251
|
He L, Palos-Jasso A, Yi Y, Qin M, Qiu L, Yang X, Zhang Y, Yu J. Bioinformatic Analysis Revealed the Essential Regulatory Genes and Pathways of Early and Advanced Atherosclerotic Plaque in Humans. Cells 2022; 11:cells11243976. [PMID: 36552740 PMCID: PMC9776921 DOI: 10.3390/cells11243976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis (AS) is a lipid-induced, chronic inflammatory, autoimmune disease affecting multiple arteries. Although much effort has been put into AS research in the past decades, it is still the leading cause of death worldwide. The complex genetic network regulation underlying the pathogenesis of AS still needs further investigation to provide effective targeted therapy for AS. We performed a bioinformatic microarray data analysis at different atherosclerotic plaque stages from the Gene Expression Omnibus database with accession numbers GSE43292 and GSE28829. Using gene set enrichment analysis, we further confirmed the immune-related pathways that play an important role in the development of AS. We are reporting, for the first time, that the metabolism of the three branched-chain amino acids (BCAAs; leucine, isoleucine, and valine) and short-chain fatty acids (SCFA; propanoate, and butanoate) are involved in the progression of AS using microarray data of atherosclerotic plaque tissue. Immune and muscle system-related pathways were further confirmed as highly regulated pathways during the development of AS using gene expression pattern analysis. Furthermore, we also identified four modules mainly involved in histone modification, immune-related processes, macroautophagy, and B cell activation with modular differential connectivity in the dataset of GSE43292, and three modules related to immune-related processes, B cell activation, and nuclear division in the dataset of GSE28829 also display modular differential connectivity based on the weighted gene co-expression network analysis. Finally, we identified eight key genes related to the pathways of immune and muscle system function as potential therapeutic biomarkers to distinguish patients with early or advanced stages in AS, and two of the eight genes were validated using the gene expression dataset from gene-deficient mice. The results of the current study will improve our understanding of the molecular mechanisms in the progression of AS. The key genes and pathways identified could be potential biomarkers or new drug targets for AS management.
Collapse
Affiliation(s)
- Luling He
- Key Laboratory for Pharmacology and Translational Research of Traditional Chinese Medicine of Nanchang, Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
- Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Nanchang 330006, China
| | - Andrea Palos-Jasso
- Department of Cardiovascular Sciences and Centre for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Yao Yi
- Institute of Gynecology and Obstetrics of traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Manman Qin
- Key Laboratory for Pharmacology and Translational Research of Traditional Chinese Medicine of Nanchang, Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
- Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Nanchang 330006, China
| | - Liang Qiu
- Key Laboratory for Pharmacology and Translational Research of Traditional Chinese Medicine of Nanchang, Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
- Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Nanchang 330006, China
| | - Xiaofeng Yang
- Department of Cardiovascular Sciences and Centre for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Yifeng Zhang
- Key Laboratory for Pharmacology and Translational Research of Traditional Chinese Medicine of Nanchang, Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
- Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Nanchang 330006, China
- Correspondence:
| | - Jun Yu
- Department of Cardiovascular Sciences and Centre for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
252
|
Zhai M, Gong S, Luan P, Shi Y, Kou W, Zeng Y, Shi J, Yu G, Hou J, Yu Q, Jian W, Zhuang J, Feinberg MW, Peng W. Extracellular traps from activated vascular smooth muscle cells drive the progression of atherosclerosis. Nat Commun 2022; 13:7500. [PMID: 36473863 PMCID: PMC9723654 DOI: 10.1038/s41467-022-35330-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular DNA traps (ETs) represent an immune response by which cells release essential materials like chromatin and granular proteins. Previous studies have demonstrated that the transdifferentiation of vascular smooth muscle cells (VSMCs) plays a crucial role in atherosclerosis. This study seeks to investigate the interaction between CD68+ VSMCs and the formation of ETs and highlight its function in atherosclerosis. Here we show that ETs are inhibited, and atherosclerotic plaque formation is alleviated in male Myh11CrePad4flox/flox mice undergoing an adeno-associated-virus-8 (AAV8) mediating overexpression of proprotein convertase subtilisin/kexin type 9 mutation (PCSK9) injection and being challenged with a high-fat diet. Obvious ETs generated from CD68+ VSMCs are inhibited by Cl-amidine and DNase I in vitro. By utilizing VSMCs-lineage tracing technology and single-cell RNA sequencing (scRNA-seq), we demonstrate that the ETs from CD68+ VSMCs influence the progress of atherosclerosis by regulating the direction of VSMCs' transdifferentiation through STING-SOCS1 or TLR4 signaling pathway.
Collapse
Affiliation(s)
- Ming Zhai
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Shiyu Gong
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Peipei Luan
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Yefei Shi
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Wenxin Kou
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Yanxi Zeng
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jiayun Shi
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Guanye Yu
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jiayun Hou
- grid.8547.e0000 0001 0125 2443Biomedical Research Center, Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai, China
| | - Qing Yu
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Weixia Jian
- grid.13402.340000 0004 1759 700XDepartment of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Jianhui Zhuang
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Mark W. Feinberg
- grid.38142.3c000000041936754XCardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Wenhui Peng
- grid.13402.340000 0004 1759 700XDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, China
| |
Collapse
|
253
|
Fasolo F, Paloschi V, Maegdefessel L. Long non-coding RNAs at the crossroad of vascular smooth muscle cell phenotypic modulation in atherosclerosis and neointimal formation. Atherosclerosis 2022:S0021-9150(22)01542-8. [PMID: 36513554 DOI: 10.1016/j.atherosclerosis.2022.11.021] [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: 07/22/2022] [Revised: 11/15/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022]
Abstract
Despite extraordinary advances in the comprehension of the pathophysiology of atherosclerosis and the employment of very effective treatments, cardiovascular diseases are still a major cause of mortality and represent a large share of health expenditure worldwide. Atherosclerosis is a disease affecting the medium and large arteries, which consists of a progressive accumulation of fatty substances, cellular waste products and fibrous elements, which culminates in the buildup of a plaque obstructing the blood flow. Endothelial dysfunction represents an early pathological event, favoring immune cells recruitment and triggering local inflammation. The release of inflammatory cytokines and other signaling molecules stimulates phenotypic modifications in the underlying vascular smooth muscle cells, which, in physiological conditions, are responsible for the maintenance of vessels architecture while regulating vascular tone. Vascular smooth muscle cells are highly plastic and may respond to disease stimuli by de-differentiating and losing their contractility, while increasing their synthetic, proliferative, and migratory capacity. This phenotypic switching is considered a pathological hallmark of atherogenesis and is ruled by the activation of selective gene programs. The advent of genomics and the improvement of sequencing technologies deepened our knowledge of the complex gene expression regulatory networks mediated by non-coding RNAs, and favored the rise of innovative therapeutic approaches targeting the non-coding transcriptome. In the context of atherosclerosis, long non-coding RNAs have received increasing attention as potential translational targets, due to their contribution to the molecular dynamics modulating the expression of vascular smooth muscle cells contractile/synthetic gene programs. In this review, we will focus on the most well-characterized long non-coding RNAs contributing to atherosclerosis by controlling expression of the contractile apparatus and genes activated in perturbed vascular smooth muscle cells.
