1
|
Tao Y, Xiong M, Peng Y, Yao L, Zhu H, Zhou Q, Ouyang J. Machine learning-based identification and validation of immune-related biomarkers for early diagnosis and targeted therapy in diabetic retinopathy. Gene 2025; 934:149015. [PMID: 39427825 DOI: 10.1016/j.gene.2024.149015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
The early diagnosis of diabetic retinopathy (DR) is challenging, highlighting the urgent need to identify new biomarkers. Immune responses play a crucial role in DR, yet there are currently no reports of machine learning (ML) algorithms being utilized for the development of immune-related molecular markers in DR. Based on the datasets GSE102485 and GSE160306, differentially expressed genes (DEGs) were screened using Weighted Gene Co-expression Network Analysis (WGCNA). Five ML algorithms including Bayesian, Learning Vector Quantization (LVQ), Wrapper (Boruta), Random Forest (RF), and Logistic Regression were employed to select immune-related genes associated with DR (DR.Sig). Seven ML algorithms including Naive Bayes (NB), RF, Support Vector Machine (SVM), AdaBoost Classification Trees (AdaBoost), Boosted Logistic Regressions (LogitBoost), K-Nearest Neighbors (KNN), and Cancerclass were utilized to construct a predictive model for DR. The relationship between DR.Sig genes and immune cells was analyzed using single-sample Gene Set Enrichment Analysis (ssGSEA). Additionally, drug sensitivity prediction of DR.Sig genes and molecular docking were performed. Through the utilization of 5 ML algorithms, 6 immune-related biomarkers closely related to the occurrence of DR were identified, including FCGR2B, CSRP1, EDNRA, SDC2, TEK, and CIITA. The DR predictive model constructed based on these 6 DR.Sig genes using the Cancerclass algorithm demonstrated superior predictive performance compared to 4 previously published DR-related biomarkers. In vivo and in vitro experiments also provided strong validation of the expression of the 6 genes in DR. Positive correlations were observed between these genes and 22 types of immune cells. Molecular docking results revealed that CSRP1, EDNRA, and TEK exhibited the highest affinities with the small molecule compounds etoposide, FR-139317, and camptothecin, respectively. The models constructed based on various ML algorithms can effectively predict the occurrence of DR events and hold potential for targeted drug therapies, providing a basis for the early diagnosis and targeted treatment of DR.
Collapse
Affiliation(s)
- Yulin Tao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; Department of Ophthalmology, Jiujiang No 1 Peoples Hospital, Jiujiang 332000, China
| | - Minqi Xiong
- The Chinese University of Hong Kong, Shenzhen 518100, China
| | - Yirui Peng
- School of Life Sciences, Xiamen University, Xiamen 361000, China
| | - Lili Yao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Haibo Zhu
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Qiong Zhou
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| | - Jun Ouyang
- Department of Ophthalmology, Jiujiang No 1 Peoples Hospital, Jiujiang 332000, China.
