1
|
Chen H, Wu B, Guan K, Chen L, Chai K, Ying M, Li D, Zhao W. Identification of lipid metabolism related immune markers in atherosclerosis through machine learning and experimental analysis. Front Immunol 2025; 16:1549150. [PMID: 40070840 PMCID: PMC11893410 DOI: 10.3389/fimmu.2025.1549150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background Atherosclerosis is a significant contributor to cardiovascular disease, and conventional diagnostic methods frequently fall short in the timely and accurate detection of early-stage atherosclerosis. Abnormal lipid metabolism plays a critical role in the development of atherosclerosis. Consequently, the identification of new diagnostic markers is essential for the precise diagnosis of this condition. Method The datasets related to atherosclerosis utilized in this research were obtained from the GEO database (GSE2470, GSE24495, GSE100927 and GSE43292). The ssGSEA technique was first utilized to assess lipid metabolism scores in samples affected by atherosclerosis, thereby aiding in the discovery of important regulatory genes linked to lipid metabolism via WGCNA. Following this, differential expression analysis and functional evaluations were carried out, after which various machine learning approaches were employed to determine significant diagnostic genes for atherosclerosis. A diagnostic model was then developed and validated through several machine learning algorithms. Furthermore, molecular docking studies were conducted to analyze the binding affinity of these key markers with therapeutic agents for atherosclerosis. The ssGSEA technique was also used to measure immune cell scores in atherosclerotic samples, aiding the exploration of the connection between key diagnostic markers and immune cells. Finally, the expression variations of the identified pivotal genes were confirmed through experimental validation. Result WGCNA identified 302 lipid metabolism-related genes in atherosclerotic samples, and functional analysis revealed that these genes are associated with multiple immune pathways. Through further differential analysis and screening using machine learning algorithms, APLNR, PCDH12, PODXL, SLC40A1, TM4SF18, and TNFRSF25 were identified as key diagnostic genes for atherosclerosis. The diagnostic model we constructed was confirmed to predict the occurrence of atherosclerosis with high accuracy, and molecular docking studies indicated that these six key diagnostic genes have potential as drug targets. Additionally, the ssGSEA algorithm further validated the association of these diagnostic genes with various immune cells. Finally, the expression levels of these six genes were experimentally confirmed. Conclusion Our study introduces novel lipid metabolism-related diagnostic markers for atherosclerosis and emphasizes their potential as immune-related drug targets. This research provides a valuable approach for the predictive diagnosis and targeted therapy of atherosclerosis.
Collapse
Affiliation(s)
- Hang Chen
- Department of Thyroid Breast Vascular Surgery, Banan Hospital of Chongqing Medical University, Chongqing, China
| | - Biao Wu
- Department of Vascular Surgery, Changhai Hospital Affiliated to Naval Medical University, Shanghai, China
- Cancer Research Centre Nantong, Nantong Tumor Hospital, Nantong, China
| | - Kunyu Guan
- Pediatrics, Changhai Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Liang Chen
- Department of Vascular Surgery, Changhai Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Kangjie Chai
- Department of Vascular Surgery, Changhai Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Maoji Ying
- General Practice, Changhai Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Dazhi Li
- Department of Vascular Surgery, Changhai Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Weicheng Zhao
- Department of Interventional, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
2
|
Khalaf K, Chamieh M, Welc N, Singh C, Kaouk JL, Kaouk A, Mackiewicz A, Kaczmarek M, Perek B. Cellular aspects of immunity involved in the development of atherosclerosis. Front Immunol 2025; 16:1461535. [PMID: 39944697 PMCID: PMC11813763 DOI: 10.3389/fimmu.2025.1461535] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 01/09/2025] [Indexed: 05/09/2025] Open
Abstract
Atherosclerosis, previously regarded as a lipid storage disease, has now been classified as a chronic inflammatory disease. The hardening of arterial vessels characterizes atherosclerosis due to the accumulation of lipids in the arterial walls, eliciting an inflammatory response. The development of atherosclerosis occurs in various stages and is facilitated by many clinical factors, such as hypertension, hyperlipidemia, and inflammatory status. A large arsenal of cells has been implicated in its development. This review will summarize the phases of atherosclerotic formation and all the cells involved in either promoting or inhibiting its development.
