily attributed to TGF-β/Smad3 signaling dependent macrophage-myofibroblast transition (MMT), rather than apoptosis or out-migration. Furthermore, MMT caused collagen deposition is conducive to nerve regeneration. Both macrophage depletion via clodronate liposomes and MMT blockade using TGF-β/Smad3 inhibitor significantly reduced collagen deposition and impaired functional nerve regeneration. In summary, the present study indicates that TGF-β/Smad3 regulated MMT contributes to macrophage elimination and functional recovery in the injury nerve.
Collapse
Affiliation(s)
- Yunlun Li
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangzhou, Guangdong Province, China
| | - Jiale Cai
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangzhou, Guangdong Province, China
| | - Yizhou Xu
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China; Department of Spine Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ying Zou
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shuyi Xu
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangzhou, Guangdong Province, China
| | - Xinya Zheng
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lanya Fu
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jiaqi Zhang
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xinrui Ma
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ye He
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xianghai Wang
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangzhou, Guangdong Province, China
| | - Kaixian Deng
- Department of Gynecology, Shunde Hospital of Southern Medical University, Foshan, China
| | - Jiasong Guo
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangzhou, Guangdong Province, China.
| |
Collapse
|
9
|
Chen C, Lan Y, Yan W, Zhang X, Li T, Han J. Exploring Therapeutic Targets for Age-Related Macular Degeneration From Circulating Proteins to Plasma Metabolites in the European Population. Transl Vis Sci Technol 2025; 14:8. [PMID: 40327005 PMCID: PMC12063708 DOI: 10.1167/tvst.14.5.8] [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: 01/06/2025] [Accepted: 04/02/2025] [Indexed: 05/07/2025] Open
Abstract
Purpose To explore the causal associations among circulating proteins, plasma metabolites, and age-related macular degeneration (AMD). Methods We employed Mendelian randomization (MR) analysis and colocalization analysis to discern the causal relationship between proteomes and AMD. This investigation utilized data from protein quantitative trait loci (pQTL) studies in deCODE and the UK Biobank. Additionally, plasma metabolite-related genome-wide association studies (GWAS) data and AMD-related GWAS data were incorporated. Results Our findings confirmed a potential causal relationship between cytoplasmic tryptophanyl-tRNA synthetase 1 (WARS1) and a higher risk of AMD. The observed causal impact of WARS1 on the two subtypes of AMD (dry and wet) align consistently with the aforementioned outcomes. Three plasma metabolites-N-acetyl-kynurenine, N-acetyltyrosine, and caproate (6:0)-were identified as mediators of the causal effect of WARS1 on AMD, and subgroup analysis revealed that N-acetyltyrosine is a specific negative metabolite associated with WARS1 and dry AMD, whereas X-16580 is a specific positive metabolite linked to WARS1 and wet AMD. Conclusions The outcomes of this study suggest a potential causal role of specific circulating proteins in AMD and identified the mediating role of plasma metabolites between WARS1 and AMD by integrating multiple genetic analyses. Nevertheless, further research is essential to validate and strengthen these conclusions. Translational Relevance This study establishes the causal role of specific circulating proteins in AMD and identified the mediating role of plasma metabolites between WARS1 and AMD.
Collapse
Affiliation(s)
- Chengming Chen
- Department of Ophthalmology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou, China
| | - Yanyan Lan
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Weiming Yan
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou, China
| | - Xiaohong Zhang
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou, China
| | - Tian Li
- Tianjin Medical University, Tianjin, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
10
|
Liu L, Sun C, Huang B, Zhao D, Xiong C, Xu F, Wei T. Potential causal association between serum vitamin D levels and intervertebral disc degeneration: A mendelian randomization study. J Orthop Sci 2025; 30:433-439. [PMID: 39034208 DOI: 10.1016/j.jos.2024.07.001] [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: 04/17/2024] [Revised: 06/10/2024] [Accepted: 07/02/2024] [Indexed: 07/23/2024]
Abstract
OBJECTIVES Intervertebral disc degeneration (IDD) is a prevalent musculoskeletal disorder with substantial implications for disability and healthcare expenditures. The role of serum vitamin D (25-Hydroxyvitamin D, 25(OH)D) levels in the pathogenesis of various musculoskeletal conditions has been explored in prior observational studies, suggesting a potential association. While previous observational studies have suggested an association between the two conditions, it might confound the effect of 25(OH)D on IDD. This Mendelian randomization (MR) study seeks to elucidate the causal relationship between 25(OH)D and IDD. METHODS We performed a MR analysis using summary-level data from genome-wide association studies (GWAS) of 25(OH)D (sample size = 441,291 European) and IDD (sample size = 336,439 (cases = 41,669, controls = 294,770) European). Single nucleotide polymorphisms (SNPs) significantly associated with 25(OH)D (p < 5 × 10-8) were selected as instrumental variables. The associations between genetically predicted 25(OH)D and IDD were estimated using the inverse-variance weighted (IVW) method, with sensitivity analyses employing the weighted median, MR-Egger, and MR-PRESSO approaches to assess the robustness of the findings. RESULTS In the primary IVW analysis, genetically predicted 25(OH)D was unrelated associated with IDD (odds ratio (OR) = 0.9671, 95% confidence interval (CI): 0.8956-1.0444, p = 0.39). The results remained consistent across the sensitivity analyses, and no significant directional pleiotropy was detected (MR-Egger intercept: p = 0.64). CONCLUSIONS This study found no obvious evidence that 25(OH)D is causally associated with IDD risks. We call for larger sample size studies to further unravel the potential causal relationship and the exact mechanism.
Collapse
Affiliation(s)
- Libangxi Liu
- Department of Orthopaedics, General Hospital of Central Theater Command of PLA, 627 Wuluo Road, Wuchang District, Wuhan, Hubei, PR China; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, PR China
| | - Chao Sun
- Department of Orthopaedics, General Hospital of Central Theater Command of PLA, 627 Wuluo Road, Wuchang District, Wuhan, Hubei, PR China
| | - Biwang Huang
- Department of Orthopaedics, General Hospital of Central Theater Command of PLA, 627 Wuluo Road, Wuchang District, Wuhan, Hubei, PR China
| | - Dongdong Zhao
- Department of Orthopaedics, General Hospital of Central Theater Command of PLA, 627 Wuluo Road, Wuchang District, Wuhan, Hubei, PR China
| | - Chengjie Xiong
- Department of Orthopaedics, General Hospital of Central Theater Command of PLA, 627 Wuluo Road, Wuchang District, Wuhan, Hubei, PR China; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, PR China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, PR China.
| | - Feng Xu
- Department of Orthopaedics, General Hospital of Central Theater Command of PLA, 627 Wuluo Road, Wuchang District, Wuhan, Hubei, PR China.
| | - Tanjun Wei
- Department of Orthopaedics, General Hospital of Central Theater Command of PLA, 627 Wuluo Road, Wuchang District, Wuhan, Hubei, PR China.
| |
Collapse
|
11
|
Li C, Liu W, Fu A, Yang H, Yi G. Potential therapeutic strategies targeting efferocytosis for inflammation resolution and tissue repair in inflammatory bowel disease. Cell Immunol 2025; 411-412:104957. [PMID: 40315792 DOI: 10.1016/j.cellimm.2025.104957] [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: 08/21/2024] [Revised: 04/11/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025]
Abstract
Efferocytosis, the process by which apoptotic cells (ACs) are recognized and cleared by phagocytes, is a critical mechanism in maintaining intestinal immune homeostasis and promoting the resolution of inflammation. Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), is characterized by chronic intestinal inflammation, wherein defective efferocytosis contributes to the accumulation of ACs, secondary necrosis, and sustained mucosal damage. This review delineates the molecular mechanisms underlying efferocytosis and systematically examines its functional roles across five key intestinal phagocytic cell types: macrophages, dendritic cells (DCs), neutrophils, intestinal epithelial cells (IECs), and Paneth cells (PCs). Particular emphasis is placed on the dysregulation of efferocytosis capacity in IBD pathogenesis and the consequences of impaired apoptotic cell clearance in both professional and non-professional phagocytes. Furthermore, we evaluate emerging therapeutic strategies designed to restore or enhance efferocytosis, including modulation of macrophage polarization, LC3-associated phagocytosis pathways, nanotechnology-enabled delivery systems, and stem cell-based interventions. A comprehensive understanding of cell-type-specific efferocytosis in the intestinal microenvironment offers promising directions for the development of targeted, inflammation-resolving therapies for IBD.
Collapse
Affiliation(s)
- Chaoquan Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Wanting Liu
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Aoni Fu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Haotian Yang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Guanghui Yi
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
12
|
Chen L, Li J, Zhang W, Wang J. Antiepileptic Effects of Acorus tatarinowii Schott in a Rat Model of Epilepsy: Regulation of Metabolic Axes and Gut Microbiota. BIOLOGY 2025; 14:488. [PMID: 40427677 PMCID: PMC12108817 DOI: 10.3390/biology14050488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025]
Abstract
As a phytotherapeutic agent with historical applications in epilepsy management, Acorus tatarinowii Schott (ATS) remains pharmacologically enigmatic, particularly regarding its pathophysiological mechanisms. This knowledge gap significantly hinders the clinical application of ATS-based treatments. To explore the potential of ATS in combating epileptogenesis, we utilized a pentylenetetrazole (PTZ)-induced chronic epilepsy rat model. Brain metabolomic analysis was performed by ultra-performance liquid chromatography coupled with mass spectrometry (UPLC/MS). Principal component analysis (PCA) and orthogonal projections to latent structures-discriminant analysis (OPLS-DA) were performed for screening differential metabolites. Gut microbiota composition was analyzed through 16S rRNA gene sequencing and examined using Spearman correlation analysis. The results show that oral ATS (50 mg/kg) significantly improved the seizure latency and pathology of rats with epilepsy. Ascorbate and aldarate metabolism, glycerophospholipid metabolism, arachidonic acid metabolism, and intestinal flora were crucial for ATS's ability to counteract epilepsy. The therapeutic effects of ATS against epilepsy were investigated with brain metabolomics and gut microbiota analysis, providing the basis for further comprehensive research.
Collapse
Affiliation(s)
- Liang Chen
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (L.C.); (J.L.); (W.Z.)
| | - Jiaxin Li
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (L.C.); (J.L.); (W.Z.)
| | - Wenhui Zhang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (L.C.); (J.L.); (W.Z.)
| | - Jiepeng Wang
- School of Basic Medical Sciences, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| |
Collapse
|
13
|
Yao H, Chen J, Wang Y, Li Y, Tang P, Liang M, Jiang Q. Uncovering therapeutic targets for Pre-eclampsia and pregnancy hypertension via multi-tissue data integration. BMC Pregnancy Childbirth 2025; 25:479. [PMID: 40269770 PMCID: PMC12020376 DOI: 10.1186/s12884-025-07608-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 04/15/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Pre-eclampsia (PE) and pregnancy hypertension (PH) are common and serious complications during pregnancy, which can lead to maternal and fetal death in severe cases. Therefore, further research on the potential therapeutic targets of PE and PH is of great significance for developing new treatment strategies. METHODS This study used the summary data-based Mendelian randomization (SMR) method to analyze expression quantitative trait loci (eQTL) data from blood, aorta, and uterus with Genome-wide association studies (GWAS) data on PE and PH, exploring potential genetic loci involved in PE and PH. Since proteinuria is a clinical manifestation of PE, we also analyzed genes related to the kidney and PE. The HEIDI test was used for heterogeneity testing, and results were adjusted using FDR. The cis-eQTL data were obtained from the blood summary-level data of the eQTLGen Consortium and the aorta and uterus data from the V8 release of the GTEx eQTL summary data. The GWAS data for PE and PH were obtained from the FinnGen Documentation of R10 release. This study utilized the STROBE-MR checklist for reporting Mendelian Randomization (MR) studies. RESULTS This study identified several potential therapeutic targets by integrating eQTL data from blood, uterus, and aorta with GWAS data for PE and PH, as well as kidney eQTL data with GWAS data for PE. Additionally, the study discovered some genes with common roles in PE and PH, offering new insights into the shared pathological mechanisms of these two conditions. These findings not only provide new clues to the pathogenesis of PE and PH but also offer crucial foundational data for the development of future therapeutic strategies. CONCLUSION This study revealed multiple potential therapeutic targets for PE and PH, providing new insights for basic experimental research and clinical treatment to mitigate the severe consequences of PE and PH. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Hang Yao
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, China
| | - Jiahao Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Wang
- Graduate School of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yuxin Li
- Graduate School of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Peiyu Tang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, China
| | - Mingpeng Liang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, China
| | - Qingling Jiang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, China.
| |
Collapse
|
14
|
Wang T, He Q, Chan KHK. A Multi-omics approach to identify and validate shared genetic architecture in rheumatoid arthritis, multiple sclerosis, and type 1 diabetes: integrating GWAS, GEO, MSigDB, and scRNA-seq data. Funct Integr Genomics 2025; 25:91. [PMID: 40254686 PMCID: PMC12009781 DOI: 10.1007/s10142-025-01598-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/22/2025]
Abstract
The notable comorbidity among autoimmune diseases underscores their shared genetic underpinnings, particularly evident in rheumatoid arthritis (RA), type 1 diabetes (T1D), and multiple sclerosis (MS). However, the exact components and mechanisms of this shared genetic structure remain poorly understood. Here we show that ROMO1 is a key shared genetic component among RA, MS, and T1D. Using differential gene expression (DGE) and LASSO regression analyses of bulk RNA-seq data from whole blood tissues, we identified ROMO1 as a potential shared genetic factor. A multi-sample analysis with external Gene Expression Omnibus (GEO) data revealed ROMO1's consistent association with immune cell patterns across tissues in all three diseases. Single-gene Gene Set Enrichment Analysis (GSEA) suggested ROMO1's involvement in the reactive oxygen species (ROS) pathway, which was further substantiated by conjoint analysis with 256 ROS pathway-related genes(ROSGs) from Molecular Signatures Database (MSigDB). Single-gene Receiver Operating Characteristic (ROC) analysis highlighted ROMO1's potential as a disease biomarker. Single-cell RNA sequencing (scRNA-seq) analysis showed significantly altered ROMO1 expression in monocytes and other immune cells compared to healthy control (HC). Immune infiltration analysis revealed ROMO1's significant association with monocytes across all three diseases. Furthermore, two-sample Mendelian randomization (MR) analysis using genome-wide association studies (GWAS) data demonstrated that ROMO1 could regulate epitopes on monocytes, potentially lowering autoimmune disease risk. Our findings clarify the importance of ROMO1 in the shared genetic architecture of RA, MS, and T1D, and its underlying mechanism in disease development.