Collapse
Affiliation(s)
- Francesca Fasolo
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Berlin, Germany.
| | - Valentina Paloschi
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Berlin, Germany
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Berlin, Germany; Molecular Vascular Medicine Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
254
|
Zhang G, Liu Z, Deng J, Liu L, Li Y, Weng S, Guo C, Zhou Z, Zhang L, Wang X, Liu G, Guo J, Bai J, Wang Y, Du Y, Li TS, Tang J, Zhang J. Smooth muscle cell fate decisions decipher a high-resolution heterogeneity within atherosclerosis molecular subtypes. J Transl Med 2022; 20:568. [PMID: 36474294 PMCID: PMC9724432 DOI: 10.1186/s12967-022-03795-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Mounting evidence has revealed the dynamic variations in the cellular status and phenotype of the smooth muscle cell (SMC) are vital for shaping the atherosclerotic plaque microenvironment and ultimately mapping onto heterogeneous clinical outcomes in coronary artery disease. Currently, the underlying clinical significance of SMC evolutions remains unexplored in atherosclerosis. METHODS The dissociated cells from diseased segments within the right coronary artery of four cardiac transplant recipients and 1070 bulk samples with atherosclerosis from six bulk cohorts were retrieved. Following the SMC fate trajectory reconstruction, the MOVICS algorithm integrating the nearest template prediction was used to develop a stable and robust molecular classification. Subsequently, multi-dimensional potential biological implications, molecular features, and cell landscape heterogeneity among distinct clusters were decoded. RESULTS We proposed an SMC cell fate decision signature (SCFDS)-based atherosclerosis stratification system and identified three SCFDS subtypes (C1-C3) with distinguishing features: (i) C1 (DNA-damage repair type), elevated base excision repair (BER), DNA replication, as well as oxidative phosphorylation status. (ii) C2 (immune-activated type), stronger immune activation, hyper-inflammatory state, the complex as well as varied lesion microenvironment, advanced stage, the most severe degree of coronary stenosis severity. (iii) C3 (stromal-rich type), abundant fibrous content, stronger ECM metabolism, immune-suppressed microenvironment. CONCLUSIONS This study uncovered atherosclerosis complex cellular heterogeneity and a differentiated hierarchy of cell populations underlying SMC. The novel high-resolution stratification system could improve clinical outcomes and facilitate individualized management.
Collapse
Affiliation(s)
- Ge Zhang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Zaoqu Liu
- grid.412633.10000 0004 1799 0733Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Jinhai Deng
- grid.13097.3c0000 0001 2322 6764Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King’s College London, London, UK
| | - Long Liu
- grid.412633.10000 0004 1799 0733Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yu Li
- grid.260463.50000 0001 2182 8825Medical College, Nanchang University, Nanchang, 330006 Jiangxi China
| | - Siyuan Weng
- grid.412633.10000 0004 1799 0733Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Chunguang Guo
- grid.412633.10000 0004 1799 0733Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Zhaokai Zhou
- grid.412633.10000 0004 1799 0733Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Li Zhang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Xiaofang Wang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Gangqiong Liu
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Jiacheng Guo
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Jing Bai
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Yunzhe Wang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Youyou Du
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Tao-Sheng Li
- grid.174567.60000 0000 8902 2273Department of Stem Cell Biology, Atomic Bomb Diseases Institute, Nagasaki University, Nagasaki, 852-8523 Japan
| | - Junnan Tang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Jinying Zhang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| |
Collapse
|
255
|
KLF4-PFKFB3-driven glycolysis is essential for phenotypic switching of vascular smooth muscle cells. Commun Biol 2022; 5:1332. [PMID: 36470917 PMCID: PMC9722670 DOI: 10.1038/s42003-022-04302-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/26/2022] [Indexed: 12/12/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) within atherosclerotic lesions undergo a phenotypic switching in a KLF4-dependent manner. Glycolysis plays important roles in transdifferentiation of somatic cells, however, it is unclear whether and how KLF4 mediates the link between glycolytic switch and VSMCs phenotypic transitions. Here, we show that KLF4 upregulation accompanies VSMCs phenotypic switching in atherosclerotic lesions. KLF4 enhances the metabolic switch to glycolysis through increasing PFKFB3 expression. Inhibiting glycolysis suppresses KLF4-induced VSMCs phenotypic switching, demonstrating that glycolytic shift is required for VSMCs phenotypic switching. Mechanistically, KLF4 upregulates expression of circCTDP1 and eEF1A2, both of which cooperatively promote PFKFB3 expression. TMAO induces glycolytic shift and VSMCs phenotypic switching by upregulating KLF4. Our study indicates that KLF4 mediates the link between glycolytic switch and VSMCs phenotypic transitions, suggesting that a previously unrecognized KLF4-eEF1A2/circCTDP1-PFKFB3 axis plays crucial roles in VSMCs phenotypic switching.
Collapse
|
256
|
Chen MY, Zhang ZH, Ke JF, Li TT, Li MF, Lu JX, Li LX. Chaetocin attenuates atherosclerosis progression and inhibits vascular smooth muscle cell phenotype switching. J Cardiovasc Transl Res 2022; 15:1270-1282. [PMID: 35428928 DOI: 10.1007/s12265-022-10258-5] [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: 10/09/2021] [Accepted: 04/06/2022] [Indexed: 12/16/2022]
Abstract
We aimed to explore the effect of chaetocin on atherosclerosis and its possible mechanism. In vitro, we observed that chaetocin treatment significantly inhibited the proliferation of VSMCs in concentration- and time-dependent manner. We also found that chaetocin suppressed the migration of VSMCs. Moreover, chaetocin treatment induced a contractile phenotype in VSMCs by increasing α-SMA and SM22α expression. In addition, chaetocin treatment attenuated the accumulation of H3K9me3 on VSMCs contractile gene promoters, which promoted the expression of α-SMA and SM22α. In vivo, chaetocin treatment decreased the H3K9me3 expression, diminished atherosclerotic plaque formation, and increased plaque stability by decreasing necrotic core area and lipid accumulation and increasing collagen content and contractile VSMC phenotype. We demonstrated a new function of chaetocin in inhibiting atherosclerosis progression and increasing plaque stability partly by inhibiting pathological phenotypic switching of VSMCs. These newly identified roles of chaetocin might provide a novel therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Ming-Yun Chen
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China
- Department of Endocrinology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
| | - Zhi-Hui Zhang
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China
| | - Jiang-Feng Ke
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China
| | - Ting-Ting Li
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China
| | - Mei-Fang Li
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Jun-Xi Lu
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China
| | - Lian-Xi Li
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China.