Collapse
Affiliation(s)
- Khalil Khalaf
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marc Chamieh
- Department of Spine Disorders and Pediatric Orthopedics, Poznan University of Medical Sciences, Poznań, Poland
| | - Natalia Welc
- Department of Dermatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Chandpreet Singh
- Department of Internal Medicine, University of California, Los Angeles (UCLA) - Kern Medical Center, Bakersfield, CA, United States
| | - Joanne Lynn Kaouk
- Department of Science, Louisiana State University, Lousiana, LA, United States
| | - Aiden Kaouk
- Department of Natural Sciences, The University of Texas at Austin, Texas, TX, United States
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
| | - Bartlomiej Perek
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
3
|
Zhao Z, Qin Y, Wu R, Li W, Dong Y. Single-cell analysis identified key macrophage subpopulations associated with atherosclerosis. Open Med (Wars) 2024; 19:20241088. [PMID: 39726810 PMCID: PMC11669903 DOI: 10.1515/med-2024-1088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/13/2024] [Accepted: 10/18/2024] [Indexed: 12/28/2024] Open
Abstract
Background Atherosclerosis is a lipid-driven inflammatory disease characterized by plaque formation in major arteries. These plaques contain lipid-rich macrophages that accumulate through monocyte recruitment, local macrophage differentiation, and proliferation. Objective We identify the macrophage subsets that are closely related to atherosclerosis and reveal the key pathways in the progression of atherosclerotic disease. Materials and methods In this study, we characterize the single-cell landscape of atherosclerosis, identifying macrophage subsets closely related to the disease and revealing key pathways in its progression. Using analytical methods like CytoTRACE, Monocle2, Slingshot, and CellChat, we study macrophage differentiation and infer cell trajectory. Results The 8,417 macrophages were divided into six subtypes, macrophages: C0 C1QC+ macrophages, C1 SPP1+ macrophages, C2 FCN1+ macrophages, C3 IGKC+ macrophages, C4 FCER1A+ macrophages, C5CALD1+ macrophages. The results of gene set enrichment analysis, Monocle2, and Slingshot suggest that C2 FCN1+ macrophages may play an important role in the progression of atherosclerosis. C2 FCN1+ macrophages interact with endothelial cells via CCL, CXCL, APP, and other pathways to regulate the progression of atherosclerosis. Conclusion We identify a key macrophage subgroup (C2 FCN1+ macrophages) associated with atherosclerosis, which interacts with endothelial cells via CCL, CXCL, APP, and other pathways to regulate disease progression.
Collapse
Affiliation(s)
- Zhenzhen Zhao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
- Department of Cardiovascular Disease, The Second Affiliated Hospital of Shandong University of Chinese Medicine, Jinan, 250001, China
| | - Yuelong Qin
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Rui Wu
- Pingyi County Hospital of Traditional Chinese Medicine Cardiology Department, Linyi, 273300, China
| | - Wenwu Li
- Department of Burn Plastic and Wound Repair Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Laboratory of the Atherosclerosis and Ischemic Cardiovascular Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yujiang Dong
- Department of Cardiovascular Disease, The Second Affiliated Hospital of Shandong University of Chinese Medicine, Jinan, 250001, China
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| |
Collapse
|
4
|
Chen H, Zhang L, Liu M, Li Y, Chi Y. Multi-Omics Research on Angina Pectoris: A Novel Perspective. Aging Dis 2024:AD.2024.1298. [PMID: 39751862 DOI: 10.14336/ad.2024.1298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Angina pectoris (AP), a clinical syndrome characterized by paroxysmal chest pain, is caused by insufficient blood supply to the coronary arteries and sudden temporary myocardial ischemia and hypoxia. Long-term AP typically induces other cardiovascular events, including myocardial infarction and heart failure, posing a serious threat to patient safety. However, AP's complex pathological mechanisms and developmental processes introduce significant challenges in the rapid diagnosis and accurate treatment of its different subtypes, including stable angina pectoris (SAP), unstable angina pectoris (UAP), and variant angina pectoris (VAP). Omics research has contributed significantly to revealing the pathological mechanisms of various diseases with the rapid development of high-throughput sequencing approaches. The application of multi-omics approaches effectively interprets systematic information on diseases from the perspective of genes, RNAs, proteins, and metabolites. Integrating multi-omics research introduces novel avenues for identifying biomarkers to distinguish different AP subtypes. This study reviewed articles related to multi-omics and AP to elaborate on the research progress in multi-omics approaches (including genomics, transcriptomics, proteomics, and metabolomics), summarized their applications in screening biomarkers employed to discriminate multiple AP subtypes, and delineated integration methods for multi-omics approaches. Finally, we discussed the advantages and disadvantages of applying a single-omics approach in distinguishing diverse AP subtypes. Our review demonstrated that the integration of multi-omics technologies is preferable for quick and precise diagnosis of the three AP types, namely SAP, UAP, and VAP.