Collapse
Affiliation(s)
- Tailin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Qian He
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Kei Hang Katie Chan
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China.
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China.
- Department of Epidemiology, Centre for Global Cardiometabolic Health, Brown University, Rhode Island, USA.
- City University of Hong Kong, Room 1A-313, 3/F, Block 1, To Yuen Building, 31 To Yuen Street, Kowloon Tong, Kowloon, HKSAR, China.
| |
Collapse
|
15
|
Lei Y, Lin H, Chen Y, Wan B, Ao C, Liu J, Wang W. Epigenetic regulation of physiological resilience to ammonia nitrogen stress in the Pacific whiteleg shrimp Penaeus vannamei: Evidence from genome-wide DNA methylation dynamics. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101510. [PMID: 40220697 DOI: 10.1016/j.cbd.2025.101510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
Although DNA methylation has emerged as an essential epigenetic mechanism modulating organismal responses to abiotic stresses, its involvement in the physiological resilience of marine invertebrates like shrimp to ammonia nitrogen toxicity remains enigmatic. Here, we performed the first comprehensive dissection of genome-wide DNA methylation dynamics in the Pacific whiteleg shrimp Penaeus vannamei exposed to ammonia nitrogen, based on whole-genome bisulfite sequencing and transcriptome analyses. In the genome of P. vannamei, three DNA methyltransferases (DNMT1, DNMT2 and DNMT3a), one DNA demethylase (TET2) and four methyl-CpG binding proteins (MBD2, MBD4, Kaiso, and UHRF1) were present. About 1.68-1.87 % of cytosine nucleotides were methylated, and higher percentages of cytosines in the CpG context (5.23 %-6.34 %) was methylated compared with the CHG and CHH contexts. Methylated cytosines were mostly enriched in the coding DNA sequence, and methylation peaks occurred near the transcription end sites. Following ammonia exposure, 4203 differentially expressed genes (DEGs) and 1100 differentially methylated genes (DMGs) were identified. The DMGs accounted for 4.4 % of the total gene reservoir in P. vannamei genome, and 212 shared genes were found between the DEGs and DMGs. Genes exhibiting significant methylation and expression changes were enriched in various pathways including the FoxO signaling pathway, autophagy and endocytosis. Among them was a group of genes related to energy metabolism, antioxidation response and detoxification metabolism, highlighting involvement of DNA methylation in fine-tuning these crucial physiological processes. These findings provide new insights into the regulatory roles of DNA methylation in the physiological resilience of marine invertebrates to aquatic stressors.
Collapse
Affiliation(s)
- Yiguo Lei
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Hanliang Lin
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yunhua Chen
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Boquan Wan
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Chunmei Ao
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Jianyong Liu
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Wei Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China.
| |
Collapse
|
16
|
Bai Z, Zhang G. The application of mendelian randomization in cancer research: a bibliometric analysis. Discov Oncol 2025; 16:463. [PMID: 40185974 PMCID: PMC11971120 DOI: 10.1007/s12672-025-02226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
Cancer is a major public health and economic issue faced globally today, significantly affecting human health and life. Due to various constraints, exploring the causal relationship between risk factors and cancer is challenging and not exactly accurate. The advent of Mendelian randomization (MR) effectively addresses these issues, providing new avenues for exploring causal relationships. We downloaded literature related to the application of MR in cancer from the Web of Science Core Collection (WoSCC) from 2005 to October 21, 2024, limiting the document type to articles and the language to English, resulting in a total of 2058 articles. We downloaded them in plain text format and extracted information on countries, authors, institutions, keywords, journals, citation counts, and publication dates, utilizing VOSviewer, CiteSpace, and R language for bibliometric analysis. From 2005 to 2024, the number of publications on the application of MR in cancer has shown a growth trend. China was the most productive country (1305); the University of Bristol was the most prolific institution (213); Smith, George Davey published the most articles in this field (59) with a total citation count of 5344; the most prolific journal in this field is Scientific Reports (71). Chronic diseases and cancer, inflammation and cancer, and sex hormones and cancer are three hot topics in the current research on the application of MR in cancer. In the future, optimizing statistical methods, standardizing research processes, collecting data from a broader range of populations, expanding data scale, and integrating other research methods to enhance research quality will be the development trends of MR in cancer research. In summary, this study employed bibliometric methods to comprehensively analyze the literature on the application of MR in cancer over the past 20 years, evaluating the historical development, current applications, research hotspots, and future trends of MR in the field of cancer.
Collapse
Affiliation(s)
- Zhongtao Bai
- Department of General Surgery, Suzhou Hospital of Anhui Medical University, Suzhou, 234000, Anhui Province, China
| | - Genlong Zhang
- Department of General Surgery, Suzhou Hospital of Anhui Medical University, Suzhou, 234000, Anhui Province, China.
| |
Collapse
|
17
|
Li C, Hao R, Li C, Liu L, Ding Z. Integration of single-cell and bulk RNA sequencing data using machine learning identifies oxidative stress-related genes LUM and PCOLCE2 as potential biomarkers for heart failure. Int J Biol Macromol 2025; 300:140793. [PMID: 39929468 DOI: 10.1016/j.ijbiomac.2025.140793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/24/2025] [Accepted: 02/06/2025] [Indexed: 02/23/2025]
Abstract
Oxidative stress (OS) is a pivotal mechanism driving the progression of cardiovascular diseases, particularly heart failure (HF). However, the comprehensive characterisation of OS-related genes in HF remains largely unexplored. In the present study, we analysed single-cell RNA sequencing datasets from the Gene Expression Omnibus and OS gene sets from GeneCards. We identified 167 OS-related genes potentially linked to HF by applying algorithms, such as AUCell, UCell, singscore, ssgsea, and AddModuleScore, combined with correlation analysis. Subsequently, we used feature selection algorithms, including least absolute shrinkage and selection operator, XGBoost, Boruta, random forest, gradient boosting machines, decision trees, and support vector machine recursive feature elimination, to identify lumican (LUM) and procollagen C-endopeptidase enhancer 2 (PCOLCE2) as key biomarker candidates with significant diagnostic potential. Bulk RNA-sequencing confirmed their elevated expression in patients with HF, highlighting their predictive utility. Single-cell analysis further revealed their upregulation primarily in fibroblasts, emphasising their cell-specific role in HF. To validate these findings, we developed a transverse aortic constriction-induced HF mouse model that showed enhanced cardiac OS activity and significant PCOLCE2 upregulation in the HF group. These results provide strong evidence of the involvement of OS-related mechanisms in HF. Herein, we propose a diagnostic strategy that provides novel insights into the molecular mechanisms underlying HF. However, further studies are required to validate its clinical utility and ensure its application in the diagnosis of HF.
Collapse
Affiliation(s)
- Chaofang Li
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ruijinlin Hao
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chuanfu Li
- Departments of Surgery, East Tennessee State University, Johnson City, TN 37614, USA
| | - Li Liu
- Department of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhengnian Ding
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
18
|
Tong H, Zhao Y, Cui Y, Yao J, Zhang T. Multi-omic studies on the pathogenesis of Sepsis. J Transl Med 2025; 23:361. [PMID: 40128726 PMCID: PMC11934817 DOI: 10.1186/s12967-025-06366-w] [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: 11/20/2024] [Accepted: 03/08/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Sepsis is a life-threatening inflammatory condition, and its underlying genetic mechanisms are not yet fully elucidated. We applied methods such as Mendelian randomization (MR), genetic correlation analysis, and colocalization analysis to integrate multi-omics data and explore the relationship between genetically associated genes and sepsis, as well as sepsis-related mortality, with the goal of identifying key genetic factors and their potential mechanistic pathways. METHODS To identify therapeutic targets for sepsis and sepsis-related mortality, we conducted an MR analysis on 11,643 sepsis cases and 1,896 cases of 28-day sepsis mortality from the UK Biobank cohort. The exposure data consisted of 15,944 potential druggable genes (expression quantitative trait loci, eQTL) and 4,907 plasma proteins (protein quantitative trait loci, pQTL). We then performed sensitivity analysis, SMR analysis, reverse MR analysis, genetic correlation analysis, colocalization analysis, enrichment analysis, and protein-protein interaction network analysis on the overlapping genes. Validation was conducted using 17,133 sepsis cases from FinnGen R12. Drug prediction and molecular docking were subsequently used to further assess the therapeutic potential of the identified drug targets, while PheWAS was used to evaluate potential side effects. Finally, mediation analysis was conducted to identify the mediating role of related metabolites. RESULTS The MR analysis results identified a significant causal relationship between 24 genes and sepsis. The robustness of these causal associations was further strengthened by SMR analysis, sensitivity analysis, and reverse MR analysis. Genetic correlation analysis revealed that only two of these genes were genetically correlated with sepsis. Colocalization analysis showed that only one gene was closely associated with sepsis, while validation using the FinnGen dataset identified three genes. In the MR analysis of 28-day sepsis mortality, seven genes were found to have significant associations, with reverse MR analysis excluding one gene. The remaining genes passed sensitivity analysis, with no significant genes identified in genetic correlation and colocalization analyses. Molecular docking demonstrated excellent binding affinity between drugs and proteins with available structural data. PheWAS at the gene level did not reveal any potential side effects of the related drugs. CONCLUSIONS The identified drug targets, associated pathways, and metabolites have enhanced our understanding of the complex relationships between genes and sepsis. These genes and metabolites can serve as effective targets for sepsis treatment, paving new pathways in this field and laying a foundation for future research.
Collapse
Affiliation(s)
- Hongjie Tong
- Department of Critical Care Medicine, Jinhua Hospital Affiliated to Zhejiang University, Jinhua, Zhejiang, China
- Zhejiang University School of Medicine, Hangzhou, China
| | - Yuhang Zhao
- Department of Neurology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Ying Cui
- Department of Critical Care Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jiali Yao
- Department of Critical Care Medicine, Jinhua Hospital Affiliated to Zhejiang University, Jinhua, Zhejiang, China.
| | - Tianlong Zhang
- Department of Critical Care Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
19
|
Peng S, Fang J, Mo W, Hu G, Wu S. Identifying cross-tissue molecular targets of lung function by multi-omics integration analysis from DNA methylation and gene expression of diverse human tissues. BMC Genomics 2025; 26:289. [PMID: 40128644 PMCID: PMC11931835 DOI: 10.1186/s12864-025-11476-2] [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: 01/27/2025] [Accepted: 03/12/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Previous studies have reported several genetic loci associated with lung function. However, the mediating mechanism between these genetic loci and lung function phenotype is rarely explored. In this research, we used a cross-tissue multi-omics post-GWAS analysis to explain the associations between DNA methylation, gene expression, and lung function. METHODS We conducted integration analyses of lung function traits using genome-wide association study (GWAS) summary data alongside expression quantitative trait loci (eQTLs) and DNA methylation quantitative trait loci (mQTLs) derived from whole blood, utilizing multi-omics SMR and Bayesian colocalization analysis. Considering the genetic differences of tissues, we replicated the shared causal signals of eQTLs and lung function in 48 diverse tissues and the shared causal signals of mQTLs and lung function in 8 diverse tissues. Multi-trait colocalization analyses were utilized to identify the causal signals between gene expression in blood, blood cell traits, and lung function, as well as between cross-tissue gene expression in diverse tissues and lung function. RESULTS Eight genes from blood tissue were prioritized as FEV1 causal genes using multi-omics SMR analysis and COLOC colocalization analysis: EML3, UBXN2A, ROM1, ZBTB38, RASGRP3, FAIM, PABPC4, and SNIP1. Equally, five genes (CD46, EML3, UBXN2A, ZBTB38, and LMCD1) were prioritized as FVC causal genes and one gene (LMCD1) was prioritized as FEV1/FVC causal genes. The causal signals between 8 genes (EML3, ROM1, UBXN2A, ZBTB38, RASGRP3, FAIM, PABPC4, and CD46) and lung function were successfully replicated in diverse tissues. More importantly, MOLCO colocalization analysis showed that 3 genes (CD46, LMCD1, and ZBTB38) expression in blood, blood cell traits, and lung function traits shared the same causal signals. Finally, through cross-tissue colocalization analysis of multiple traits, we found that the heart-lung axis EML3 expressions and lung function mediate the same causal signal. CONCLUSION This study identified potential cross-tissue molecular targets associated with lung function traits from DNA methylation and gene expression of diverse tissues and explored the probable regulation mechanism of these molecular targets. This provides multi-omics and cross-tissue evidence for the molecular regulation mechanism of lung function and may provide new insight into the influence of crosstalk between organs and tissues on lung function.