| |
Collapse
|
257
|
Daoud F, Arévalo Martínez M, Holst J, Holmberg J, Albinsson S, Swärd K. Role of smooth muscle YAP and TAZ in protection against phenotypic modulation, inflammation, and aneurysm development. Biochem Pharmacol 2022; 206:115307. [DOI: 10.1016/j.bcp.2022.115307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 11/02/2022]
|
258
|
Imai T, Van TM, Pasparakis M, Polykratis A. Smooth muscle cell specific NEMO deficiency inhibits atherosclerosis in ApoE−/− mice. Sci Rep 2022; 12:12538. [PMID: 35869246 PMCID: PMC9307802 DOI: 10.1038/s41598-022-16737-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022] Open
Abstract
The development of atherosclerotic plaques is the result of a chronic inflammatory response coordinated by stromal and immune cellular components of the vascular wall. While endothelial cells and leukocytes are well-recognised mediators of inflammation in atherosclerosis, the role of smooth muscle cells (SMCs) remains incompletely understood. Here we aimed to address the role of canonical NF-κB signalling in SMCs in the development of atherosclerosis. We investigated the role of NF-κB signalling in SMCs in atherosclerosis by employing SMC-specific ablation of NEMO, an IKK complex subunit that is essential for canonical NF-κB activation, in ApoE−/− mice. We show that SMC-specific ablation of NEMO (NEMOSMCiKO) inhibited high fat diet induced atherosclerosis in ApoE−/− mice. NEMOSMCiKO/ApoE−/− mice developed less and smaller atherosclerotic plaques, which contained fewer macrophages, decreased numbers of apoptotic cells and smaller necrotic areas and showed reduced inflammation compared to the plaques of ApoE−/− mice. In addition, the plaques of NEMOSMCiKO/ApoE−/− mice showed higher expression of α-SMA and lower expression of the transcriptional factor KLF4 compared to those of ApoE−/− mice. Consistently, in vitro, NEMO-deficient SMCs exhibited reduced proliferation and migration, as well as decreased KLF4 expression and lower production of IL-6 and MCP-1 upon inflammatory stimulus (TNF or LPS) compared to NEMO-expressing SMCs. In conclusion, NEMO-dependent activation of NF-κB signalling in SMCs critically contributes to the pathogenesis of atherosclerosis by regulating SMC proliferation, migration and phenotype switching in response to inflammatory stimuli.
Collapse
|
259
|
Luse MA, Krüger N, Good ME, Biwer LA, Serbulea V, Salamon A, Deaton RA, Leitinger N, Gödecke A, Isakson BE. Smooth muscle cell FTO regulates contractile function. Am J Physiol Heart Circ Physiol 2022; 323:H1212-H1220. [PMID: 36306211 PMCID: PMC9678421 DOI: 10.1152/ajpheart.00427.2022] [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: 08/16/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 12/14/2022]
Abstract
The fat mass and obesity gene (FTO) is a N6-methyladenosine RNA demethylase that was initially linked by Genome-wide association studies to increased rates of obesity. Subsequent studies have revealed multiple mass-independent effects of the gene, including cardiac myocyte contractility. We created a mouse with a conditional and inducible smooth muscle cell deletion of Fto (Myh11 Cre+ Ftofl/fl) and did not observe any changes in mouse body mass or mitochondrial metabolism. However, the mice had significantly decreased blood pressure (hypotensive), despite increased heart rate and sodium, and significantly increased plasma renin. Remarkably, the third-order mesenteric arteries from these mice had almost no myogenic tone or capacity to constrict to smooth muscle depolarization or phenylephrine. Microarray analysis from Fto-/--isolated smooth muscle cells demonstrated a significant decrease in serum response factor (Srf) and the downstream effectors Acta2, Myocd, and Tagln; this was confirmed in cultured human coronary arteries with FTO siRNA. We conclude Fto is an important component to the contractility of smooth muscle cells.NEW & NOTEWORTHY We show a key role for the fat mass obesity (FTO) gene in regulating smooth muscle contractility, possibly by methylation of serum response factor (Srf).
Collapse
Affiliation(s)
- Melissa A Luse
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Nenja Krüger
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Institute of Animal Developmental and Molecular Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Miranda E Good
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Lauren A Biwer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Vlad Serbulea
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Anita Salamon
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Rebecca A Deaton
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Norbert Leitinger
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Axel Gödecke
- Institute of Animal Developmental and Molecular Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
260
|
Shi X, Zhu S, Liu M, Stone SS, Rong Y, Mao K, Xu X, Ma C, Jiang Z, Zha Y, Yan C, Yu X, Wu D, Liu G, Mi J, Zhao J, Li Y, Ding Y, Wang X, Zhang YB, Ji X. Single-Cell RNA-Seq Reveals a Population of Smooth Muscle Cells Responsible for Atherogenesis. Aging Dis 2022; 13:1939-1953. [PMID: 36465170 PMCID: PMC9662277 DOI: 10.14336/ad.2022.0313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 03/13/2022] [Indexed: 01/30/2024] Open
Abstract
Understanding the regional propensity differences of atherosclerosis (AS) development is hindered by the lack of animal models suitable for the study of the disease process. In this paper, we used 3S-ASCVD dogs, an ideal large animal human-like models for AS, to interrogate the heterogeneity of AS-prone and AS-resistant arteries; and at the single-cell level, identify the dominant cells involved in AS development. Here we present data from 3S-ASCVD dogs which reliably mimic human AS pathophysiology, predilection for lesion sites, and endpoint events. Our analysis combined bulk RNA-seq with single-cell RNA-seq to depict the transcriptomic profiles and cellular atlas of AS-prone and AS-resistant arteries in 3S-ASCVD dogs. Our results revealed the integral role of smooth muscle cells (SMCs) in regional propensity for AS. Notably, TNC+ SMCs were major contributors to AS development in 3S-ASCVD dogs, indicating enhanced extracellular matrix remodeling and transition to myofibroblasts during the AS process. Moreover, TNC+ SMCs were also present in human AS-prone carotid plaques, suggesting a potential origin of myofibroblasts and supporting the relevance of our findings. Our study provides a promising large animal model for pre-clinical studies of ASCVD and add novel insights surrounding the regional propensity of AS development in humans, which may lead to interventions that delay or prevent lesion progression and adverse clinical events.
Collapse
Affiliation(s)
- Xiaofeng Shi
- School of Engineering Medicine, Beihang University, Beijing, China.
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.
| | - Shangming Zhu
- School of Engineering Medicine, Beihang University, Beijing, China.
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Meijing Liu
- School of Engineering Medicine, Beihang University, Beijing, China.
| | - Sara Saymuah Stone
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Yao Rong
- School of Engineering Medicine, Beihang University, Beijing, China.
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Ke Mao
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Xiaopeng Xu
- School of Engineering Medicine, Beihang University, Beijing, China.
| | - Chao Ma
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Zhuoyuan Jiang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Yan Zha
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Chun Yan
- School of Engineering Medicine, Beihang University, Beijing, China.
| | - Xiaofan Yu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Guiyou Liu
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.
| | - Jidong Mi
- Beijing SINOGENE Biotechnology Co., Ltd, Beijing, China.
| | - Jianping Zhao
- Beijing SINOGENE Biotechnology Co., Ltd, Beijing, China.
| | - Yuan Li
- Beijing SINOGENE Biotechnology Co., Ltd, Beijing, China.