| |
Collapse
|
2
|
Brown SD, Klimi E, Bakker WAM, Beqqali A, Baker AH. Non-coding RNAs to treat vascular smooth muscle cell dysfunction. Br J Pharmacol 2025; 182:246-280. [PMID: 38773733 DOI: 10.1111/bph.16409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/19/2024] [Accepted: 03/14/2024] [Indexed: 05/24/2024] Open
Abstract
Vascular smooth muscle cell (vSMC) dysfunction is a critical contributor to cardiovascular diseases, including atherosclerosis, restenosis and vein graft failure. Recent advances have unveiled a fascinating range of non-coding RNAs (ncRNAs) that play a pivotal role in regulating vSMC function. This review aims to provide an in-depth analysis of the mechanisms underlying vSMC dysfunction and the therapeutic potential of various ncRNAs in mitigating this dysfunction, either preventing or reversing it. We explore the intricate interplay of microRNAs, long-non-coding RNAs and circular RNAs, shedding light on their roles in regulating key signalling pathways associated with vSMC dysfunction. We also discuss the prospects and challenges associated with developing ncRNA-based therapies for this prevalent type of cardiovascular pathology. LINKED ARTICLES: This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
MESH Headings
- Animals
- Humans
- Cardiovascular Diseases/drug therapy
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- RNA, Circular/genetics
- RNA, Circular/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- RNA, Untranslated/pharmacology
- RNA, Untranslated/therapeutic use
Collapse
Affiliation(s)
- Simon D Brown
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Eftychia Klimi
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | - Abdelaziz Beqqali
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Andrew H Baker
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
3
|
Xiong J, Chen G, Lin B, Zhong L, Jiang X, Lu H. Integrative analysis of single-Cell RNA sequencing and experimental validation in the study of abdominal aortic aneurysm progression. Gene 2024; 929:148820. [PMID: 39103059 DOI: 10.1016/j.gene.2024.148820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/13/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a complex vascular disorder characterized by the progressive dilation of the abdominal aorta, with a high risk of rupture and mortality. Understanding the cellular interactions and molecular mechanisms underlying AAA development is critical for identifying potential therapeutic targets. METHODS This study utilized datasets GSE197748, GSE164678 and GSE183464 from the GEO database, encompassing bulk and single-cell RNA sequencing data from AAA and control samples. We performed principal component analysis, differential expression analysis, and functional enrichment analysis to identify key pathways involved in AAA. Cell-cell interactions were investigated using CellPhoneDB, focusing on fibroblasts, vascular smooth muscle cells (VSMCs), and macrophages. We further validated our findings using a mouse model of AAA induced by porcine pancreatic enzyme infusion, followed by gene expression analysis and co-immunoprecipitation experiments. RESULTS Our analysis revealed significant alterations in gene expression profiles between AAA and control samples, with a pronounced immune response and cell adhesion pathways being implicated. Single-cell RNA sequencing data highlighted an increased proportion of pro-inflammatory macrophages, along with changes in the composition of fibroblasts and VSMCs in AAA. CellPhoneDB analysis identified critical ligand-receptor interactions, notably collagen type I alpha 1 chain (COL1A1)/COL1A2-CD18 and thrombospondin 1 (THBS1)-CD3, suggesting complex communication networks between fibroblasts and VSMCs. In vivo experiments confirmed the upregulation of these genes in AAA mice and demonstrated the functional interaction between COL1A1/COL1A2 and CD18. CONCLUSION The interaction between fibroblasts and VSMCs, mediated by specific ligand-receptor pairs such as COL1A1/COL1A2-CD18 and THBS1-CD3, plays a pivotal role in AAA pathogenesis.
Collapse
MESH Headings
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Animals
- Mice
- Single-Cell Analysis/methods
- Humans
- Sequence Analysis, RNA/methods
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Macrophages/metabolism
- Disease Progression
- Fibroblasts/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Male
- Mice, Inbred C57BL
- Gene Expression Profiling/methods
- Cell Communication/genetics
- Collagen Type I/genetics
- Collagen Type I/metabolism
Collapse
Affiliation(s)
- Jie Xiong
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China
| | - Guojun Chen
- Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou 510515, China
| | - Beiyou Lin
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China
| | - Lintao Zhong
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China
| | - Xiaofei Jiang
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China.
| | - Hongyun Lu
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China.
| |
Collapse
|
4
|
Maegdefessel L, Fasolo F. Long Noncoding RNA Function in Smooth Muscle Cell Plasticity and Atherosclerosis. Arterioscler Thromb Vasc Biol 2024. [PMID: 39633574 DOI: 10.1161/atvbaha.124.320393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
In the healthy mature artery, vascular cells, including endothelial cells, smooth muscle cells (SMCs), and fibroblasts are organized in different layers, performing specific functions. SMCs located in the media are in a differentiated state and exhibit a contractile phenotype. However, in response to vascular injury within the intima, stimuli from activated endothelial cells and recruited inflammatory cells reach SMCs and induce a series of remodeling events in them, known as phenotypic switching. Indeed, SMCs retain a certain degree of plasticity and are able to transdifferentiate into other cell types that are crucial for both the formation and development of atherosclerotic lesions. Because of their highly cell-specific expression profiles and their widely recognized contribution to physiological and disease-related biological processes, long noncoding RNAs have received increasing attention in atherosclerosis research. Dynamic fluctuations in their expression have been implicated in the regulation of SMC identity. Sophisticated technologies are now available to allow researchers to access single-cell transcriptomes and study long noncoding RNA function with unprecedented precision. Here, we discuss the state of the art of long noncoding RNAs regulation of SMC phenotypic switching, describing the methodologies used to approach this issue and evaluating the therapeutic perspectives of exploiting long noncoding RNAs as targets in atherosclerosis.