Collapse
Affiliation(s)
- Shisheng Peng
- The Tenth Affiliated Hospital, Southern Medical University, Dongguan People's Hospital, Dongguan, Guangdong, 523059, China
| | - Jinlong Fang
- The Tenth Affiliated Hospital, Southern Medical University, Dongguan People's Hospital, Dongguan, Guangdong, 523059, China
| | - Weiliang Mo
- The Tenth Affiliated Hospital, Southern Medical University, Dongguan People's Hospital, Dongguan, Guangdong, 523059, China
| | - Guodong Hu
- The Tenth Affiliated Hospital, Southern Medical University, Dongguan People's Hospital, Dongguan, Guangdong, 523059, China.
| | - Senquan Wu
- The Tenth Affiliated Hospital, Southern Medical University, Dongguan People's Hospital, Dongguan, Guangdong, 523059, China.
| |
Collapse
|
20
|
Shen C, Liu H, Chen Y, Liu M, Wang Q, Liu J, Liu J. Helicobacter pylori induces GBA1 demethylation to inhibit ferroptosis in gastric cancer. Mol Cell Biochem 2025; 480:1845-1863. [PMID: 39283563 DOI: 10.1007/s11010-024-05105-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 08/21/2024] [Indexed: 02/21/2025]
Abstract
This research investigates potential therapeutic targets for gastric cancer, focusing on ferroptosis-related genes. Gastric cancer is known for its lower survival rates, necessitating new treatment strategies. This study employed Mendelian randomization to identify ferroptosis-related genes and methylation sites in gastric cancer, examining correlations between Helicobacter pylori infection, GBA1 gene expression, and promoter methylation with single-cell datasets and the TCGA-STAD database. We used Helicobacter pylori-infected gastric cancer cell models and used next-generation sequencing to monitor methylation changes pre- and post-infection. GBA1 expression levels were assessed via qRT-PCR and Western blot both before and after infection. The effect of Helicobacter pylori on GC cell proliferation was analyzed using CCK-8 and EdU assays after knocking down the GBA1 gene. The association between Helicobacter pylori infection and ferroptosis, including its reversibility after GBA1 knockdown, was evaluated using FerrOrange, GSH, MDA, and C11-BODIPY assays. Mass spectrometry measured the impact of Helicobacter pylori and GBA1 knockdown on lipid metabolism. An in vivo subcutaneous tumor-bearing model was also established to confirm these findings. Mendelian randomization analysis revealed that high GBA1 expression and reduced methylation levels of its promoter are risk factors for gastric cancer. Single-cell sequencing and TCGA-STAD datasets indicated a positive correlation between Helicobacter pylori infection and GBA1 expression, with a concurrent negative correlation between GBA1 promoter methylation and GBA1 expression. In gastric cancer cell lines, Helicobacter pylori infection was observed to enhance GBA1 expression and decrease methylation levels at its promoter. Additionally, Helicobacter pylori promoted GC cell proliferation, an effect mitigated by knocking down GBA1. Infection also reduced lipid peroxidation, increased glutathione levels, and impeded ferroptosis in GC cells; however, these effects were reversed following GBA1 knockdown. Changes in sphingolipid metabolism induced by I were detected in GC cell lines. In vivo experiments using a subcutaneous tumor-bearing model demonstrated that Helicobacter pylori infection fosters tumorigenesis in GC cells. Our study demonstrates that Helicobacter pylori infection triggers demethylation and upregulation of GBA1, subsequently inhibiting ferroptosis in gastric cancer cells. These findings suggest that targeting the GBA1 pathway may offer a novel therapeutic approach for managing gastric cancer.
Collapse
Affiliation(s)
- Chenjie Shen
- Department of Oncology, Jiangnan University Affiliated Hospital, No. 1000, Hefeng Road, Wuxi, Jiangsu, China
- Department of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu, China
| | - Huan Liu
- Department of Oncology, Jiangnan University Affiliated Hospital, No. 1000, Hefeng Road, Wuxi, Jiangsu, China
- Department of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu, China
| | - Yuhan Chen
- Department of Oncology, Jiangnan University Affiliated Hospital, No. 1000, Hefeng Road, Wuxi, Jiangsu, China
- Department of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu, China
| | - Mengpei Liu
- Department of Oncology, Jiangnan University Affiliated Hospital, No. 1000, Hefeng Road, Wuxi, Jiangsu, China
- Department of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu, China
| | - Qian Wang
- Department of Oncology, Jiangnan University Affiliated Hospital, No. 1000, Hefeng Road, Wuxi, Jiangsu, China
| | - Jiaqi Liu
- Department of Integrative Medicine, Huashan Hospital Fudan University, 12 Middle Wulumuqi Road, Shanghai, China.
| | - Jingjing Liu
- Department of Oncology, Jiangnan University Affiliated Hospital, No. 1000, Hefeng Road, Wuxi, Jiangsu, China.
- Department of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu, China.
| |
Collapse
|
21
|
Hou K, Tan Y, Zhang Q. Investigating the causal relationship between skin microbiota and hypertrophic scar using bidirectional mendelian randomization. Burns 2025; 51:107376. [PMID: 39778466 DOI: 10.1016/j.burns.2025.107376] [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: 08/30/2024] [Revised: 12/16/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
BACKGROUND Hypertrophic scar (HS) is acknowledged as a pathological fibro-proliferative disease of the dermis, resulting from excessive connective tissue growth. HS significantly impacts patient quality of life due to both social and functional issues. Despite various treatments, therapeutic effectiveness remains limited, necessitating further exploration of underlying factors and mechanisms. OBJECTIVE The current study was designed to determine the causal relationship between skin microbiota and HS employing a bidirectional Mendelian randomization (MR) approach. METHODS We utilized genome-wide association study (GWAS) data from the PopGen cohort and the FinnGen database. Independent single nucleotide polymorphisms (SNPs) linked to the skin microbiota were identified as instrumental variables (IVs) chosen for the two-sample MR analysis. Key analytical approaches included inverse variance weighting (IVW), MR-Egger, simple median, simple mode, and weighted mode, with MR-Egger intercept test and Cochrane's Q test used to detect potential horizontal pleiotropy and heterogeneity. RESULTS The two-sample MR analysis identified significant causal relationships between specific skin microbiota features and HS. Notably, Enhydrobacter, Micrococcus, and Acinetobacter on moist skin exhibited protective effects against HS, whereas Finegoldia and Lactobacillales on dry skin were linked to an increased risk of HS. Sensitivity analyses verified the strength of these results, revealing no notable horizontal pleiotropy or heterogeneity. CONCLUSION Our research reveals a unidirectional causal relationship between certain skin microbiota and HS, suggesting that modulation of skin microbiota could be a novel therapeutic approach for HS management. These results emphasize the significance of considering skin microbiota in the pathogenesis and treatment of HS.
Collapse
Affiliation(s)
- Kai Hou
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yufang Tan
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
22
|
Zhang H, Tian Y, Xu C, Chen M, Xiang Z, Gu L, Xue H, Xu Q. Crosstalk between gut microbiotas and fatty acid metabolism in colorectal cancer. Cell Death Discov 2025; 11:78. [PMID: 40011436 DOI: 10.1038/s41420-025-02364-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/30/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common malignancy globally and the second leading cause of cancer-related mortality. Its development is a multifactorial and multistage process influenced by a dynamic interplay between gut microbiota, environmental factors, and fatty acid metabolism. Dysbiosis of intestinal microbiota and abnormalities in microbiota-associated metabolites have been implicated in colorectal carcinogenesis, highlighting the pivotal role of microbial and metabolic interactions. Fatty acid metabolism serves as a critical nexus linking dietary patterns with gut microbial activity, significantly impacting intestinal health. In CRC patients, reduced levels of short-chain fatty acids (SCFAs) and SCFA-producing bacteria have been consistently observed. Supplementation with SCFA-producing probiotics has demonstrated tumor-suppressive effects, while therapeutic strategies aimed at modulating SCFA levels have shown potential in enhancing the efficacy of radiation therapy and immunotherapy in both preclinical and clinical settings. This review explores the intricate relationship between gut microbiota, fatty acid metabolism, and CRC, offering insights into the underlying mechanisms and their potential translational applications. Understanding this interplay could pave the way for novel diagnostic, therapeutic, and preventive strategies in the management of CRC.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Yuan Tian
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Chunjie Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Miaomiao Chen
- Department of Radiology, Huashan Hospital, National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200040, PR China
| | - Zeyu Xiang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Lei Gu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Hanbing Xue
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Qing Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
23
|
Han S, Cho SA, Choi W, Eilbeck K, Coon H, Nho K, Lee Y. Interaction of genetic variants and methylation in transcript-level expression regulation in Alzheimer's disease by multi-omics data analysis. BMC Genomics 2025; 26:170. [PMID: 39979805 PMCID: PMC11844006 DOI: 10.1186/s12864-025-11362-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) presents a significant public health problem and major cause of dementia. Not only genetic but epigenetic factors contribute to complex and heterogeneous molecular mechanisms underlying AD risk; in particular, single nucleotide polymorphisms (SNPs) and DNA methylation can lead to dysregulation of gene expression in the AD brain. Each of these regulators has been independently studied well in AD progression, however, their interactive roles, particularly when they are located differently, still remains unclear. Here, we aimed to explore the interplay between SNPs and DNA methylation in regulating transcript expression levels in the AD brain through an integrative analysis of whole-genome sequencing, RNA-seq, and methylation data measured from the dorsolateral prefrontal cortex. RESULTS We identified 179 SNP-methylation combination pairs that showed statistically significant interactions associated with the expression of 67 transcripts (63 unique genes), enriched in functional pathways, including immune-related and post-synaptic assembly pathways. Particularly, a number of HLA family genes (HLA-A, HLA-B, HLA-C, HLA-DRB1, HLA-DRB5, HLA-DPA1, HLA-K, HLA-DQB1, and HLA-DMA) were observed as having expression changes associated with the interplay. CONCLUSIONS Our findings especially implicate immune-related pathways as targets of these regulatory interactions. SNP-methylation interactions may thus contribute to the molecular complexity underlying immune-related pathogenies in AD patients. Our study provides a new molecular knowledge in the context of the interplay between genetic and epigenetic regulations, in that it concerns transcript expression status in AD.
Collapse
Affiliation(s)
- Seonggyun Han
- Department of Biomedical Informatics, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Soo-Ah Cho
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Wongyung Choi
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Karen Eilbeck
- Department of Biomedical Informatics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hilary Coon
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences and Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Younghee Lee
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
24
|
Xu Y, Liu S, Zhou Z, Qin H, Zhang Y, Zhang G, Ma H, Han X, Liu H, Liu Z. Integrated multi-omics analysis revealed the molecular networks and potential targets of cellular senescence in Alzheimer's disease. Hum Mol Genet 2025; 34:381-391. [PMID: 39690817 DOI: 10.1093/hmg/ddae189] [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: 06/10/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024] Open
Abstract
Cellular senescence (CS) is a hallmark of Alzheimer's disease (AD). However, the mechanisms through which CS contributes to AD pathogenesis remain poorly understood. We found that CS level in AD was higher compared with the healthy control group. Transcriptome-based differential expression analysis identified 113 CS-related genes in blood and 410 in brain tissue as potential candidate genes involved in AD. To further explore the causal role of these genes, an integrative mendelian randomization analysis was conducted, combining AD genome-wide association study summary statistics with expression quantitative trait loci (eQTL) and DNA methylation quantitative trait loci (mQTL) data from blood samples, which identified five putative AD-causal genes (CENPW, EXOSC9, HSPB11, SLC44A2, and SLFN12) and 18 corresponding DNA methylation probes. Additionally, integrative analysis between eQTLs and mQTLs from blood uncovered two genes and 12 corresponding regulatory elements involved in AD. Furthermore, two genes (CDKN2B and ITGAV) were prioritized as putative causal genes in brain tissue and were validated through in vitro experiments. The multi-omics integration study revealed the potential role and underlying biological mechanisms of CS driven by genetic predisposition in AD. This study contributed to fundamental understanding of CS in AD pathogenesis and facilitated the identification of potential therapeutic targets for AD prevention and treatment.