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Xiaogang Wang
- School of Engineering Medicine, Beihang University, Beijing, China.
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University) Ministry of Industry and Information Technology, Beijing, China.
| | - Yong-Biao Zhang
- School of Engineering Medicine, Beihang University, Beijing, China.
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University) Ministry of Industry and Information Technology, Beijing, China.
| | - Xunming Ji
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
261
|
Reed E, Fellows A, Lu R, Rienks M, Schmidt L, Yin X, Duregotti E, Brandt M, Krasemann S, Hartmann K, Barallobre-Barreiro J, Addison O, Cuello F, Hansen A, Mayr M. Extracellular Matrix Profiling and Disease Modelling in Engineered Vascular Smooth Muscle Cell Tissues. Matrix Biol Plus 2022; 16:100122. [PMID: 36193159 PMCID: PMC9526190 DOI: 10.1016/j.mbplus.2022.100122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/22/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
Aortic smooth muscle cells (SMCs) have an intrinsic role in regulating vessel homeostasis and pathological remodelling. In two-dimensional (2D) cell culture formats, however, SMCs are not embedded in their physiological extracellular matrix (ECM) environment. To overcome the limitations of conventional 2D SMC cultures, we established a 3D in vitro model of engineered vascular smooth muscle cell tissues (EVTs). EVTs were casted from primary murine aortic SMCs by suspending a SMC-fibrin master mix between two flexible silicon-posts at day 0 before prolonged culture up to 14 days. Immunohistochemical analysis of EVT longitudinal sections demonstrated that SMCs were aligned, viable and secretory. Mass spectrometry-based proteomics analysis of murine EVT lysates was performed and identified 135 matrisome proteins. Proteoglycans, including the large aggregating proteoglycan versican, accumulated within EVTs by day 7 of culture. This was followed by the deposition of collagens, elastin-binding proteins and matrix regulators up to day 14 of culture. In contrast to 2D SMC controls, accumulation of versican occurred in parallel to an increase in versikine, a cleavage product mediated by proteases of the A Disintegrin and Metalloproteinase with Thrombospondin motifs (ADAMTS) family. Next, we tested the response of EVTs to stimulation with transforming growth factor beta-1 (TGFβ-1). EVTs contracted in response to TGFβ-1 stimulation with altered ECM composition. In contrast, treatment with the pharmacological activin-like kinase inhibitor (ALKi) SB 431542 suppressed ECM secretion. As a disease stimulus, we performed calcification assays. The ECM acts as a nidus for calcium phosphate deposition in the arterial wall. We compared the onset and extent of calcification in EVTs and 2D SMCs cultured under high calcium and phosphate conditions for 7 days. Calcified EVTs displayed increased tissue stiffness by up to 30 % compared to non-calcified controls. Unlike the rapid calcification of SMCs in 2D cultures, EVTs sustained expression of the calcification inhibitor matrix Gla protein and allowed for better discrimination of the calcification propensity between independent biological replicates. In summary, EVTs are an intuitive and versatile model to investigate ECM synthesis and turnover by SMCs in a 3D environment. Unlike conventional 2D cultures, EVTs provide a more relevant pathophysiological model for retention of the nascent ECM produced by SMCs.
Collapse
Key Words
- 2D, Two-dimensional
- 3D cell culture
- 3D, Three-dimensional
- ADAMTS, A disintegrin and metalloproteinase with thrombospondin motifs
- ALKi, Activin-like kinase inhibitor
- Calcification
- ECM
- ECM, Extracellular matrix
- EHT, Engineered heart tissue
- EVT, Engineered vascular smooth muscle cell tissue
- LC-MS/MS, Liquid chromatography with tandem mass spectrometry
- Proteomics
- SMC, Smooth muscle cell
- Smooth muscle cells
- TCP, Tissue culture polystyrene
- TGFβ-1, Transforming growth factor beta-1
- Tissue engineering
Collapse
Affiliation(s)
- Ella Reed
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Adam Fellows
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Ruifang Lu
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Marieke Rienks
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Lukas Schmidt
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Xiaoke Yin
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Elisa Duregotti
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Mona Brandt
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, University Medical Center Hamburg-Eppendorf, Germany
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Kristin Hartmann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Javier Barallobre-Barreiro
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| | - Owen Addison
- Centre of Oral, Clinical & Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, University Medical Center Hamburg-Eppendorf, Germany
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, University Medical Center Hamburg-Eppendorf, Germany
| | - Manuel Mayr
- King's British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London SE5 9NU, UK
| |
Collapse
|
262
|
Xue Y, Luo M, Hu X, Li X, Shen J, Zhu W, Huang L, Hu Y, Guo Y, Liu L, Wang L, Luo S. Macrophages regulate vascular smooth muscle cell function during atherosclerosis progression through IL-1β/STAT3 signaling. Commun Biol 2022; 5:1316. [PMID: 36456628 PMCID: PMC9715630 DOI: 10.1038/s42003-022-04255-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/13/2022] [Indexed: 12/05/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) play a central role in atherosclerosis progression, but the functional changes in VSMCs and the associated cellular crosstalk during atherosclerosis progression remain unknown. Here we show that scRNA-seq analysis of proximal adjacent (PA) and atherosclerotic core (AC) regions of human carotid artery plaques identifies functional alterations in macrophage-like VSMCs, elucidating the main state differences between PA and AC VSMCs. And, IL-1β mediates macrophage-macrophage-like VSMC crosstalk through regulating key transcription factors involved in macrophage-like VSMCs functional alterations during atherosclerosis progression. In vitro assays reveal VSMCs trans-differentiated into a macrophage-like phenotype and then functional alterations in response to macrophage-derived stimuli. IL-1β promots the adhesion, inflammation, and apoptosis of macrophage-like VSMCs in a STAT3 dependent manner. The current findings provide interesting insight into the macrophages-macrophage-like VSMC crosstalk, which would drive functional alterations in the latter cell type through IL-1β/STAT3 axis during atherosclerosis progression.