Collapse
Affiliation(s)
- Lars Maegdefessel
- Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University Munich, Germany (L.M., F.F.)
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Berlin, Germany (L.M., F.F.)
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden (L.M.)
| | - Francesca Fasolo
- Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University Munich, Germany (L.M., F.F.)
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Berlin, Germany (L.M., F.F.)
| |
Collapse
|
5
|
Li J, Wang X. Functional roles of conserved lncRNAs and circRNAs in eukaryotes. Noncoding RNA Res 2024; 9:1271-1279. [PMID: 39036601 PMCID: PMC11260338 DOI: 10.1016/j.ncrna.2024.06.014] [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: 12/20/2023] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 07/23/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) have emerged as critical regulators in essentially all biological processes across eukaryotes. They exert their functions through chromatin remodeling, transcriptional regulation, interacting with RNA-binding proteins (RBPs), serving as microRNA sponges, etc. Although non-coding RNAs are typically more species-specific than coding RNAs, a number of well-characterized lncRNA (such as XIST and NEAT1) and circRNA (such as CDR1as and ciRS-7) are evolutionarily conserved. The studies on conserved lncRNA and circRNAs across multiple species could facilitate a comprehensive understanding of their roles and mechanisms, thereby overcoming the limitations of single-species studies. In this review, we provide an overview of conserved lncRNAs and circRNAs, and summarize their conserved roles and mechanisms.
Collapse
Affiliation(s)
- Jingxin Li
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, The RNA Institute, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China (UTSC), Hefei, 230027, Anhui, China
| | - Xiaolin Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, The RNA Institute, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China (UTSC), Hefei, 230027, Anhui, China
| |
Collapse
|
6
|
Hou X, Liang F, Li J, Yang Y, Wang C, Qi T, Sheng W. Mapping cell diversity in human sporadic cerebral cavernous malformations. Gene 2024; 924:148605. [PMID: 38788816 DOI: 10.1016/j.gene.2024.148605] [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: 10/23/2023] [Revised: 05/12/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Cerebral cavernous malformation (CCM) is a low-flow, bleeding-prone vascular disease that can cause cerebral hemorrhage, seizure and neurological deficits. Its inheritance mode includes sporadic or autosomal dominant inheritance with incomplete penetrance, namely sporadic CCM (SCCM) and familial CCM. SCCM is featured by single lesion and single affection in a family. Among CCM patients especially SCCM, the pathogenesis of the corresponding phenotypes and pathological features or candidate genes have not been fully elucidated yet. METHODS Here, we performed in-depth single-cell RNA sequencing (scRNA-Seq) and bulk assay for transposase-accessible chromatin sequencing (ATAC-Seq) in SCCM and control patients. Further validation was conducted for the gene of interest using qPCR and RNA in situ hybridization (RNA FISH) techniques to provide further atlas and evidence for SCCM generative process. RESULTS We identified six cell types in the SCCM and control vessels and found that the expression of NEK1, RNPC3, FBRSL1, IQGAP2, MCUB, AP3B1, ESCO1, MYO9B and PVT1 were up-regulated in SCCM tissues. Among the six cell types, we found that compared with control conditions, PVT1 showed a rising peak which followed the pseudo-time axis in endothelial cell clusters of SCCM samples, while showed an increasing trend in smooth muscle cell clusters of SCCM samples. Further experiments indicated that, compared with the control vessels, PVT1 exhibited significantly elevated expression in SCCM samples. CONCLUSION In SCCM conditions, We found that in the process of development from control to lesion conditions, PVT1 showed a rising peak in endothelial cells and showed an increasing trend in smooth muscle cells at the same time. Overall, there was a significantly elevated expression of NEK1, RNPC3, FBRSL1, IQGAP2, MCUB, AP3B1, ESCO1, MYO9B and PVT1 in SCCM specimens compared to control samples.