Collapse
Affiliation(s)
- Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
| | - Zhaokai Zhou
- Department of Urology Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
| | - Hongzhuo Qin
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
| | - Hongxuan Ma
- Department of Kidney Transportation, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
- Interventional Institute of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
| | - Huimin Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
- Interventional Institute of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou, Henan 450052, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Dongcheng District, Beijing 100730, China
| |
Collapse
|
25
|
Li S, Yuan H, Li L, Li Q, Lin P, Li K. Oxidative Stress and Reprogramming of Lipid Metabolism in Cancers. Antioxidants (Basel) 2025; 14:201. [PMID: 40002387 PMCID: PMC11851681 DOI: 10.3390/antiox14020201] [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: 12/10/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Oxidative stress is a common event involved in cancer pathophysiology, frequently accompanied by unique lipid metabolic reprogramming phenomena. Oxidative stress is caused mainly by an imbalance between the production of reactive oxygen species (ROS) and the antioxidant system in cancer cells. Emerging evidence has reported that oxidative stress regulates the expression and activity of lipid metabolism-related enzymes, leading to the alteration of cellular lipid metabolism; this involves a significant increase in fatty acid synthesis and a shift in the way in which lipids are taken up and utilized. The dysregulation of lipid metabolism provides abundant intermediates to synthesize biological macromolecules for the rapid proliferation of cancer cells; moreover, it contributes to the maintenance of intracellular redox homeostasis by producing a variety of reducing agents. Moreover, lipid derivatives and metabolites play critical roles in signal transduction within cancer cells and in the tumor microenvironment that evades immune destruction and facilitates tumor invasion and metastasis. These findings suggest a close relationship between oxidative stress and lipid metabolism during the malignant progression of cancers. This review focuses on the crosstalk between the redox system and lipid metabolic reprogramming, which provides an in-depth insight into the modulation of ROS on lipid metabolic reprogramming in cancers and discusses potential strategies for targeting lipid metabolism for cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Ping Lin
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (S.L.); (H.Y.); (L.L.); (Q.L.)
| | - Kai Li
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (S.L.); (H.Y.); (L.L.); (Q.L.)
| |
Collapse
|
26
|
An Z, Zeng M, Wang X. A bidirectional Mendelian randomization study integrating genome-wide association studies, expression quantitative trait locus, and methylation quantitative trait locus data revealed causal relationship between heavy cigarette dependence and Barrett's esophagus. SAGE Open Med 2025; 13:20503121251316595. [PMID: 39925959 PMCID: PMC11806485 DOI: 10.1177/20503121251316595] [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: 10/15/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025] Open
Abstract
Background The association between smoking dependence and the risk of developing Barrett's esophagus remains unclear. This study aimed to investigate whether a causal relationship exists between smoking dependence and Barrett's esophagus, using Mendelian randomization analysis. Methods Two-sample Mendelian randomization analysis was conducted to evaluate the impact of smoking and Barrett's esophagus. Additionally, we applied summary data-based Mendelian randomization techniques to combine information from genome-wide association studies (GWAS) with expression quantitative trait locus and methylation quantitative trait locus. Results Multivariable Mendelian randomization showed an association between smoking per day (odds ratio = 1.2, 95% confidence interval: 1.038-1.38, p = 0.014) or current smoking (odds ratio = 2.41, 95% confidence interval: 1.06-5.5, p = 0.037) and Barrett's esophagus. Inverse variance-weighted methods of bidirectional Mendelian randomization also revealed that smoking per day was significantly associated with elevated risks of Barrett's esophagus (odds ratio = 1.34, 95% confidence interval: 1.092-1.649, p = 0.005), while Barrett's esophagus was also a susceptibility factor for smoking per day (odds ratio = 1.05, 95% confidence interval: 1.017-1.087, p = 0.003). By incorporating consistent summary data-based Mendelian randomization associations between DNA methylation and smoke/Barrett's esophagus, gene expression and smoke/Barrett's esophagus, and DNA methylation and gene expression, we identified that the genetic variant-cg00935895-RBM43 (ENSG00000184898)-smoke/Barrett's esophagus axis exerted an effect on smoke/Barrett's esophagus by altering the DNA methylation level, which regulated the expression level of RBM43. Conclusions Our study provides evidence of a bidirectional causal association between smoking and Barrett's esophagus from a genetic perspective, which sheds new light on the potential role of RBM43 as a mediator in facilitating the impact of smoking and Barrett's esophagus.
Collapse
Affiliation(s)
- Zhou An
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Meichun Zeng
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xianhua Wang
- Department of Endoscope, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
27
|
Su Z, Luo Z, Wu D, Liu W, Li W, Yin Z, Xue R, Wu L, Cheng Y, Wan Q. Causality between diabetes and membranous nephropathy: Mendelian randomization. Clin Exp Nephrol 2025; 29:227-235. [PMID: 39375304 DOI: 10.1007/s10157-024-02566-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/11/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND Membranous nephropathy (MN) has not yet been fully elucidated regarding its relationship with Type I and II Diabetes. This study aims to evaluate the causal effect of multiple types of diabetes and MN by summarizing the evidence from the Mendelian randomization (MR) study. METHODS The statistical data for MN was obtained from a GWAS study encompassing 7979 individuals. Regarding diabetes, fasting glucose, fasting insulin, and HbA1C data, we accessed the UK-Biobank, within family GWAS consortium, MAGIC, FinnGen database, MRC-IEU, and Neale Lab, which provided sample sizes ranging from 17,724 to 298,957. As a primary method in this MR analysis, we employed the Inverse Variance Weighted (IVW), Weighted Median, Weighted mode, MR-Egger, Mendelian randomization pleiotropy residual sum, and outlier (MR-PRESSO) and Leave-one-out sensitivity test. Reverse MR analysis was utilized to investigate whether MN affects Diabetes. Meta-analysis was applied to combine study-specific estimates. RESULTS It has been determined that type 2 diabetes, gestational diabetes, type 1 diabetes with or without complications, maternal diabetes, and insulin use pose a risk to MN. Based on the genetic prediction, fasting insulin, fasting blood glucose, and HbA1c levels were not associated with the risk of MN. No heterogeneity, horizontal pleiotropy, or reverse causal relationships were found. The meta-analysis results further validated the accuracy. CONCLUSIONS The MR analysis revealed the association between MN and various subtypes of diabetes. This study has provided a deeper understanding of the pathogenic mechanisms connecting MN and diabetes.
Collapse
Affiliation(s)
- Zhihang Su
- Department of Nephrology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 3002 Sungang West Road, Shenzhen, 518000, China
| | - Ziqi Luo
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Di Wu
- Department of Nephrology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 3002 Sungang West Road, Shenzhen, 518000, China
| | - Wen Liu
- Department of Nephrology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 3002 Sungang West Road, Shenzhen, 518000, China
| | - Wangyang Li
- Department of Nephrology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 3002 Sungang West Road, Shenzhen, 518000, China
| | - Zheng Yin
- Department of Nephrology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 3002 Sungang West Road, Shenzhen, 518000, China
| | - Rui Xue
- Department of Nephrology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 3002 Sungang West Road, Shenzhen, 518000, China
| | - Liling Wu
- Department of Nephrology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 3002 Sungang West Road, Shenzhen, 518000, China
| | - Yuan Cheng
- Department of Nephrology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 3002 Sungang West Road, Shenzhen, 518000, China
| | - Qijun Wan
- Department of Nephrology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 3002 Sungang West Road, Shenzhen, 518000, China.
| |
Collapse
|
28
|
Tian W, Song P, Zang J, Zhao J, Liu Y, Wang C, Fang H, Wang H, Zhao Y, Liu X, Gao Y, Cao L. Tanshinone IIA, a component of Salvia miltiorrhiza Bunge, attenuated sepsis-induced liver injury via the SIRT1/Sestrin2/HO-1 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119169. [PMID: 39617088 DOI: 10.1016/j.jep.2024.119169] [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: 07/26/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a traditional Chinese medicine, Salvia miltiorrhiza Bunge has been widely used to treat ischemic and inflammation-related diseases for more than 2000 years. S. miltiorrhiza Bunge has hepatoprotective effects, but the underlying mechanism is not fully understood. OBJECTIVE To verify the effect of tanshinone IIA (Tan IIA), the main fat-soluble component of S. miltiorrhiza Bunge, on liver damage induced by sepsis/LPS-induced inflammation and further explore the underlying mechanisms. MATERIALS AND METHODS Mice were administered Tan IIA 2 h before cecal ligation and puncture (CLP). Liver damage was evaluated by hematoxylin-eosin staining and changes in related serum factor levels. The expression of silent information regulator sirtuin 1 (SIRT1), Sestrin2, HO-1 and inflammatory cytokines was examined by immunohistochemistry or western blotting. LPS was used to induce the inflammatory response in vitro, and the activity of the related signaling pathway in response to Tan IIA was detected by western blotting. The SIRT1 inhibitor EX-527 and small interfering RNAs (siRNAs) were employed to determine the key roles of SIRT1 and Sestrin2 in Tan IIA's function. RESULTS We found that Tan IIA significantly improved the pathological changes and function of the liver, and alleviated liver damage in CLP mice. Additionally, SIRT1, Sestrin2, and HO-1 expression was significantly elevated after Tan IIA treatment compared with that in the CLP group both in vivo and in vitro, and Tan IIA treatment additionally suppressed pro-inflammatory cytokine release. However, inhibition of either SIRT1 or Sestrin2 remarkably abrogated the protective effects of Tan IIA. Most importantly, Sestrin2 appeared to function downstream of SIRT1 based on their expression changes after EX-527 or siRNA treatment. CONCLUSION Tan IIA inhibited sepsis/LPS-induced inflammation through the SIRT1/Sestrin2/HO-1 pathway, thereby protecting against sepsis-induced liver injury (SLI). This study suggests that Tan IIA has therapeutic potential against SLI and that the SIRT1/Sestrin2/HO-1 signaling pathway might be a viable target for SLI treatment.
Collapse
Affiliation(s)
- Wencong Tian
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, 300122, PR China.
| | - Peng Song
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, 300122, PR China.
| | - Junhao Zang
- School of Medicine, Nankai University Tianjin, 300071, PR China.
| | - Jia Zhao
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, 300122, PR China.
| | - Yanhong Liu
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, 300122, PR China.
| | - Chuntao Wang
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, 300122, PR China.
| | - Hong Fang
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, 300122, PR China.
| | - Hongzhi Wang
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, 300122, PR China.
| | - Yongjie Zhao
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, 300122, PR China; Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center Tianjin, 300122, PR China.
| | - Xingqiang Liu
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, 300122, PR China.
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, 300350, PR China.
| | - Lei Cao
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, 300122, PR China; Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center Tianjin, 300122, PR China.
| |
Collapse
|
29
|
Gao Y, Wang D, Wang Q, Wang J, Li S, Wang T, Hu X, Wan C. Causal Impacts of Psychiatric Disorders on Cognition and the Mediating Effect of Oxidative Stress: A Mendelian Randomization Study. Antioxidants (Basel) 2025; 14:162. [PMID: 40002349 PMCID: PMC11852177 DOI: 10.3390/antiox14020162] [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: 12/31/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Many psychiatric disorders are associated with major cognitive deficits. However, it is uncertain whether these deficits develop as a result of psychiatric disorders and what shared risk factors might mediate this relationship. Here, we utilized the Mendelian randomization (MR) analysis to investigate the complex causal relationship between nine major psychiatric disorders and three cognitive phenotypes, while also examining the potential mediating role of oxidative stress as a shared biological underpinning. Schizophrenia (SZ), major depressive disorder (MDD), and attention deficit hyperactivity disorder (ADHD) showed a decreasing effect on cognitive performance, intelligence, and education, while bipolar disorder (BPD) increased educational attainment. MR-Clust results exhibit the shared genetic basis between SZ and other psychiatric disorders in relation to cognitive function. Furthermore, when oxidative stress was considered as a potential mediating factor, the associations between SZ and the three dimensions of cognition, as well as between MDD and intelligence and ADHD and intelligence, exhibited larger effect sizes than the overall. Mediation MR analysis also supported the causal effects between psychiatric disorders and cognition via oxidative stress traits, including carotene, vitamin E, bilirubin, and uric acid. Finally, summary-based MR identified 29 potential causal associations of oxidative stress genes with both cognitive performance and psychiatric disorders. Our findings highlight the importance of considering oxidative stress in understanding and potentially treating cognitive impairments associated with psychiatric conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaowen Hu
- Bio-X Institutes, Key Laboratory for The Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai 200030, China; (Y.G.); (D.W.); (Q.W.); (J.W.); (S.L.); (T.W.)
| | - Chunling Wan
- Bio-X Institutes, Key Laboratory for The Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai 200030, China; (Y.G.); (D.W.); (Q.W.); (J.W.); (S.L.); (T.W.)
| |
Collapse
|
30
|
Krstulović J, Augustin G, Romić I, Tavra A, Batinović F, Hrgović Z. Hyperbaric Oxygen Therapy in the Treatment of Crohn's Disease. Healthcare (Basel) 2025; 13:128. [PMID: 39857155 PMCID: PMC11765433 DOI: 10.3390/healthcare13020128] [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: 12/09/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Our study describes hyperbaric oxygen therapy (HBOT) as an additional therapy in the conservative treatment of Crohn's disease (CD) and its benefit in the early postoperative period to prevent surgical complications and improve gastrointestinal motility. Methods: This retrospective study evaluated HBOT in patients hospitalized at the Clinical Hospital Center Split for complications of CD between 2015 and 2020. Patients (N = 61) aged 19 to 67 with perianal fistulas, abscesses, fistulas, obstruction, stenosis, or bleeding were included, excluding those with ulcerative colitis or requiring intensive care. Patients were retrospectively divided into conservatively and surgically treated groups, and HBOT was administered over 15-25 days, with treatment lasting 60 min at 2.2 absolute atmospheres (ATA). We analyzed treatment outcomes between the HBOT-treated surgical and conservative groups and compared patients treated with HBOT to a cohort from the preceding five years who did not receive HBOT. Results: We treated 61 CD patients with HBOT, including 34 conservatively and 27 surgically treated patients. HBOT significantly reduced disease activity indices (311.7 ± 59.1 vs. 114 ± 29.8; 203.6 ± 24.1 vs. 83.8 ± 15, for conservatively treated patients, and 352.8 ± 45.7 vs. 109 ± 22.8; 270.4 ± 19.7 vs. 140.3 ± 10.6 for surgically treated patients) and accelerated bowel peristalsis recovery, with 94.1% of conservatively treated patients achieving remission. Comparison with a historical cohort showed faster recovery and improved outcomes in the HBOT group. Conclusions: HBOT is useful in postponing or avoiding surgical treatment, and in operated patients, it improves postoperative recovery and reduces the rate of postoperative complications.