Collapse
Affiliation(s)
- Yuzhou Xue
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Minghao Luo
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiankang Hu
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang Li
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Shen
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenyan Zhu
- Medical Department, Yidu Cloud (Beijing) Technology Co., Ltd., Beijing, China
- Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Longxiang Huang
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Hu
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yongzheng Guo
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Liu
- Department of Dermatology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingbang Wang
- Department of Orthopedic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Suxin Luo
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
263
|
The Role of Hydrogen Sulfide in Plaque Stability. Antioxidants (Basel) 2022; 11:antiox11122356. [PMID: 36552564 PMCID: PMC9774534 DOI: 10.3390/antiox11122356] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/11/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Atherosclerosis is the greatest contributor to cardiovascular events and is involved in the majority of deaths worldwide. Plaque rapture or erosion precipitates life-threatening thrombi, resulting in the obstruction blood flow to the heart (acute coronary syndrome), brain (ischemic stroke) or low extremities (peripheral vascular diseases). Among these events, major causation dues to the plaque rupture. Although the initiation, procession, and precise time of controlling plaque rupture are unclear, foam cell formation and apoptosis, cell death, extracellular matrix components, protease expression and activity, local inflammation, intraplaque hemorrhage, and calcification contribute to the plaque instability. These alterations tightly associate with the function regulation of intraplaque various cell populations. Hydrogen sulfide (H2S) is gasotransmitter derived from methionine metabolism and exerts a protective role in the genesis of atherosclerosis. Recent progress also showed H2S mediated the plaque stability. In this review, we discuss the progress of endogenous H2S modulation on functions of vascular smooth muscle cells, monocytes/macrophages, and T cells, and the molecular mechanism in plaque stability.
Collapse
|
264
|
Cao G, Xuan X, Hu J, Zhang R, Jin H, Dong H. How vascular smooth muscle cell phenotype switching contributes to vascular disease. Cell Commun Signal 2022; 20:180. [PMID: 36411459 PMCID: PMC9677683 DOI: 10.1186/s12964-022-00993-2] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/22/2022] [Indexed: 11/22/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the most abundant cell in vessels. Earlier experiments have found that VSMCs possess high plasticity. Vascular injury stimulates VSMCs to switch into a dedifferentiated type, also known as synthetic VSMCs, with a high migration and proliferation capacity for repairing vascular injury. In recent years, largely owing to rapid technological advances in single-cell sequencing and cell-lineage tracing techniques, multiple VSMCs phenotypes have been uncovered in vascular aging, atherosclerosis (AS), aortic aneurysm (AA), etc. These VSMCs all down-regulate contractile proteins such as α-SMA and calponin1, and obtain specific markers and similar cellular functions of osteoblast, fibroblast, macrophage, and mesenchymal cells. This highly plastic phenotype transformation is regulated by a complex network consisting of circulating plasma substances, transcription factors, growth factors, inflammatory factors, non-coding RNAs, integrin family, and Notch pathway. This review focuses on phenotypic characteristics, molecular profile and the functional role of VSMCs phenotype landscape; the molecular mechanism regulating VSMCs phenotype switching; and the contribution of VSMCs phenotype switching to vascular aging, AS, and AA. Video Abstract.
Collapse
Affiliation(s)
- Genmao Cao
- grid.452845.a0000 0004 1799 2077Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Xuezhen Xuan
- grid.452845.a0000 0004 1799 2077Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Jie Hu
- grid.452845.a0000 0004 1799 2077Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Ruijing Zhang
- grid.452845.a0000 0004 1799 2077Department of Nephrology, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Haijiang Jin
- grid.452845.a0000 0004 1799 2077Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Honglin Dong
- grid.452845.a0000 0004 1799 2077Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| |
Collapse
|
265
|
Macrophage-, Dendritic-, Smooth Muscle-, Endothelium-, and Stem Cells-Derived Foam Cells in Atherosclerosis. Int J Mol Sci 2022; 23:ijms232214154. [PMID: 36430636 PMCID: PMC9695208 DOI: 10.3390/ijms232214154] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Atherosclerosis is an inflammatory disease depending on the buildup, called plaque, of lipoproteins, cholesterol, extracellular matrix elements, and various types of immune and non-immune cells on the artery walls. Plaque development and growth lead to the narrowing of the blood vessel lumen, blocking blood flow, and eventually may lead to plaque burst and a blood clot. The prominent cellular components of atherosclerotic plaque are the foam cells, which, by trying to remove lipoprotein and cholesterol surplus, also participate in plaque development and rupture. Although the common knowledge is that the foam cells derive from macrophages, studies of the last decade clearly showed that macrophages are not the only cells able to form foam cells in atherosclerotic plaque. These findings give a new perspective on atherosclerotic plaque formation and composition and define new targets for anti-foam cell therapies for atherosclerosis prevention. This review gives a concise description of foam cells of different pedigrees and describes the main mechanisms participating in their formation and function.
Collapse
|
266
|
Lycopene Scavenges Cellular ROS, Modulates Autophagy and Improves Survival through 7SK snRNA Interaction in Smooth Muscle Cells. Cells 2022; 11:cells11223617. [PMID: 36429045 PMCID: PMC9688495 DOI: 10.3390/cells11223617] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
The chance of survival rate and autophagy of smooth muscle cells under calcium stress were drastically improved with a prolonged inclusion of Lycopene in the media. The results showed an improved viability from 41% to 69% and a reduction in overall autophagic bodies from 7% to 3%, which was well in agreement with the LC3II and III mRNA levels. However, the proliferation was slow compared to the controls. The fall in the major inflammatory marker TNF-α and improved antioxidant enzyme GPx were regarded as significant restoration markers of cell survival. The reactive oxygen species (ROS) were reduced from 8 fold to 3 fold post addition of lycopene for 24 h. Further, the docking studies revealed binding of lycopene molecules with 7SK snRNA at 7.6 kcal/mol docking energy with 300 ns stability under physiological conditions. Together, these results suggest that Lycopene administration during ischemic heart disease might improve the functions of the smooth muscle cells and 7SK snRNA might be involved in the binding of lycopene and its antioxidant protective effects.
Collapse
|
267
|
Han Y, Yan L, Xia L, Li S, Zhang Q, jin C. Global trends and Frontier topics about vascular smooth muscle cells phenotype switch: A bibliometric analysis from 1999 to 2021. Front Pharmacol 2022; 13:1004525. [PMID: 36452231 PMCID: PMC9702355 DOI: 10.3389/fphar.2022.1004525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/02/2022] [Indexed: 03/31/2024] Open
Abstract
Objective: Vascular smooth muscle cell phenotype switch (VSMCPS) plays a significant role in vascular remodeling. This study aimed to conduct a bibliometric analysis and visualize the knowledge map of research on VSMCPS. Methods: We retrieved publications focusing on VSMCPS from the Web of Science Core Collection database (SCI-EXPANDED) from 1999 to 2021. Using bibliometric tools, VOSviewer and CiteSpace, we identified the most productive researchers, journals, institutions, and countries. At the same time, the trends, hot topics, and knowledge networks were analyzed and visualized. Results: A total of 2213 publications were included in this analysis. The number of annual publications in the VSMCPS field exhibited an upward trend and could be roughly divided into three phases. Until 2006, the most prolific authors were from the United States. As of 2008, the number of articles published in China increased dramatically to reach 126 papers in 2020. As of 2014, China was the most productive country in this field. The United States ranked first in the number of highly-influential authors, institutions, and literature from 1999 to 2022. Owens GK, Hata, Akiko, and Wen, jin-kun were the most prolific authors. Arteriosclerosis Thrombosis and Vascular Biology, Circulation Research, and Cardiovascular Research were the top-ranked journals in this field. "Vascular remodeling," "atherosclerosis," "neointima," "hypertension", and "inflammation" were the main researched topics. New diseases, new mechanisms, and new phenotype (e.g., micro RNA, macrophage-like-cell, hypoxia, autophagy, long noncoding RNA, oxidative stress, endoplasmic reticulum stress, senescence, aging, abdominal aortic aneurysm, and aortic dissection) represent the trending topics in recent years. Conclusion: This study systematically analyzed and visualized the knowledge map of VSMCPS over the past 2 decades. Our findings provide a comprehensive overview for scholars who want to understand current trends and new research frontiers in this area.