Collapse
Affiliation(s)
- Xiaocan Hou
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Feng Liang
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Jiaoxing Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Yibing Yang
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Chuhuai Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, China.
| | - Tiewei Qi
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, China.
| | - Wenli Sheng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China.
| |
Collapse
|
7
|
Gluba-Sagr A, Franczyk B, Rysz-Górzyńska A, Olszewski R, Rysz J. The Role of Selected lncRNAs in Lipid Metabolism and Cardiovascular Disease Risk. Int J Mol Sci 2024; 25:9244. [PMID: 39273193 PMCID: PMC11395304 DOI: 10.3390/ijms25179244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 09/15/2024] Open
Abstract
Lipid disorders increase the risk for the development of cardiometabolic disorders, including type 2 diabetes, atherosclerosis, and cardiovascular disease. Lipids levels, apart from diet, smoking, obesity, alcohol consumption, and lack of exercise, are also influenced by genetic factors. Recent studies suggested the role of long noncoding RNAs (lncRNAs) in the regulation of lipid formation and metabolism. Despite their lack of protein-coding capacity, lncRNAs are crucial regulators of various physiological and pathological processes since they affect the transcription and epigenetic chromatin remodelling. LncRNAs act as molecular signal, scaffold, decoy, enhancer, and guide molecules. This review summarises available data concerning the impact of lncRNAs on lipid levels and metabolism, as well as impact on cardiovascular disease risk. This relationship is significant because altered lipid metabolism is a well-known risk factor for cardiovascular diseases, and lncRNAs may play a crucial regulatory role. Understanding these mechanisms could pave the way for new therapeutic strategies to mitigate cardiovascular disease risk through targeted modulation of lncRNAs. The identification of dysregulated lncRNAs may pose promising candidates for therapeutic interventions, since strategies enabling the restoration of their levels could offer an effective means to impede disease progression without disrupting normal biological functions. LncRNAs may also serve as valuable biomarker candidates for various pathological states, including cardiovascular disease. However, still much remains unknown about the functions of most lncRNAs, thus extensive studies are necessary elucidate their roles in physiology, development, and disease.
Collapse
Affiliation(s)
- Anna Gluba-Sagr
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland
| | - Aleksandra Rysz-Górzyńska
- Department of Ophthalmology and Visual Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland
| | - Robert Olszewski
- Department of Gerontology, Public Health and Didactics, National Institute of Geriatrics, Rheumatology and Rehabilitation in Warsaw, 02-637 Warsaw, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland
| |
Collapse
|
8
|
Zhao C, Wang Y, Wang H, Sharma A, Wu Y, Schmidt-Wolf IGH, Wang Z. CSRP1 gene: a potential novel prognostic marker in acute myeloid leukemia with implications for immune response. Discov Oncol 2024; 15:248. [PMID: 38937285 PMCID: PMC11211298 DOI: 10.1007/s12672-024-01088-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/06/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia, constituting a majority of leukemias, grapples with a 24% 5-year survival rate. Recent strides in research have unveiled fresh targets for drug therapies. LIM-only, a pivotal transcription factor within LIM proteins, oversees cell development and is implicated in tumor formation. Among these critical LIM proteins, CSRP1, a Cysteine-rich protein, emerges as a significant player in various diseases. Despite its recognition as a potential prognostic factor and therapeutic target in various cancers, the specific link between CSRP1 and acute myeloid leukemia remains unexplored. Our previous work, identifying CSRP1 in a prognostic model for AML patients, instigates a dedicated exploration into the nuanced role of CSRP1 in acute myeloid leukemia. METHODS R tool was conducted to analyze the public data. qPCR was applied to evaluate the expression of CSRP1 mRNA for clinical samples and cell line. Unpaired t test, Wilcoxon Rank Sum test, KM curves, spearman correlation test and Pearson correlation test were included in this study. RESULTS CSRP1 displays notable expression variations between normal and tumor samples in acute myeloid leukemia (AML). It stands out as an independent prognostic factor for AML patients, showing correlations with clinical factors like age and cytogenetics risk. Additionally, CSRP1 correlates with immune-related pathways, immune cells, and immune checkpoints in AML. Furthermore, the alteration of CSRP1 mRNA levels is observed upon treatment with a DNMT1 inhibitor for THP1 cells. CONCLUSION The CSRP1 has potential as a novel prognostic factor and appears to influence the immune response in acute myeloid leukemia. Additionally, there is an observed association between CSRP1 and DNA methylation in acute myeloid leukemia.