Collapse
Affiliation(s)
- Jure Krstulović
- School of Medicine, University of Split, Šoltanska 2, 21000 Split, Croatia; (J.K.); (A.T.); (F.B.); (Z.H.)
- Department of Surgery, University Hospital of Split, Spinčićeva 1, 21000 Split, Croatia
| | - Goran Augustin
- Department of Surgery, University Hospital Centre Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia;
| | - Ivan Romić
- Department of Surgery, University Hospital Centre Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia;
| | - Ante Tavra
- School of Medicine, University of Split, Šoltanska 2, 21000 Split, Croatia; (J.K.); (A.T.); (F.B.); (Z.H.)
| | - Franko Batinović
- School of Medicine, University of Split, Šoltanska 2, 21000 Split, Croatia; (J.K.); (A.T.); (F.B.); (Z.H.)
- Department of Otorhinolaryngology, University Hospital of Split, Spinčićeva 1, 21000 Split, Croatia
| | - Zrinka Hrgović
- School of Medicine, University of Split, Šoltanska 2, 21000 Split, Croatia; (J.K.); (A.T.); (F.B.); (Z.H.)
| |
Collapse
|
31
|
Xia B, Lu YL, Peng J, Liang JW, Li FQ, Ding JY, Wan CW, Le CY, Dai JL, Jie-Wang, Guo B, Huang J. Galactin-8 DNA methylation mediates macrophage autophagy through the MAPK/mTOR pathway to alleviate atherosclerosis. Sci Rep 2025; 15:603. [PMID: 39747459 PMCID: PMC11695939 DOI: 10.1038/s41598-024-85036-1] [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: 08/05/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025] Open
Abstract
DNA methylation modifications are an important mechanism affecting the process of atherosclerosis (AS). Previous studies have shown that Galectin-8 (GAL8) DNA methylation level is associated with sudden death of coronary heart disease or acute events of coronary heart disease. However, the mechanism of GAL8 DNA methylation and gene expression in AS has not been elucidated, prompting us to carry out further research on it. ApoE-/- mice were used to establish an atherosclerosis model, and DNA methylation inhibitor DO05 and MAPK/mTOR inhibitor UO126 were used for intervention. Pyrosequencing was used to detect changes in GAL8 DNA methylation levels of the mouse aorta between groups. ROC curve analysis was performed to assess the relationship between GAL8 DNA methylation and atherosclerosis. Aortic staining with hematoxylin and eosin (H&E) was used to observe the aortic intima, plaque area, and characteristics of secondary lesions within the plaque. Oil Red O staining was used to detect lipid deposition in mouse arterial plaques or macrophages. Movat staining was used to detect the number of foam cells in the plaque. Immunohistochemistry (IHC) and Western blot were used to quantify the localization and expression levels of DNA methyltransferase1 (DNMT1), GAL8, MAPK/mTOR pathway proteins, Light Chain3 (LC3), Beclin1, Sequestosome1 (p62), Tumor Necrosis Factor-α (TNF-α), and other proteins. Immunofluorescence (IF) was used to detect the fluorescence intensity of GAL8, LC3, Monocyte chemoattractant protein-1(MCP-1), and other proteins. Detection of autophagosomes in macrophages by transmission electron microscopy was also performed. The foam cell model was induced with human monocytes (THP-1) and co-cultured with foam cells using siRNAs targeting GAL8, DO05, and UO126. The level of DNMT1 was detected by Western blot; Oil red O staining was used to detect lipid deposition in foam cells in each group, and the localization and expression levels of GAL8, MAPK/mTOR pathway proteins, LC3, Beclin1, p62, and TNF-α were quantitatively determined by Western blot. Immunofluorescence (IF) was used to detect the fluorescence intensity of GAL8, MAPK/mTOR pathway protein, LC3, p62, TNF-α, and other proteins. The GAL-8 promoter region harbors six CpG sites susceptible to DNA methylation. Following DNMT1 inhibition, the DC05 group displayed a significant decrease in methylation across all six CpG sites compared to the C57 and AS groups. Conversely, the UO126 group exhibited increased methylation at the first three CpG loci relative to the AS group. ROC curve analysis revealed GAL8 DNA methylation as an independent risk factor for atherosclerosis: GAL8, along with inflammation-related proteins MCP-1, MMP9, and TNF-α, were upregulated in the mouse lesion group, while expression of autophagy-related proteins LC3 and Beclin1 was downregulated. Additionally, phosphorylated MAPK/mTOR pathway proteins were detected in the mouse model of atherosclerosis. After inhibiting the methylation level of GAL-8 DNA, the expression of GAL-8 was up-regulated, macrophage autophagy was inhibited, inflammation was increased, and atherosclerotic lesions in mice were aggravated. After direct inhibition of the activity of the MAPK/mTOR pathway, macrophage autophagy was further weakened, the inflammatory response was further aggravated, and the atherosclerotic lesions of mice were further aggravated. After the specific knockdown of GAL-8 using siRNA GAL-8 using foam cells, the above phenomenon was reversed, macrophage autophagy was promoted, the inflammatory response was reduced, and the degree of atherosclerosis was alleviated. The degree of GAL8 DNA methylation is related to the progression of atherosclerosis, and its hypomethylation can aggravate atherosclerotic lesions. The mechanism may be through the regulation of MAPK/mTOR pathway to slow down the autophagy of macrophages, and then aggravate the inflammation in plaques. Targeting GAL8 DNA methylation may be a new target for the diagnosis and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Bing Xia
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Yan-Lin Lu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Jin Peng
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, China
- School of Basic Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Jing-Wei Liang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Fang-Qin Li
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Jiu-Yang Ding
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Chang-Wu Wan
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Cui-Yun Le
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Jia-Lin Dai
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Jie-Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Bing Guo
- School of Basic Medicine, Guizhou Medical University, Guiyang, 550004, China.
| | - Jiang Huang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
32
|
Huang Y, Cong A, Li J, Zhou Z, Zhou H, Su C, Hu Z, Hou FF, Cao W. Glycolysis in Peritubular Endothelial Cells and Microvascular Rarefaction in CKD. J Am Soc Nephrol 2025; 36:19-33. [PMID: 39226371 PMCID: PMC11706556 DOI: 10.1681/asn.0000000000000488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024] Open
Abstract
Key Points Peritubular endothelial cells have a hypoglycolytic metabolism in CKD. Restoration of glycolysis in CKD peritubular endothelial cells by overexpressing 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase attenuates microvascular rarefaction and kidney fibrosis. Strategies targeting the metabolic defect in glycolysis in peritubular endothelial cells may be effective in the treatment of CKD. Background Peritubular endothelial cell dropout leading to microvascular rarefaction is a common manifestation of CKD. The role of metabolism reprogramming in peritubular endothelial cell loss in CKD is undetermined. Methods Single-cell sequencing and metabolic analysis were used to characterize the metabolic profile of peritubular endothelial cells from patients with CKD and from CKD mouse models. In vivo and in vitro models demonstrated metabolic reprogramming in peritubular endothelial cells in conditions of CKD and its contribution to microvascular rarefaction. Results In this study, we identified glycolysis as a top dysregulated metabolic pathway in peritubular endothelial cells from patients with CKD. Specifically, CKD peritubular endothelial cells were hypoglycolytic while displaying an antiangiogenic response with decreased proliferation and increased apoptosis. The hypoglycolytic phenotype of peritubular endothelial cells was recapitulated in CKD mouse models and in peritubular endothelial cells stimulated by hydrogen peroxide. Mechanically, oxidative stress, through activating a redox sensor kruppel-like transcription factor 9, downregulated the glycolytic activator 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase expression, thereby reprogramming peritubular endothelial cells toward a hypoglycolytic phenotype. 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase overexpression in peritubular endothelial cells restored hydrogen peroxide–induced reduction in glycolysis and cellular ATP levels and enhanced the G1/S cell cycle transition, enabling peritubular endothelial cells to improve proliferation and reduce apoptosis. Consistently, restoration of peritubular endothelial cell glycolysis in CKD mice, by overexpressing endothelial Pfkfb3, reversed the antiangiogenic response in peritubular endothelial cells and protected the kidney from microvascular rarefaction and fibrosis. By contrast, suppression of glycolysis by endothelial Pfkfb3 deletion exacerbated microvascular rarefaction and fibrosis in CKD mice. Conclusions Our study revealed a disrupted regulation of glycolysis in peritubular endothelial cells as an initiator of microvascular rarefaction in CKD. Restoration of peritubular endothelial cell glycolysis in CKD kidney improved microvascular rarefaction and ameliorated fibrotic lesions.
Collapse
Affiliation(s)
- Yujie Huang
- Division of Nephrology, Nanfang Hospital, Southern Medical University; State Key Laboratory of Organ Failure Research; National Clinical Research Center for Kidney Disease; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Yin Z, Fu L, Wang Y, Tai S. Impact of gut microbiota on cardiac aging. Arch Gerontol Geriatr 2025; 128:105639. [PMID: 39312851 DOI: 10.1016/j.archger.2024.105639] [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: 08/02/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024]
Abstract
Recent research has suggested imbalances in gut microbiota composition as contributors to cardiac aging. An individual's physical condition, along with lifestyle-associated factors, including diet and medication, are significant determinants of gut microbiota composition. This review discusses evidence of bidirectional associations between aging and gut microbiota, identifying gut microbiota-derived metabolites as potential regulators of cardiac aging. It summarizes the effects of gut microbiota on cardiac aging diseases, including cardiac hypertrophy and fibrosis, heart failure, and atrial fibrillation. Furthermore, this review discusses the potential anti-aging effects of modifying gut microbiota composition through dietary and pharmacological interventions. Lastly, it underscores critical knowledge gaps and outlines future research directions. Given the current limited understanding of the direct relationship between gut microbiota and cardiac aging, there is an urgent need for preclinical and clinical investigations into the mechanistic interactions between gut microbiota and cardiac aging. Such endeavors hold promise for shedding light on the pathophysiology of cardiac aging and uncovering new therapeutic targets for cardiac aging diseases.
Collapse
Affiliation(s)
- Zhiyi Yin
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China
| | - Liyao Fu
- Hunan Key Laboratory of Cardiometabolic Medicine, Department of Cardiology, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China
| | - Yongjun Wang
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China.
| | - Shi Tai
- Hunan Key Laboratory of Cardiometabolic Medicine, Department of Cardiology, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China.
| |
Collapse
|
34
|
Lu Y, Su Y, Wang N, Li D, Zhang H, Xu H. Identification of O-glycosylation related genes and subtypes in ulcerative colitis based on machine learning. PLoS One 2024; 19:e0311495. [PMID: 39739658 DOI: 10.1371/journal.pone.0311495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/17/2024] [Indexed: 01/02/2025] Open
Abstract
Ulcerative colitis (UC) is an immune-related inflammatory bowel disease, with its underlying mechanisms being a central area of clinical research. O-GlcNAcylation plays a critical role in regulating immunity progression and the occurrence of inflammatory diseases and tumors. Yet, the mechanism of O-GlcNAc-associated colitis remains to be elucidated. To this end, the transcriptional and clinical data of GSE75214 and GSE92415 from the GEO database was hereby examined, and genes MUC1, ADAMTS1, GXYLT2, and SEMA5A were found to be significantly related to O-GlcNAcylation using machine learning methods. Based on the four hub genes, two UC subtypes were built. Notably, subtype B might be prone to developing colitis-associated colorectal cancer (CAC). This study delved into the role of intestinal glycosylation changes, especially the O-GlcNAcylation, and forged a foundation for further research on the occurrence and development of UC. Overall, understanding the role of O-GlcNAcylation in UC could have significant implications for diagnosis and treatment, offering valuable insights into the disease's progression.