Collapse
Affiliation(s)
- Ying Han
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Langchao Yan
- Mini-invasive Neurosurgery and Translational Medical Center, Xi’an Central Hospital, Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Xia
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Shifu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chen jin
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
268
|
Volpini X, Natali L, Brugo MB, de la Cruz-Thea B, Baigorri RE, Cerbán FM, Fozzatti L, Motran CC, Musri MM. Trypanosoma cruzi Infection Promotes Vascular Remodeling and Coexpression of α-Smooth Muscle Actin and Macrophage Markers in Cells of the Aorta. ACS Infect Dis 2022; 8:2271-2290. [PMID: 36083791 DOI: 10.1021/acsinfecdis.2c00318] [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: 01/29/2023]
Abstract
Chagas disease is an emerging global health problem; however, it remains neglected. Increased aortic stiffness (IAS), a predictor of cardiovascular events, has recently been reported in asymptomatic chronic Chagas patients. After vascular injury, smooth muscle cells (SMCs) can undergo alterations associated with phenotypic switch and transdifferentiation, promoting vascular remodeling and IAS. By studying different mouse aortic segments, we tested the hypothesis that Trypanosoma cruzi infection promotes vascular remodeling. Interestingly, the thoracic aorta was the most affected by the infection. Decreased expression of SMC markers and increased expression of proliferative markers were observed in the arteries of acutely infected mice. In acutely and chronically infected mice, we observed cells coexpressing SMC and macrophage (Mo) markers in the media and adventitia layers of the aorta, indicating that T. cruzi might induce cellular processes associated with SMC transdifferentiation into Mo-like cells or vice versa. In the adventitia, the Mo cell functional polarization was associated with an M2-like CD206+arginase-1+ phenotype despite the T. cruzi presence in the tissue. Only Mo-like cells in inflammatory foci were CD206+iNOS+. In addition to the disorganization of elastic fibers, we found thickening of the aortic layers during the acute and chronic phases of the disease. Our findings indicate that T. cruzi infection induces a vascular remodeling with SMC dedifferentiation and increased cell populations coexpressing α-SMA and Mo markers that could be associated with IAS promotion. These data highlight the importance of studying large vessel homeostasis in Chagas disease.
Collapse
Affiliation(s)
- Ximena Volpini
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Lautaro Natali
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Maria Belén Brugo
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Benjamin de la Cruz-Thea
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina
| | - Ruth Eliana Baigorri
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Fabio Marcelo Cerbán
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Laura Fozzatti
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Claudia Cristina Motran
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Melina Mara Musri
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina.,Departamento de Fisiología, Facultad de Ciencias Exactas Físicas y Naturales. Universidad Nacional de Córdoba (FCEFyN-UNC). Av. Velez Sarfield 299, Centro, Córdoba, PC X5000JJC, Argentina
| |
Collapse
|
269
|
Barnhart MD, Yang Y, Nakagaki-Silva EE, Hammond TH, Pizzinga M, Gooding C, Stott K, Smith CWJ. Phosphorylation of the smooth muscle master splicing regulator RBPMS regulates its splicing activity. Nucleic Acids Res 2022; 50:11895-11915. [PMID: 36408906 PMCID: PMC9723635 DOI: 10.1093/nar/gkac1048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/12/2022] [Accepted: 10/24/2022] [Indexed: 11/22/2022] Open
Abstract
We previously identified RBPMS as a master regulator of alternative splicing in differentiated smooth muscle cells (SMCs). RBPMS is transcriptionally downregulated during SMC dedifferentiation, but we hypothesized that RBPMS protein activity might be acutely downregulated by post-translational modifications. Publicly available phosphoproteomic datasets reveal that Thr113 and Thr118 immediately adjacent to the RRM domain are commonly both phosphorylated. An RBPMS T113/118 phosphomimetic T/E mutant showed decreased splicing regulatory activity both in transfected cells and in a cell-free in vitro assay, while a non-phosphorylatable T/A mutant retained full activity. Loss of splicing activity was associated with a modest reduction in RNA affinity but significantly reduced RNA binding in nuclear extract. A lower degree of oligomerization of the T/E mutant might cause lower avidity of multivalent RNA binding. However, NMR analysis also revealed that the T113/118E peptide acts as an RNA mimic which can loop back and antagonize RNA-binding by the RRM domain. Finally, we identified ERK2 as the most likely kinase responsible for phosphorylation at Thr113 and Thr118. Collectively, our data identify a potential mechanism for rapid modulation of the SMC splicing program in response to external signals during the vascular injury response and atherogenesis.
Collapse
Affiliation(s)
- Michael D Barnhart
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Yi Yang
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | | | - Thomas H Hammond
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | | | - Clare Gooding
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Katherine Stott
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | | |
Collapse
|
270
|
Rangarajan S, Orujyan D, Rangchaikul P, Radwan MM. Critical Role of Inflammation and Specialized Pro-Resolving Mediators in the Pathogenesis of Atherosclerosis. Biomedicines 2022; 10:2829. [PMID: 36359349 PMCID: PMC9687471 DOI: 10.3390/biomedicines10112829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2023] Open
Abstract
Recent research on how the body resolves this inflammation is gaining traction and has shed light on new avenues for future management of cardiovascular diseases. In this narrative review, we discuss the pathophysiological mechanisms of atherosclerosis, the recent development in the understanding of a new class of molecules called Specialized Pro-resolving Mediators (SPMs), and the impact of such findings in the realm of cardiovascular treatment options. We searched the MEDLINE database restricting ourselves to original research articles as much as possible on the complex pathophysiology of atherosclerosis and the role of SPMs. We expect to see further research in translating these findings to bedside clinical trials in treating conditions with a pathophysiological basis of inflammation, such as coronary artery disease, asthma, and periodontal disease.