Collapse
Affiliation(s)
- Chunxia Zhao
- Department of Nursing, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yulu Wang
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Huan Wang
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital of Bonn, Bonn, Germany
- Department of Neurosurgery, University Hospital of Bonn, Bonn, Germany
| | - Yun Wu
- Department of Hematology, Shangrao People's Hospital, The Affiliated Shangrao Hospital of Nanchang University, Shangrao, China
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital of Bonn, Bonn, Germany
| | - Zifeng Wang
- Department of Hematology, Shangrao People's Hospital, The Affiliated Shangrao Hospital of Nanchang University, Shangrao, China.
| |
Collapse
|
9
|
Liu Y, Sun X, Gou Z, Deng Z, Zhang Y, Zhao P, Sun W, Bai Y, Jing Y. Epigenetic modifications in abdominal aortic aneurysms: from basic to clinical. Front Cardiovasc Med 2024; 11:1394889. [PMID: 38895538 PMCID: PMC11183338 DOI: 10.3389/fcvm.2024.1394889] [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: 03/11/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Abdominal Aortic Aneurysm (AAA) is a disease characterized by localized dilation of the abdominal aorta, involving multiple factors in its occurrence and development, ultimately leading to vessel rupture and severe bleeding. AAA has a high mortality rate, and there is a lack of targeted therapeutic drugs. Epigenetic regulation plays a crucial role in AAA, and the treatment of AAA in the epigenetic field may involve a series of related genes and pathways. Abnormal expression of these genes may be a key factor in the occurrence of the disease and could potentially serve as promising therapeutic targets. Understanding the epigenetic regulation of AAA is of significant importance in revealing the mechanisms underlying the disease and identifying new therapeutic targets. This knowledge can contribute to offering AAA patients better clinical treatment options beyond surgery. This review systematically explores various aspects of epigenetic regulation in AAA, including DNA methylation, histone modification, non-coding RNA, and RNA modification. The analysis of the roles of these regulatory mechanisms, along with the identification of relevant genes and pathways associated with AAA, is discussed comprehensively. Additionally, a comprehensive discussion is provided on existing treatment strategies and prospects for epigenetics-based treatments, offering insights for future clinical interventions.
Collapse
Affiliation(s)
- YuChen Liu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - XiaoYun Sun
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Zhen Gou
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - ZhenKun Deng
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - YunRui Zhang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - PingPing Zhao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Wei Sun
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Yang Bai
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - YuChen Jing
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
He T, Li C, Chen Q, Li R, Luo J, Mao J, Yang Z. Combined analysis of lncRNA and mRNA emphasizes the potential role of tryptophan-mediated regulation of muscle development in weaned piglets by lncRNA. J Anim Sci 2024; 102:skae264. [PMID: 39276131 PMCID: PMC11465388 DOI: 10.1093/jas/skae264] [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: 06/24/2024] [Accepted: 09/13/2024] [Indexed: 09/16/2024] Open
Abstract
Pork is an important high-value protein source that fulfills the nutritional requirements for normal growth development, repair, and metabolism. Tryptophan (Trp), a crucial amino acid for piglet growth performance and muscle development, has an essential yet unclear regulatory mechanism. To investigate the biological basis of Trp regulation of piglet muscle development and identify the related regulatory pathways, we studied 20 weaned piglets. The piglets were divided into control (CON, 0.14% Trp) and high Trp (HT, 0.35% Trp) groups. They were fed with different Trp concentrations for 28 d, after which we collected the longissimus dorsi (LD) muscle for histomorphometric analysis and RNA extraction. Our results showed that the HT diet significantly increased the average daily weight gain, myocyte number, and muscle fiber density in weaned piglets. We then analyzed the differentially expressed (DE) genes in the LD muscle through RNA sequencing (RNA-seq). We identified 253 lncRNAs and 1,055 mRNAs mainly involved in myoblast proliferation and myofiber formation, particularly through the FoxO and AMPK signaling pathways and metabolism. Further analysis of the DE lncRNA targeting relationship and construction of a protein-protein interaction network resulted in the discovery of a novel lncRNA, XLOC_021675, or FRPMD, and elucidated its role in regulating piglet muscle development. Finally, we confirmed the RNA-seq results by reverse transcription polymerase chain reaction (RT-PCR). This study provides valuable insights into the regulatory mechanism of lncRNA-mediated Trp regulation of muscle development in weaned piglets offering a theoretical basis for optimizing piglet dietary ratios and enhancing pork production.