Collapse
Affiliation(s)
- Yue Lu
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yi Su
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Nan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Dongyue Li
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Huichao Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyu Xu
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
35
|
Qi C, Li A, Su F, Wang Y, Zhou L, Tang C, Feng R, Mao R, Chen M, Chen L, Koppelman GH, Bourgonje AR, Zhou H, Hu S. An atlas of the shared genetic architecture between atopic and gastrointestinal diseases. Commun Biol 2024; 7:1696. [PMID: 39719505 DOI: 10.1038/s42003-024-07416-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024] Open
Abstract
Comorbidity among atopic diseases (ADs) and gastrointestinal diseases (GIDs) has been repeatedly demonstrated by epidemiological studies, whereas the shared genetic liability remains largely unknown. Here we establish an atlas of the shared genetic architecture between 10 ADs or related traits and 11 GIDs, comprehensively investigating the comorbidity-associated genomic regions, cell types, genes and genetically predicted causality. Although distinct genetic correlations between AD-GID are observed, including 14 genome-wide and 28 regional correlations, genetic factors of Crohn's disease (CD), ulcerative colitis (UC), celiac disease and asthma subtypes are converged on CD4+ T cells consistently across relevant tissues. Fourteen genes are associated with comorbidities, with three genes are known treatment targets, showing probabilities for drug repurposing. Lower expressions of WDR18 and GPX4 in PBMC CD4+ T cells predict decreased risk of CD and asthma, which could be novel drug targets. MR unveils certain ADs led to higher risk of GIDs or vice versa. Taken together, here we show distinct genetic correlations between AD-GID pairs, but the correlated genomic loci converge on the dysregulation of CD4+ T cells. Inhibiting WDR18 and GPX4 expressions might be candidate therapeutic strategies for CD and asthma. Estimated causality indicates potential guidance for preventing comorbidity.
Collapse
Affiliation(s)
- Cancan Qi
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - An Li
- Department of Periodontology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Fengyuan Su
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yu Wang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Longyuan Zhou
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ce Tang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Rui Feng
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Gastroenterology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-Sen University, Nanning, Guangxi, China
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Lianmin Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Gerard H Koppelman
- University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Centre Groningen, Beatrix Children's Hospital, Department of Paediatric Pulmonology and Paediatric Allergology, Groningen, the Netherlands
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Hongwei Zhou
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Shixian Hu
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
36
|
Bian R, Li D, Xu X, Zhang L. The impact of immunity on the risk of coronary artery disease: insights from a multiomics study. Postgrad Med J 2024; 101:50-59. [PMID: 39180487 DOI: 10.1093/postmj/qgae105] [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: 06/17/2024] [Revised: 07/16/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Immune inflammation is intricately associated with coronary artery disease (CAD) progression, necessitating the pursuit of more efficacious therapeutic strategies. This study aimed to uncover potential therapeutic targets for CAD and myocardial infarction (MI) by elucidating the causal connection between regulatory immune-related genes (RIRGs) and these disorders. METHODOLOGY We performed summary data-based Mendelian randomization analysis to assess the therapeutic targets linked to expression quantitative trait loci and methylation quantitative trait loci of RIRGs in relation to CAD and MI. Independent validation cohorts and datasets from coronary artery and left ventricular heart tissue were analyzed. To strengthen causal inference, colocalization analysis and PhenoScanner phenotype scans were employed. RESULTS Utilizing multiomics integration, we pinpointed EIF2B2, FCHO1, and DDT as CAD risk genes. Notably, EIF2B2 and FCHO1 displayed significant associations with MI. High EIF2B2 expression, regulated by cg16144293, heightened CAD and MI risk at rs175438. In contrast, enhanced FCHO1 expression, modulated by cg18329931, reduced CAD and MI risk at rs13382133. DDT upregulation influenced by cg11060661 and cg09664220 was associated with decreased CAD risk at rs5760120. Colocalization analysis firmly established these relationships. CONCLUSION EIF2B2, FCHO1, and DDT represent risk loci for CAD progression within RIRGs. Our identification of these genes enhances understanding of CAD pathogenesis and directs future drug development efforts.
Collapse
Affiliation(s)
- Rutao Bian
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan 450000, China
- The Affiliated Zhengzhou Hospital of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China
| | - Dongyu Li
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan 450000, China
- The Affiliated Zhengzhou Hospital of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China
| | - Xuegong Xu
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan 450000, China
- The Affiliated Zhengzhou Hospital of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China
| | - Li Zhang
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan 450000, China
- The Affiliated Zhengzhou Hospital of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China
| |
Collapse
|
37
|
Wang J, Hu J, Qin D, Han D, Hu J. A multi-omics Mendelian randomization identifies putatively causal genes and DNA methylation sites for asthma. World Allergy Organ J 2024; 17:101008. [PMID: 39720783 PMCID: PMC11667005 DOI: 10.1016/j.waojou.2024.101008] [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: 01/25/2024] [Revised: 08/02/2024] [Accepted: 11/12/2024] [Indexed: 12/26/2024] Open
Abstract
Background Asthma is a global chronic respiratory disease with complex pathogenesis. While current therapies offer some relief, they often fall short in effectively managing symptoms and preventing exacerbations for numerous patients. Thus, understanding its mechanisms and discovering new drug targets remains a pressing need for better treatment. Methods Using the GEO dataset, we screened differentially expressed genes (DEGs) in asthma patients' blood. Employing Summary Data-based Mendelian Randomization (SMR) and Two-Sample Mendelian Randomization (TSMR), we pinpointed asthma causal genes, causal DNA methylation sites, and methylation sites affecting gene expression, cross validated with at least 2 large-scale GWAS from each source. We utilized colocalization for genetic associations, meta-analysis for data integration, two-step MR for methylation-gene-asthma mediation mechanism. Druggability was evaluated using Open Target, virtual screening, and docking. Results Among the 954 DEGs found in asthma patients' blood, increased expression of CEP95 (discovery, OR_SMR = 0.94, 95% CI: 0.91-0.97), RBM6 (discovery, OR_SMR = 0.97, 95% CI: 0.95-0.99), and ITPKB (discovery, OR_SMR = 0.82, 95% CI: 0.74-0.92) in the blood decreased the risk of asthma, higher levels of HOXB-AS1 (discovery, OR_SMR = 1.05, 95% CI: 1.03-1.07), ETS1 (discovery, OR_SMR = 1.62, 95% CI: 1.29-2.04), and JAK2 (discovery, OR_SMR = 1.13, 95% CI: 1.06-1.21) in the blood increased the risk of asthma. Additionally, a total of 8 methylation sites on ITPKB, ETS1, and JAK2 were identified to influence asthma. An increase in methylation at site cg16265553 raised the risk of asthma partially by suppressing ITPKB expression. Similarly, increased methylation at cg13661497 reduced the asthma risk totally by suppressing JAK2 expression. The impact of CEP95, HOXB-AS1, and RBM6 expressions on asthma was further confirmed in lung tissues. Except for HOXB-AS1, all the other genes were potential druggable targets. Conclusion Our study highlighted that specific gene expressions and methylation sites significantly influence asthma risk and revealed a potential methylation-to-gene-to-asthma mechanism. This provided pivotal evidence for future targeted functional studies and the development of preventive and treatment strategies.
Collapse
Affiliation(s)
- Jia Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jinxin Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dan Qin
- Research Center of Traditional Chinese Medicine Information Engineering, Beijing University of Chinese Medicine, Beijing, China
| | - Dan Han
- Department of Neonatology, The First Hospital of China Medical University, Shenyang, China
| | - Jiapeng Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
38
|
Chen C, Quan J, Chen X, Yang T, Yu C, Ye S, Yang Y, Wu X, Jiang D, Weng Y. Explore key genes of Crohn's disease based on glycerophospholipid metabolism: A comprehensive analysis Utilizing Mendelian Randomization, Multi-Omics integration, Machine Learning, and SHAP methodology. Int Immunopharmacol 2024; 141:112905. [PMID: 39173401 DOI: 10.1016/j.intimp.2024.112905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND AND AIMS Crohn's disease (CD) is a chronic, complex inflammatory condition with increasing incidence and prevalence worldwide. However, the causes of CD remain incompletely understood. We identified CD-related metabolites, inflammatory factors, and key genes by Mendelian randomization (MR), multi-omics integration, machine learning (ML), and SHAP. METHODS We first performed a mediation MR analysis on 1400 serum metabolites, 91 inflammatory factors, and CD. We found that certain phospholipids are causally related to CD. In the scRNA-seq data, monocytes were categorized into high and low metabolism groups based on their glycerophospholipid metabolism scores. The differentially expressed genes of these two groups of cells were extracted, and transcription factor prediction, cell communication analysis, and GSEA analysis were performed. After further screening of differentially expressed genes (FDR<0.05, log2FC>1), least absolute shrinkage and selection operator (LASSO) regression was performed to obtain hub genes. Models for hub genes were built using the Catboost, XGboost, and NGboost methods. Further, we used the SHAP method to interpret the models and obtain the gene with the highest contribution to each model. Finally, qRT-PCR was used to verify the expression of these genes in the peripheral blood mononuclear cells (PBMC) of CD patients and healthy subjects. RESULT MR results showed 1-palmitoyl-2-stearoyl-gpc (16:0/18:0) levels, 1-stearoyl-2-arachidonoyl-GPI (18:0/20:4) levels, 1-arachidonoyl-gpc (20:4n6) levels, 1-palmitoyl-2-arachidonoyl-gpc (16:0/20:4n6) levels, and 1-arachidonoyl-GPE (20:4n6) levels were significantly associated with CD risk reduction (FDR<0.05), with CXCL9 acting as a mediation between these phospholipids and CD. The analysis identified 19 hub genes, with Catboost, XGboost, and NGboost achieving AUC of 0.91, 0.88, and 0.85, respectively. The SHAP methodology obtained the three genes with the highest model contribution: G0S2, S100A8, and PLAUR. The qRT-PCR results showed that the expression levels of S100A8 (p = 0.0003), G0S2 (p < 0.0001), and PLAUR (p = 0.0141) in the PBMC of CD patients were higher than healthy subjects. CONCLUSION MR findings suggest that certain phospholipids may lower CD risk. G0S2, S100A8, and PLAUR may be potential pathogenic genes in CD. These phospholipids and genes could serve as novel diagnostic and therapeutic targets for CD.
Collapse
Affiliation(s)
- Changan Chen
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Juanhua Quan
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Xintian Chen
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Tingmei Yang
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Caiyuan Yu
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Shicai Ye
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Yuping Yang
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Xiu Wu
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Danxian Jiang
- Department of Medical Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China.
| | - Yijie Weng
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China.
| |
Collapse
|
39
|
Dong XX, Chen DL, Chen HM, Li DL, Hu DN, Lanca C, Grzybowski A, Pan CW. DNA methylation biomarkers and myopia: a multi-omics study integrating GWAS, mQTL and eQTL data. Clin Epigenetics 2024; 16:157. [PMID: 39538342 PMCID: PMC11562087 DOI: 10.1186/s13148-024-01772-1] [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: 08/23/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND This study aimed to identify DNA methylation biomarkers associated with myopia using summary-data-based Mendelian randomization (SMR). METHODS A systematic search of the PubMed, Web of Science, Cochrane Library, and Embase databases was conducted up to March 27, 2024. SMR analyses were performed to integrate genome-wide association study (GWAS) with methylation quantitative trait loci (mQTL) and expression quantitative trait loci (eQTL) studies. The heterogeneity in the dependent instrument (HEIDI) test was utilized to distinguish pleiotropic associations from linkage disequilibrium. RESULTS The systematic review identified 26 DNA methylation biomarkers in five studies, with no overlap observed among those identified by different studies. After integrating GWAS with multi-omics data of mQTL and eQTL, six genes were significantly associated with myopia: PRMT6 (cg00944433 and cg15468180), SH3YL1 (cg03299269, cg11361895, and cg13354988), ZKSCAN4 (cg01192291), GATS (cg17830204), NPAT (cg04826772), and UBE2I (cg03545757 and cg08025960). CONCLUSIONS We identified six methylation biomarkers associated with the risk of myopia that may be helpful to elucidate the etiology mechanisms of myopia. Further experimental validation studies are required to corroborate these findings.