Collapse
Affiliation(s)
- Subhapradha Rangarajan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Davit Orujyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Patrida Rangchaikul
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Mohamed M. Radwan
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
271
|
Rickel AP, Sanyour HJ, Kinser C, Khatiwada N, Vogel H, Hong Z. Exploring the difference in the mechanics of vascular smooth muscle cells from wild-type and apolipoprotein-E knockout mice. Am J Physiol Cell Physiol 2022; 323:C1393-C1401. [PMID: 36121132 PMCID: PMC9602701 DOI: 10.1152/ajpcell.00046.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 08/30/2022] [Accepted: 09/14/2022] [Indexed: 11/22/2022]
Abstract
Atherosclerosis-related cardiovascular diseases are a leading cause of mortality worldwide. Vascular smooth muscle cells (VSMCs) comprise the medial layer of the arterial wall and undergo phenotypic switching during atherosclerosis to a synthetic phenotype capable of proliferation and migration. The surrounding environment undergoes alterations in extracellular matrix (ECM) stiffness and composition and an increase in cholesterol content. Using an atherosclerotic murine model, we analyzed how the mechanics of VSMCs isolated from Western diet-fed apolipoprotein-E knockout (ApoE-/-) and wild-type (WT) mice were altered during atherosclerosis. Increased stiffness of ApoE-/- VSMCs correlated with a greater degree of stress fiber alignment, as evidenced by atomic force microscopy (AFM)-generated force maps and stress fiber topography images. On type-1 collagen (COL1)-coated polyacrylamide (PA) gels (referred to as substrate) of varying stiffness, ApoE-/- VSMCs had lower adhesion forces to COL1 and N-cadherin (N-Cad) compared with WT cells. ApoE-/- VSMC stiffness was significantly greater than that of WT cells. Cell stiffness increased with increasing substrate stiffness for both ApoE-/- and WT VSMCs. In addition, ApoE-/- VSMCs showed an enhanced migration capability on COL1-coated substrates and a general decreasing trend in migration capacity with increasing substrate stiffness, correlating with lowered adhesion forces as compared with WT VSMCs. Altogether, these results demonstrate the potential contribution of the alteration in VSMC mechanics in the development of atherosclerosis.
Collapse
Affiliation(s)
- Alex P Rickel
- Biomedical Engineering Department, University of South Dakota, Sioux Falls, South Dakota
| | - Hanna J Sanyour
- Biomedical Engineering Department, University of South Dakota, Sioux Falls, South Dakota
| | - Courtney Kinser
- Biomedical Engineering Department, University of South Dakota, Sioux Falls, South Dakota
| | - Nisha Khatiwada
- Biomedical Engineering Department, University of South Dakota, Sioux Falls, South Dakota
- Mechanical Engineering Department, Texas Tech University, Lubbock, Texas
| | - Hayley Vogel
- Biomedical Engineering Department, University of South Dakota, Sioux Falls, South Dakota
| | - Zhongkui Hong
- Biomedical Engineering Department, University of South Dakota, Sioux Falls, South Dakota
- Mechanical Engineering Department, Texas Tech University, Lubbock, Texas
| |
Collapse
|
272
|
Chou EL, Chaffin M, Simonson B, Pirruccello JP, Akkad AD, Nekoui M, Cardenas CLL, Bedi KC, Nash C, Juric D, Stone JR, Isselbacher EM, Margulies KB, Klattenhoff C, Ellinor PT, Lindsay ME. Aortic Cellular Diversity and Quantitative Genome-Wide Association Study Trait Prioritization Through Single-Nuclear RNA Sequencing of the Aneurysmal Human Aorta. Arterioscler Thromb Vasc Biol 2022; 42:1355-1374. [PMID: 36172868 PMCID: PMC9613617 DOI: 10.1161/atvbaha.122.317953] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 09/16/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Mural cells in ascending aortic aneurysms undergo phenotypic changes that promote extracellular matrix destruction and structural weakening. To explore this biology, we analyzed the transcriptional features of thoracic aortic tissue. METHODS Single-nuclear RNA sequencing was performed on 13 samples from human donors, 6 with thoracic aortic aneurysm, and 7 without aneurysm. Individual transcriptomes were then clustered based on transcriptional profiles. Clusters were used for between-disease differential gene expression analyses, subcluster analysis, and analyzed for intersection with genetic aortic trait data. RESULTS We sequenced 71 689 nuclei from human thoracic aortas and identified 14 clusters, aligning with 11 cell types, predominantly vascular smooth muscle cells (VSMCs) consistent with aortic histology. With unbiased methodology, we found 7 vascular smooth muscle cell and 6 fibroblast subclusters. Differentially expressed genes analysis revealed a vascular smooth muscle cell group accounting for the majority of differential gene expression. Fibroblast populations in aneurysm exhibit distinct behavior with almost complete disappearance of quiescent fibroblasts. Differentially expressed genes were used to prioritize genes at aortic diameter and distensibility genome-wide association study loci highlighting the genes JUN, LTBP4 (latent transforming growth factor beta-binding protein 1), and IL34 (interleukin 34) in fibroblasts, ENTPD1, PDLIM5 (PDZ and LIM domain 5), ACTN4 (alpha-actinin-4), and GLRX in vascular smooth muscle cells, as well as LRP1 in macrophage populations. CONCLUSIONS Using nuclear RNA sequencing, we describe the cellular diversity of healthy and aneurysmal human ascending aorta. Sporadic aortic aneurysm is characterized by differential gene expression within known cellular classes rather than by the appearance of novel cellular forms. Single-nuclear RNA sequencing of aortic tissue can be used to prioritize genes at aortic trait loci.
Collapse
Affiliation(s)
- Elizabeth L. Chou
- Division of Vascular and Endovascular Surgery,
Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
| | - Mark Chaffin
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Precision Cardiology Laboratory, The Broad Institute,
Cambridge, MA, USA 02142
| | - Bridget Simonson
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Precision Cardiology Laboratory, The Broad Institute,
Cambridge, MA, USA 02142
| | - James P. Pirruccello
- Cardiology Division, Massachusetts General Hospital,
Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Precision Cardiology Laboratory, The Broad Institute,
Cambridge, MA, USA 02142
- Demoulas Center for Cardiac Arrhythmias, Massachusetts
General Hospital, Boston, Massachusetts, USA
| | - Amer-Denis Akkad
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge,
MA, USA 02142
| | - Mahan Nekoui
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts
General Hospital, Boston, Massachusetts, USA
| | - Christian Lacks Lino Cardenas
- Cardiology Division, Massachusetts General Hospital,
Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
| | - Kenneth C. Bedi
- Perelman School of Medicine, University of Pennsylvania,
Philadelphia, PA, USA 19104
| | - Craig Nash
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Precision Cardiology Laboratory, The Broad Institute,
Cambridge, MA, USA 02142
| | - Dejan Juric
- Cancer Center, Massachusetts General Hospital, Boston,
Massachusetts, USA
| | - James R. Stone
- Department of Pathology, Massachusetts General
Hospital, Boston, Massachusetts, USA
| | - Eric M. Isselbacher
- Cardiology Division, Massachusetts General Hospital,
Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
- Thoracic Aortic Center, Massachusetts General Hospital,
Boston, Massachusetts, USA
| | - Kenneth B. Margulies
- Perelman School of Medicine, University of Pennsylvania,
Philadelphia, PA, USA 19104
| | - Carla Klattenhoff
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge,
MA, USA 02142
| | - Patrick T. Ellinor
- Cardiology Division, Massachusetts General Hospital,
Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Precision Cardiology Laboratory, The Broad Institute,
Cambridge, MA, USA 02142
- Demoulas Center for Cardiac Arrhythmias, Massachusetts
General Hospital, Boston, Massachusetts, USA
| | - Mark E. Lindsay
- Cardiology Division, Massachusetts General Hospital,
Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General
Hospital, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute,
Cambridge, Massachusetts, USA
- Thoracic Aortic Center, Massachusetts General Hospital,
Boston, Massachusetts, USA
| |
Collapse
|
273
|
Ding X, An Q, Zhao W, Song Y, Tang X, Wang J, Chang CC, Zhao G, Hsiai T, Fan G, Fan Y, Li S. Distinct patterns of responses in endothelial cells and smooth muscle cells following vascular injury. JCI Insight 2022; 7:e153769. [PMID: 36278486 PMCID: PMC9714785 DOI: 10.1172/jci.insight.153769] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/02/2022] [Indexed: 01/27/2025] Open
Abstract
Vascular smooth muscle cells (SMCs) are heterogeneous, and their differential responses to vascular injury are not well understood. To address this question, we performed single-cell analysis of vascular cells to a ligation injury in mouse carotid arteries after 3 days. While endothelial cells had a homogeneous activation of mesenchymal genes, less than 30% of SMCs responded to the injury and generated 2 distinct clusters - i.e., proinflammatory SMCs and stress-responsive SMCs. Proinflammatory SMCs were enriched with high levels of inflammatory markers such as vascular cell adhesion molecule-1 while stress-responsive SMCs overexpressed heat shock proteins. Trajectory analysis suggested that proinflammatory SMCs were potentially derived from a specific subpopulation of SMCs. Ligand-receptor pair analysis showed that the interaction between macrophages and proinflammatory SMCs was the major cell-cell communication among all cell types in the injured arteries. In vitro coculture demonstrated that VCAM1+ SMCs had a stronger chemotactic effect on macrophage recruitment than VCAM1- SMCs. Consistently, the number of VCAM1+ SMCs significantly increased in injured arteries and atherosclerotic lesions of ApoE-/- mice and human arteries. These findings provide insights at the single-cell level on the distinct patterns of endothelial cells and SMC responses to vascular injury.