Collapse
Affiliation(s)
- Tianle He
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Chenlei Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Qingyun Chen
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ruiqian Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Ju Luo
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Jiani Mao
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Zhenguo Yang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| |
Collapse
|
11
|
Kucher AN, Koroleva IA, Nazarenko MS. Pathogenetic Significance of Long Non-Coding RNAs in the Development of Thoracic and Abdominal Aortic Aneurysms. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:130-147. [PMID: 38467550 DOI: 10.1134/s0006297924010085] [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: 10/16/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 03/13/2024]
Abstract
Aortic aneurysm (AA) is a life-threatening condition with a high prevalence and risk of severe complications. The aim of this review was to summarize the data on the role of long non-coding RNAs (lncRNAs) in the development of AAs of various location. Within less than a decade of studies on the role of lncRNAs in AA, using experimental and bioinformatic approaches, scientists have obtained the data confirming the involvement of these molecules in metabolic pathways and pathogenetic mechanisms critical for the aneurysm development. Regardless of the location of pathological process (thoracic or abdominal aorta), AA was found to be associated with changes in the expression of various lncRNAs in the tissue of the affected vessels. The consistency of changes in the expression level of lncRNA, mRNA and microRNA in aortic tissues during AA development has been recordedand regulatory networks implicated in the AA pathogenesis in which lncRNAs act as competing endogenous RNAs (ceRNA networks) have been identified. It was found that the same lncRNA can be involved in different ceRNA networks and regulate different biochemical and cellular events; on the other hand, the same pathological process can be controlled by different lncRNAs. Despite some similarities in pathogenesis and overlapping of involved lncRNAs, the ceRNA networks described for abdominal and thoracic AA are different. Interactions between lncRNAs and other molecules, including those participating in epigenetic processes, have also been identified as potentially relevant to the AA pathogenesis. The expression levels of some lncRNAs were found to correlate with clinically significant aortic features and biochemical parameters. Identification of regulatory RNAs functionally significant in the aneurysm development is important for clarification of disease pathogenesis and will provide a basis for early diagnostics and development of new preventive and therapeutic drugs.
Collapse
Affiliation(s)
- Aksana N Kucher
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Iuliia A Koroleva
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Maria S Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia.
| |
Collapse
|
12
|
Fidler TP, Dunbar A, Kim E, Hardaway B, Pauli J, Xue C, Abramowicz S, Xiao T, O’Connor K, Sachs N, Wang N, Maegdefessel L, Levine R, Reilly M, Tall AR. Suppression of IL-1β promotes beneficial accumulation of fibroblast-like cells in atherosclerotic plaques in clonal hematopoiesis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:60-75. [PMID: 38362011 PMCID: PMC10868728 DOI: 10.1038/s44161-023-00405-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 11/30/2023] [Indexed: 02/17/2024]
Abstract
Clonal hematopoiesis (CH) is an independent risk factor for atherosclerotic cardiovascular disease. Murine models of CH suggest a central role of inflammasomes and IL-1β in accelerated atherosclerosis and plaque destabilization. Here we show using single-cell RNA sequencing in human carotid plaques that inflammasome components are enriched in macrophages, while the receptor for IL-1β is enriched in fibroblasts and smooth muscle cells (SMCs). To address the role of inflammatory crosstalk in features of plaque destabilization, we conducted SMC fate mapping in Ldlr-/- mice modeling Jak2VF or Tet2 CH treated with IL-1β antibodies. Unexpectedly, this treatment minimally affected SMC differentiation, leading instead to a prominent expansion of fibroblast-like cells. Depletion of fibroblasts from mice treated with IL-1β antibody resulted in thinner fibrous caps. Conversely, genetic inactivation of Jak2VF during plaque regression promoted fibroblast accumulation and fibrous cap thickening. Our studies suggest that suppression of inflammasomes promotes plaque stabilization by recruiting fibroblast-like cells to the fibrous cap.