Collapse
Affiliation(s)
- Xing-Xuan Dong
- School of Public Health, The Fourth Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, China
| | - Dong-Ling Chen
- School of Public Health, The Fourth Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, China
| | - Hui-Min Chen
- School of Public Health, The Fourth Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, China
| | - Dan-Lin Li
- School of Public Health, The Fourth Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, China
| | - Dan-Ning Hu
- New York Eye and Ear Infirmary of Mount Sinai, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Carla Lanca
- Division of Science, New York University Abu Dhabi, Abu Dhabi, UAE
- Comprehensive Health Research Center (CHRC), Escola Nacional de Saúde Pública, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Andrzej Grzybowski
- Institute for Research in Ophthalmology, Foundation for Ophthalmology Development, Poznan, Poland
| | - Chen-Wei Pan
- School of Public Health, The Fourth Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
40
|
Xie J, Ma R, Xu X, Yang M, Yu H, Wan X, Xu K, Guo J, Xu P. Identification of genetic association between mitochondrial dysfunction and knee osteoarthritis through integrating multi-omics: a summary data-based Mendelian randomization study. Clin Rheumatol 2024; 43:3487-3496. [PMID: 39259428 PMCID: PMC11489259 DOI: 10.1007/s10067-024-07136-7] [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: 02/24/2024] [Revised: 08/04/2024] [Accepted: 09/01/2024] [Indexed: 09/13/2024]
Abstract
OBJECTIVE Association between mitochondrial dysfunction and osteoarthritis (OA) has been consistently investigated, yet their genetic association remains obscure. In this study, mitochondrial-related genes were used as instrumental variables to proxy for mitochondrial dysfunction, and summary data of knee OA (KOA) were used as outcome to examine their genetic association. METHODS We obtained 1136 mitochondrial-related genes from the human MitoCarta3.0 database. Genetic proxy instruments for mitochondrial-related genes from studies of corresponding gene expression (n = 31,684) and protein (n = 35,559) quantitative trait locus (eQTLs and pQTLs), respectively. Aggregated data for KOA (62,497 KOA cases and 333,557 controls) were extracted from the largest OA genome-wide association study (GWAS). We integrated QTL data with KOA GWAS data to estimate their genetic association using summary data-based Mendelian randomization analysis (SMR). Additionally, we implemented Bayesian colocalization analysis to reveal whether suggestive mitochondrial-related genes and KOA were driven by a same genetic variant. Finally, to validate the primary findings, replication study (24,955 cases and 378,169 controls) and multi-SNP-based SMR (SMR-multi) test was performed. RESULTS Through SMR analysis, we found that the expression levels of 2 mitochondrial-related genes were associated with KOA risk. Specifically, elevated gene expression levels of the IMMP2L (odds ratio [OR] = 1.056; 95% confidence interval [CI] = 1.030-1.082; P-FDR = 0.004) increased the risk of KOA. Conversely, increased gene expression levels of AKAP10 decreased the risk of KOA (OR = 0.955; 95% CI, 0.934-0.977; P-FDR = 0.019). Colocalization analysis demonstrated that AKAP10 (PP.H4 = 0.84) and IMMP2L (PP.H4 = 0.91) shared the same genetic variant with KOA. In addition, consistent results were found in replication study and SMR-multi test, further demonstrating the reliability of our findings. CONCLUSIONS In summary, our analyses revealed the genetic association between mitochondrial dysfunction proxied by mitochondrial-related genes and KOA, providing new insight into potential pathogenesis of KOA. Furthermore, these identified candidate genes offer the possibility of clinical drug target development for KOA. Key points • This is the first SMR study to explore the genetic association between mitochondrial dysfunction proxied by mitochondrial-related genes and KOA. • Sufficient evidence to support genetic association between the expression levels of AKAP10 and IMMP2L, and KOA • Our MR analysis may provide novel new insight into potential pathogenesis of KOA. • These identified candidate genes offer the possibility of clinical drug target development for KOA.
Collapse
Affiliation(s)
- Jiale Xie
- Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xian, Shaanxi, China
| | - Rui Ma
- Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xian, Shaanxi, China
| | - Xin Xu
- Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xian, Shaanxi, China
| | - Mingyi Yang
- Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xian, Shaanxi, China
| | - Hui Yu
- Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xian, Shaanxi, China
| | - Xianjie Wan
- Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xian, Shaanxi, China
| | - Ke Xu
- Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xian, Shaanxi, China
| | - Junfei Guo
- Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xian, Shaanxi, China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xian, Shaanxi, China.
| |
Collapse
|
41
|
Jeong JS, Baek GH, Kim JW, Kim JH, Chung EH, Ko JW, Kwon MJ, Kim SK, Lee SH, Kim JS, Kim TW. Korean Red Ginseng alleviates dextran sodium sulfate-induced colitis through gut microbiota modulation in mice. J Ginseng Res 2024; 48:581-591. [PMID: 39583173 PMCID: PMC11584195 DOI: 10.1016/j.jgr.2024.08.001] [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: 03/12/2024] [Revised: 07/25/2024] [Accepted: 08/08/2024] [Indexed: 11/26/2024] Open
Abstract
Background There is a growing interest in understanding the association between the gut microbiota and inflammatory bowel disease (IBD). Natural compounds, such as Korean Red Ginseng (KRG), show promise for IBD treatment because of their ability to influence gut microbiota. This study explored the effects of KRG on gut microbiota modulation and subsequent intestinal epithelial cell regeneration in an experimental colitis model. Method Using a mouse model of colitis induced by 2 % dextran sodium sulfate, the study administered 200 or 400 mg/kg/day of KRG to evaluate its biological effects. Colitis symptoms were assessed through body weight, disease activity index, colon length, and histological analysis. The microbial composition in the fecal was determined using 16S rRNA sequencing. To evaluate regeneration signals in the colon, western blotting and immunohistochemistry assays were conducted. Result Administration of KRG effectively mitigated colitis symptoms in mice, as indicated by histological examination showing alleviated epithelial damage and inflammation, along with increased mucus production. Microbiota analysis showed that KRG significantly altered microbial diversity, favoring beneficial taxa and suppressing harmful taxa. Moreover, ameliorated β-catenin/transcription factor-4 protein expression, a key signal associated with epithelial cell regeneration, was observed in the KRG treated groups, accompanied by improved intestinal linings. Conclusion These findings suggest that KRG exerts biological effects in colitis by modulating gut microbiota and creating a favorable intestinal environment, thereby reducing regenerative signals. Further research is warranted to elucidate the cellular and molecular mechanisms underlying the interaction of KRG with gut microbiota and pave the way for effective IBD therapies.
Collapse
Affiliation(s)
- Ji-Soo Jeong
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Ga-Hyeon Baek
- Department of Nano-Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Jeong-Won Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Jin-Hwa Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Hye Chung
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Je-Won Ko
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Mi-Jin Kwon
- R&D Headquarters, Korea Ginseng Corporation, Gwacheon, Republic of Korea
| | - Sang-Kyu Kim
- R&D Headquarters, Korea Ginseng Corporation, Gwacheon, Republic of Korea
| | - Seung-Ho Lee
- R&D Headquarters, Korea Ginseng Corporation, Gwacheon, Republic of Korea
| | - Jun-Seob Kim
- Department of Nano-Bioengineering, Incheon National University, Incheon, Republic of Korea
- Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Tae-Won Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
42
|
Shao X, Wang Y, Geng Z, Liang G, Zhu X, Liu L, Meng M, Duan L, Zhu G. Novel therapeutic targets for major depressive disorder related to oxidative stress identified by integrative multi-omics and multi-trait study. Transl Psychiatry 2024; 14:443. [PMID: 39426956 PMCID: PMC11490649 DOI: 10.1038/s41398-024-03126-0] [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: 06/26/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024] Open
Abstract
Oxidative stress (OS) is strongly implicated in the pathophysiology of major depressive disorder (MDD) but the molecular mechanisms remain largely unknown. The purpose of this study is to identify genes related to both OS and MDD, and further to evaluate the utility of these genes as diagnostic markers and potential treatment targets. We searched datasets related to MDD from the Gene Expression Omnibus (GEO) database for differentially expressed genes (DEGs) also related to OS according to GeneCards. Bioinformatics analyses and machine learning algorithms were used to identify hub genes mediating OS-MDD interactions. A summary data-based Mendelian randomization (SMR) approach was employed to identify possible causal genes for MDD from blood tissue eQLT data. These investigations identified 32 genes mediating OS-MDD interactions, while SMR analysis identified KCNE1 (OR = 1.057, 95%CI = 1.013-1.102, P value = 0.010), MAPK3 (OR = 1.023, 95%CI = 1.004-1.043, P value = 0.020), and STIP1 (OR = 0.792, 95%CI = 0.641-0.979, P value = 0.031) as OS-related causal genes for MDD. These genes may thus serve as useful diagnostic markers and potential therapeutic targets.
Collapse
Affiliation(s)
- Xiaojun Shao
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Yuan Wang
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Zhongli Geng
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
- Shenyang Mental Health Center, Shenyang, 110168, China
| | | | - Xiaotong Zhu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
- Department of Psychiatry, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110005, China
| | - Lu Liu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
- Department of Psychiatry, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110005, China
| | - Ming Meng
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
- Shenyang Mental Health Center, Shenyang, 110168, China
| | - Li Duan
- Department of Health Management Research, Chengde Medical University, Chengde, 067000, China
| | - Gang Zhu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
43
|
Wang L, Wang Q, Zhou Y. Oral Microbial Translocation Genes in Gastrointestinal Cancers: Insights from Metagenomic Analysis. Microorganisms 2024; 12:2086. [PMID: 39458395 PMCID: PMC11510655 DOI: 10.3390/microorganisms12102086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Along with affecting oral health, oral microbial communities may also be endogenously translocated to the gut, thereby mediating the development of a range of malignancies in that habitat. While species-level studies have proven the capability of oral pathogens to migrate to the intestine, genetic evidence supporting this mechanism remains insufficient. In this study, we identified over 55,000 oral translocation genes (OTGs) associated with colorectal cancer (CRC) and inflammatory bowel disease (IBD). These genes are primarily involved in signal transduction and cell wall biosynthesis and show consistency in their functions between IBD and CRC. Furthermore, we found that Leclercia adecarboxylata, a newly discovered opportunistic pathogen, has a significantly high abundance in the gut microbiota of colorectal cancer patients. OTGs of this pathogen were enriched in 15 metabolic pathways, including those associated with amino acid and cofactor metabolism. These findings, for the first time, provide evidence at the genetic level of the transfer of oral pathogens to the intestine and offer new insights into the understanding of the roles of oral pathogens in the development of gastrointestinal cancers.
Collapse
Affiliation(s)
| | | | - Yan Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China; (L.W.); (Q.W.)
| |
Collapse
|
44
|
Li X, Li H, Hong T, Li Z, Wang Z. Assessing the causal relationship between obesity and hypothyroidism using Mendelian randomization. J Investig Med 2024; 72:763-775. [PMID: 38785311 DOI: 10.1177/10815589241257214] [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] [Indexed: 05/25/2024]
Abstract
To explore the causal relationship between obesity and hypothyroidism and identify risk factors and the predictive value of subclinical hypothyroidism (SCH) in obese patients using Mendelian randomization, this study employed five Mendelian randomization methods (MR Egger, Weighted Median, Inverse Variance Weighted, Simple Mode, and Weighted Mode) to analyze clinical data from 308 obese patients at the People's Hospital of Xinjiang Uygur Autonomous Region, from January 2015 to June 2023. Patients were divided based on thyroid function tests into normal (n = 173) and SCH groups (n = 56). Comparative analyses, along with univariate and multivariate logistic regression, were conducted to identify risk factors for SCH in obese patients. A significant association between obesity and hypothyroidism was established, especially highlighted by the inverse variance weighted method. SCH patients showed higher ages, thyroid-stimulating hormone levels, and thyroid autoantibody positivity rates, with lower T4 and FT4 levels. Age, FT4, thyroid autoantibodies, TPO-Ab, and Tg-Ab were confirmed as risk factors. The predictive value of FT4 levels for SCH in obesity was significant, with an Area Under the Curve (AUC) of 0.632. The study supports a potential causal link between obesity and hypothyroidism, identifying specific risk factors for SCH in obese patients. FT4 level stands out as an independent predictive factor, suggesting its utility in early diagnosis and preventive strategies for SCH.
Collapse
Affiliation(s)
- Xin Li
- Department of Minimally Invasive, Hernia and Abdominal Surgery, People's Hospital of Xinjiang Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Huilin Li
- Department of Minimally Invasive, Hernia and Abdominal Surgery, People's Hospital of Xinjiang Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Tao Hong
- Graduate School, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Zanlin Li
- Department of Minimally Invasive, Hernia and Abdominal Surgery, People's Hospital of Xinjiang Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Zhi Wang
- Department of Comprehensive Surgery, People's Hospital of Xinjiang Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
45
|
Liu X, Zhang D, Zhao F, Li S, Zhu H, Zhang X. Deciphering the role of oxidative stress genes in idiopathic pulmonary fibrosis: a multi-omics mendelian randomization approach. Genes Immun 2024; 25:389-396. [PMID: 39174688 DOI: 10.1038/s41435-024-00292-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
Oxidative stress (OS) is crucial in idiopathic pulmonary fibrosis (IPF) pathogenesis, with its genes potentially acting as both causes and consequences of the disease. We identified OS-related genes from GeneCards and performed a meta-analysis on pulmonary transcriptome datasets to discover differentially expressed genes (DEGs) related to OS in IPF. We integrated this data with the largest available IPF GWAS summaries, expression quantitative trait loci (eQTLs), and DNA methylation QTLs (mQTLs) from blood. This approach aimed to identify blood OS genes and regulatory elements linked to IPF risk, incorporating the latest pulmonary eQTLs and bronchoalveolar lavage fluid microbial QTLs (bmQTLs) for a comprehensive view of gene-lung microbiota interactions through SMR and colocalization analyses. Sensitivity analyses were conducted using two additional mendelian randomization (MR) methods. Meta-analysis revealed 1090 differentially expressed OS genes between IPF patients and controls. Integration with IPF GWAS, eQTL, and mQTL data identified key genes and regulatory elements involved in IPF pathogenesis, highlighting the role of specific genes such as KCNMA1 and SLC22A5 in modulating IPF risk through epigenetic mechanisms. Colocalization analysis further identified potential interactions between gene expression and lung microbiota. Our findings elucidate the complex interplay between OS genes and IPF, suggesting potential therapeutic targets and highlighting the importance of considering epigenetic and microbial interactions in the disease's etiology and progression.