Collapse
Affiliation(s)
- Haiyang Chen
- Department of Psycho-cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lijun Zhang
- Department of Psycho-cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Meiyan Liu
- Department of Psycho-cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yanwei Li
- Department of Psycho-cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Yunpeng Chi
- Department of Psycho-cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Huang M, Zhou P, Hang Y, Wu D, Zhao N, Yao G, Tang X, Sun L. CFL1 restores the migratory capacity of bone marrow mesenchymal stem cells in primary Sjögren's syndrome by regulating CCR1 expression. Int Immunopharmacol 2024; 128:111485. [PMID: 38183912 DOI: 10.1016/j.intimp.2024.111485] [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: 10/27/2023] [Revised: 12/26/2023] [Accepted: 01/01/2024] [Indexed: 01/08/2024]
Abstract
BACKGROUND Primary Sjögren's syndrome (pSS) is a chronic systemic autoimmune disease. There is no relevant research on whether the migratory ability of bone marrow mesenchymal stem cells (BM-MSC) is impaired in patients with pSS (pSS-BMMSC). METHODS Trajectories and velocities of BM-MSC were analyzed. Transwell migration assay and wound healing assay were used to investigate the migratory capacity of BM-MSC. The proliferative capacity of BM-MSC was evaluated by EDU and CCK8 assay. RNA-seq analysis was then performed to identify the underlying mechanism of lentivirus-mediated cofilin-1 overexpression BM-MSC (BMMSCCFL1). The therapeutic efficacy of BMMSCCFL1 was evaluated in NOD mice. RESULTS The migratory capacity of pSS-BMMSC was significantly reduced compared to normal volunteers (HC-BMMSC). The expression of the motility-related gene CFL1 was decreased in pSS-BMMSC. Lentivirus-mediated CFL1 overexpression of pSS-BMMSC promoted the migration capacity of pSS-BMMSC. Furthermore, RNA-seq revealed that CCR1 was the downstream target gene of CFL1. To further elucidate the mechanism of CFL1 in regulating BM-MSC migration and proliferation via the CCL5/CCR1 axis, we performed a rescue experiment using BX431 (a CCR1-specific inhibitor) to inhibit CCR1. The results showed that CCR1 inhibitors suppressed the migration and proliferation capacity of MSC induced by CFL1. CONCLUSION The pSS-BMMSC leads to impaired migration and proliferation, and overexpression of CFL1 can rescue the functional deficiency and alleviate disease symptoms in NOD mice. Mechanically, CFL1 can regulate the expression level of the downstream CCL5/CCR1 axis to enhance the migration and proliferation of BM-MSC.
Collapse
Affiliation(s)
- Mengxi Huang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Panpan Zhou
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Yang Hang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Dan Wu
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Nan Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China.
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China; Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China.
| |
Collapse
|