Collapse
Affiliation(s)
- Trevor P. Fidler
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Department of Physiology, University of San Francisco, San Francisco, CA, USA
| | - Andrew Dunbar
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eunyoung Kim
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Brian Hardaway
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Jessica Pauli
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich, Germany
| | - Chenyi Xue
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
| | - Sandra Abramowicz
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Tong Xiao
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Kavi O’Connor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadja Sachs
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich, Germany
| | - Nan Wang
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich, Germany
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ross Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Muredach Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Alan R. Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
13
|
Maegdefessel L, Boon RA, Dimmeler S. Noncoding RNAs in the Vasculature: Basic Mechanisms and Therapeutic Perspectives. Arterioscler Thromb Vasc Biol 2024; 44:3-6. [PMID: 38150514 DOI: 10.1161/atvbaha.123.319564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Affiliation(s)
- Lars Maegdefessel
- Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University Munich, Germany (L.M.)
- Department of Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden (L.M.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (L.M.)
| | - Reinier A Boon
- Department of Physiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, the Netherlands (R.A.B.)
- Amsterdam Cardiovascular Sciences, Microcirculation, the Netherlands (R.A.B.)
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Germany (R.A.B., S.D.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Frankfurt Rhein/Main, Germany (R.A.B., S.D.)
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Germany (R.A.B., S.D.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Frankfurt Rhein/Main, Germany (R.A.B., S.D.)
| |
Collapse
|
14
|
Wu Z, Yin H, Guo Y, Yin H, Li Y. Detection of cell-type-enriched long noncoding RNAs in atherosclerosis using single-cell techniques: A brief review. Life Sci 2023; 333:122138. [PMID: 37805167 DOI: 10.1016/j.lfs.2023.122138] [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: 06/15/2023] [Revised: 09/20/2023] [Accepted: 09/29/2023] [Indexed: 10/09/2023]
Abstract
Cardiovascular diseases are the leading causes of mortality and morbidity worldwide. Atherosclerotic plaque underlies the predominant factors and is composed of various cell types, including structure cells, such as endothelial and smooth muscle cells, and immune cells, such as macrophages and T cells. Single-cell RNA sequencing (scRNA-seq) has been extensively applied to decipher these cellular heterogeneities to expand our understanding on the mechanisms of atherosclerosis (AS) and to facilitate identifying cell-type-specific long noncoding RNAs (LncRNAs). LncRNAs have been demonstrated to deeply regulate biological activities at the transcriptional and post-transcriptional levels. A group of well-documented functional lncRNAs in AS have been studied. In our review, we selectively described several lncRNAs involved in the critical process of AS. We highlighted four novel lncRNAs (lncRNA CARMN, LINC00607, PCAT19, LINC01235) detected in scRNA-seq datasets and their functions in AS. We also reviewed open web source and bioinformatic tools, as well as the latest methods to perform an in-depth study of lncRNAs. It is fundamental to annotate functional lncRNAs in the various biological activities of AS, as lncRNAs may represent promising targets in the future for treatment and diagnosis in clinical practice.
Collapse
Affiliation(s)
- Zhiyuan Wu
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, PR China
| | - Huarun Yin
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 100730 Beijing, PR China
| | - Yongsheng Guo
- Peking University Health Science Center, 100191 Beijing, PR China
| | - Hongchao Yin
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 100730 Beijing, PR China
| | - Yongjun Li
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, PR China; Peking University Health Science Center, 100191 Beijing, PR China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 100730 Beijing, PR China
| |
Collapse
|