Collapse
Affiliation(s)
- Xin Liu
- Second Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dengfeng Zhang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fangchao Zhao
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shujun Li
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Haiyong Zhu
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Xu Zhang
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
46
|
Drăgoi CM, Diaconu CC, Nicolae AC, Dumitrescu IB. Redox Homeostasis and Molecular Biomarkers in Precision Therapy for Cardiovascular Diseases. Antioxidants (Basel) 2024; 13:1163. [PMID: 39456418 PMCID: PMC11504313 DOI: 10.3390/antiox13101163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Precision medicine is envisioned as the future of cardiovascular healthcare, offering a more tailored and effective method for managing cardiovascular diseases compared to the traditional one-size-fits-all approaches. The complex role of oxidative stress in chronic diseases within the framework of precision medicine was carefully explored, delving into the cellular redox status and its critical involvement in the pathophysiological complexity of cardiovascular diseases (CVDs). The review outlines the mechanisms of reactive oxygen species generation and the function of antioxidants in maintaining redox balance. It emphasizes the elevated reactive oxygen species concentrations observed in heart failure and their detrimental impact on cardiovascular health. Various sources of ROS within the cardiovascular system are examined, including mitochondrial dysfunction, which contributes to oxidative stress and mitochondrial DNA degradation. The article also addresses oxidative stress's role in myocardial remodeling, a process pivotal to the progression of heart diseases. By integrating these aspects, the review underscores the importance of redox homeostasis and identifies molecular biomarkers that can enhance precision therapy for CVDs. The insights provided aim to pave the way for targeted therapeutic strategies that mitigate oxidative stress, thereby improving patient outcomes in cardiovascular medicine.
Collapse
Affiliation(s)
- Cristina Manuela Drăgoi
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania; (C.M.D.); (I.-B.D.)
| | - Camelia Cristina Diaconu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania;
- Department of Internal Medicine, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Alina Crenguța Nicolae
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania; (C.M.D.); (I.-B.D.)
| | - Ion-Bogdan Dumitrescu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania; (C.M.D.); (I.-B.D.)
| |
Collapse
|
47
|
Chen J, Rao H, Zheng X. Identification of novel targets associated with cholesterol metabolism in nonalcoholic fatty liver disease: a comprehensive study using Mendelian randomization combined with transcriptome analysis. Front Genet 2024; 15:1464865. [PMID: 39359475 PMCID: PMC11445148 DOI: 10.3389/fgene.2024.1464865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024] Open
Abstract
Background There is limited research on cholesterol metabolism-related genes (CM-RGs) in non-alcoholic fatty liver disease (NAFLD), despite hypercholesterolemia being a recognized risk factor. The role of CM-RGs in NAFLD remains unclear. Methods The differentially expressed genes (DEGs) between NAFLD and control were acquired by differential expression analysis. The differentially expressed genes associated with cholesterol metabolism (DE-CM-RGs) were identified and functional enrichment analyses were performed. Protein-protein interaction network analysis and a two-sample Mendelian randomization study were utilized for identifying hub genes. Nomogram model, competing endogenous RNA and messenger RNA-drug networks were established. In addition, immunoinfiltration analysis was performed. Results We identified four hub genes (MVK, HMGCS1, TM7SF2, and FDPS) linked to NAFLD risk. MVK and TM7SF2 were protective factors, HMGCS1 and FDPS were risk factors for NAFLD. The area under the curve values of nomograms in GSE135251 and GSE126848 were 0.79 and 0.848, respectively. The gene set enrichment analysis indicated that hub genes participated in calcium signaling pathways and biosynthesis of unsaturated fatty acids. NAFLD patients showed increased CD56dim NK cells and Th17. Tretinoin, alendronate, zoledronic acid, and quercetin are potential target agents in NAFLD. Conclusion Our study has linked cholesterol metabolism genes (MVK, HMGCS1, TM7SF2, and FDPS) to NAFLD, providing a promising diagnostic framework, identifying treatment targets, and offering novel perspectives into its mechanisms.
Collapse
Affiliation(s)
- Juan Chen
- Department of Gastroenterology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Huajing Rao
- Emergency Internal Medicine, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaoling Zheng
- Department of Endoscopy, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| |
Collapse
|
48
|
Wu K, Zha H, Wu T, Liu H, Peng R, Lin Z, Lv D, Liao X, Sun Y, Zhang Z. Cytosolic Hmgb1 accumulation in mesangial cells aggravates diabetic kidney disease progression via NFκB signaling pathway. Cell Mol Life Sci 2024; 81:408. [PMID: 39287634 PMCID: PMC11408458 DOI: 10.1007/s00018-024-05433-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
Diabetic kidney disease (DKD) is the predominant type of end-stage renal disease. Increasing evidence suggests thatglomerular mesangial cell (MC) inflammation is pivotal for cell proliferation and DKD progression. However, the exactmechanism of MC inflammation remains largely unknown. This study aims to elucidate the role of inflammatoryfactor high-mobility group box 1 (Hmgb1) in DKD. Inflammatory factors related to DKD progression are screened viaRNA sequencing (RNA-seq). In vivo and in vitro experiments, including db/db diabetic mice model, CCK-8 assay, EdUassay, flow cytometric analysis, Co-IP, FISH, qRT-PCR, western blot, single cell nuclear RNA sequencing (snRNA-seq),are performed to investigate the effects of Hmgb1 on the inflammatory behavior of MCs in DKD. Here, wedemonstrate that Hmgb1 is significantly upregulated in renal tissues of DKD mice and mesangial cells cultured withhigh glucose, and Hmgb1 cytopasmic accumulation promotes MC inflammation and proliferation. Mechanistically,Hmgb1 cytopasmic accumulation is two-way regulated by MC-specific cyto-lncRNA E130307A14Rik interaction andlactate-mediated acetylated and lactylated Hmgb1 nucleocytoplasmic translocation, and accelerates NFκB signalingpathway activation via directly binding to IκBα. Together, this work reveals the promoting role of Hmgb1 on MCinflammation and proliferation in DKD and helps expound the regulation of Hmgb1 cytopasmic accumulation in twoways. In particular, Hmgb1 may be a promising therapeutic target for DKD.
Collapse
Affiliation(s)
- Keqian Wu
- Department of Nephrology, The Second Affiliated Hospital, Basic Medicine College, Key Laboratory of Major Brain Disease and Aging Research(Ministry of Education), Chongqing Medical University, Chongqing, 400010, China
| | - He Zha
- Department of Laboratory Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou, China
| | - Tianhui Wu
- Department of Nephrology, The Second Affiliated Hospital, Basic Medicine College, Key Laboratory of Major Brain Disease and Aging Research(Ministry of Education), Chongqing Medical University, Chongqing, 400010, China
- School of Public Health and Laboratory, Qilu Medical University, Zibo, 255300, Shandong, China
| | - Handeng Liu
- Laboratory of Tissue and Cell Biology, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, 400016, China
| | - Rui Peng
- Department of Bioinformatics, Chongqing Medical University, Chongqing, 400016, China
| | - Ziyue Lin
- Department of Nephrology, The Second Affiliated Hospital, Basic Medicine College, Key Laboratory of Major Brain Disease and Aging Research(Ministry of Education), Chongqing Medical University, Chongqing, 400010, China
| | - Dan Lv
- Department of Nephrology, The Second Affiliated Hospital, Basic Medicine College, Key Laboratory of Major Brain Disease and Aging Research(Ministry of Education), Chongqing Medical University, Chongqing, 400010, China
| | - Xiaohui Liao
- Department of Nephrology, The Second Affiliated Hospital, Basic Medicine College, Key Laboratory of Major Brain Disease and Aging Research(Ministry of Education), Chongqing Medical University, Chongqing, 400010, China.
| | - Yan Sun
- Department of Nephrology, The Second Affiliated Hospital, Basic Medicine College, Key Laboratory of Major Brain Disease and Aging Research(Ministry of Education), Chongqing Medical University, Chongqing, 400010, China.
| | - Zheng Zhang
- Department of Nephrology, The Second Affiliated Hospital, Basic Medicine College, Key Laboratory of Major Brain Disease and Aging Research(Ministry of Education), Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
49
|
Fang Y, Zhang X, Huang R, Liu J, Li Z. Gut microbiota and autoimmune thyroid disease: a bidirectional Mendelian randomization study and mediation analysis. Front Microbiol 2024; 15:1443643. [PMID: 39351300 PMCID: PMC11439789 DOI: 10.3389/fmicb.2024.1443643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Background The gut microbiota (GM) plays a pivotal role in influencing various health outcomes, including immune-mediated conditions, but its potential association with autoimmune thyroid disease (AITD) remains underexplored. We aimed to investigate the potentially pathogenic or protective causal impacts of specific GM on two types of AITD, namely Graves' disease and Hashimoto's thyroiditis, and analyzed the mediating effect of 731 immune cell phenotypes. Methods Leveraging pooled genome-wide association study (GWAS) data of 211 gut microbiota traits, 731 immune cell phenotypes, and two types of AITD (Hashimoto's thyroiditis and Graves' disease), we performed bidirectional Mendelian randomization (MR) analyses to explore the causal relationships between the GM and AITD. Subsequently, we employed a multivariable MR analysis to discover potential mediating immune cell traits. Additionally, sensitivity analyses were utilized to ensure the reliability of the outcomes. Results Our analysis revealed that a total of 7 GM taxa were positively associated with AITD, and other 14 taxa showed a negative correlation with AITD. Furthermore, we identified several immune cell traits that mediated the effects of GM on AITD. Most notably, Actinobacteria (p) presented protective effects on Hashimoto's thyroiditis via CCR2 on myeloid Dendritic Cell (5.0%), and Bifidobacterium (g) showed facilitating effects on Graves' disease through CD39+ CD4+ T cell %CD4+ T cell (5.0%) and CD14 on CD33+ HLA DR+ CD14dim (12.2%). Conclusion The current MR study provides evidence supporting the causal relationships between several specific GM taxa and AITD, and further identified potential mediating immunophenotypes.
Collapse
Affiliation(s)
- Yiqiao Fang
- Division of Thyroid Surgery, Department of General Surgery, Laboratory of Thyroid and Parathyroid Diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-Related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyue Zhang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-Related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Huang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaye Liu
- Division of Thyroid Surgery, Department of General Surgery, Laboratory of Thyroid and Parathyroid Diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-Related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihui Li
- Division of Thyroid Surgery, Department of General Surgery, Laboratory of Thyroid and Parathyroid Diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-Related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
Liu Y, Huang M, Sun Y, Dai W. Exploring the effect of lifestyle behaviors and socioeconomic status on atrial fibrillation: the mediating role of 91 inflammatory cytokines. Front Cardiovasc Med 2024; 11:1401384. [PMID: 39328240 PMCID: PMC11424413 DOI: 10.3389/fcvm.2024.1401384] [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: 04/08/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Background Atrial fibrillation (AF) is one of the most prevalent cardiac arrhythmias and has a significant economic and social burden. Whether it is associated with lifestyle behaviors and socioeconomic status is currently poorly understood. This study aimed to explore the relationship among these factors and determine the role of inflammatory cytokines. Method We investigated the causal effects of lifestyle behaviors and socioeconomic status on AF using bidirectional two-sample Mendelian randomization (MR). Instrumental variables were obtained from a publicly available genome-wide association study. A two-step MR was conducted to determine the mediating role of 91 inflammatory cytokines. Inverse variance weighted was used as the main method with four supplementary MR methods. To obtain more robust results, several sensitivity analyses were conducted. Result The results indicated that seven of the lifestyle behaviors [smoking initiation, vegetable intake, coffee consumption (cups/day), dozing, lifetime smoking index, napping, and alcohol abuse] were potential risk factors for AF. One socioeconomic status, education attainment (years of education), was causally associated with a decreased risk of AF. Moreover, we found that thymic stromal lymphopoietin, CD40l receptor, C-X-C motif chemokine 6, and C-X-C motif chemokine 11 levels mediated the causal effect, at proportions of 13.6%, 4.1%, 4.3%, and 6.9%, respectively. Conclusion Our findings provide insight into the relationship between lifestyle behaviors, socioeconomic status, and AF. Inflammatory cytokines are potential mediators of this relationship.
Collapse
Affiliation(s)
- Yiheng Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingsheng Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Sun
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiran Dai
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|