Collapse
Affiliation(s)
- Xili Ding
- Department of Bioengineering, University of California, Los Angeles, California, USA
- School of Engineering Medicine and
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Qin An
- Department of Human Genetics, David Geffen School of Medicine
| | - Weikang Zhao
- Department of Bioengineering, University of California, Los Angeles, California, USA
| | - Yang Song
- Department of Bioengineering, University of California, Los Angeles, California, USA
| | - Xiaokai Tang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jing Wang
- Department of Human Genetics, David Geffen School of Medicine
| | - Chih-Chiang Chang
- Department of Bioengineering, University of California, Los Angeles, California, USA
| | - Gexin Zhao
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, and
| | - Tzung Hsiai
- Department of Bioengineering, University of California, Los Angeles, California, USA
- Department of Medicine, University of California, Los Angeles, California, USA
| | - Guoping Fan
- Department of Human Genetics, David Geffen School of Medicine
| | - Yubo Fan
- School of Engineering Medicine and
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, California, USA
- Department of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
274
|
Lu CW, Nguyen NTK, Shen SC, Wu YB, Liang HJ, Wu CH. Botanical Antcin K Alleviates High-Fat Damage in Palm Acid Oil-Treated Vascular Endothelial Cells and Macrophages. PLANTS (BASEL, SWITZERLAND) 2022; 11:2812. [PMID: 36365265 PMCID: PMC9656957 DOI: 10.3390/plants11212812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 06/16/2023]
Abstract
Lipid metabolism disorder is the most critical risk factor for atherosclerosis, and the process involves lipid deposition in the arterial intima. In Taiwan, antcin K, an active triterpenoid from the fruiting bodies of Antrodia camphorata, has been considered a potential lipid-lowering agent. Despite this, the possible therapeutic mechanisms of antcin K remain unclear. To explore the crucial role of botanical antcin K in reducing atherosclerotic plaque, we used SVEC4-10 vascular endothelial cells and RAW264.7 macrophages with palm acid oil-induced high-fat damage as our cell models. Our results showed through using the DPPH assay that antcin K had excellent free radical scavenging ability. Antcin K treatment can significantly alleviate the high-fat damage and reduce the levels of inflammatory factors of TNF-α and IL-1β in vascular endothelial cells and macrophages, as shown through MTT assay and ELISA. Furthermore, antcin K treatment can effectively enhance migration ability and clear lipid deposition in macrophages, as shown by using cell migration assay and oil red O staining. When stained with immunofluorescence, antcin K was shown to significantly decrease the expression of adhesion molecules of vascular cell adhesion molecule 1 (VCAM-1) in vascular endothelial cells involved in monocyte migration and inflammation. Antcin K not only reduced the expression of the CD36 scavenger receptor but also augmented the expression of Kruppel-like factor 4 (KLF4) transcription factor in macrophages, which inhibits the transformation of macrophages into foam cells underlying the pathological process of atherosclerosis. Taking our findings into account, we suggested that botanical antcin K could have therapeutic potential for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Chen-Wen Lu
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Ngan Thi Kim Nguyen
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Szu-Chuan Shen
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Yeh-Bin Wu
- ARJIL Pharmaceuticals LLC, Hsinchu City 30013, Taiwan
| | - Hui-Ju Liang
- ARJIL Pharmaceuticals LLC, Hsinchu City 30013, Taiwan
| | - Chung-Hsin Wu
- School of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| |
Collapse
|
275
|
Wang Y, Gao H, Wang F, Ye Z, Mokry M, Turner AW, Ye J, Koplev S, Luo L, Alsaigh T, Adkar SS, Elishaev M, Gao X, Maegdefessel L, Björkegren JLM, Pasterkamp G, Miller CL, Ross EG, Leeper NJ. Dynamic changes in chromatin accessibility are associated with the atherogenic transitioning of vascular smooth muscle cells. Cardiovasc Res 2022; 118:2792-2804. [PMID: 34849613 PMCID: PMC9586565 DOI: 10.1093/cvr/cvab347] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
AIMS De-differentiation and activation of pro-inflammatory pathways are key transitions vascular smooth muscle cells (SMCs) make during atherogenesis. Here, we explored the upstream regulators of this 'atherogenic transition'. METHODS AND RESULTS Genome-wide sequencing studies, including Assay for Transposase-Accessible Chromatin using sequencing and RNA-seq, were performed on cells isolated from both murine SMC-lineage-tracing models of atherosclerosis and human atherosclerotic lesions. At the bulk level, alterations in chromatin accessibility were associated with the atherogenic transitioning of lesional SMCs, especially in relation to genes that govern differentiation status and complement-dependent inflammation. Using computational biology, we observed that a transcription factor previously related to coronary artery disease, Activating transcription factor 3 (ATF3), was predicted to be an upstream regulator of genes altered during the transition. At the single-cell level, our results indicated that ATF3 is a key repressor of SMC transitioning towards the subset of cells that promote vascular inflammation by activating the complement cascade. The expression of ATF3 and complement component C3 was negatively correlated in SMCs from human atherosclerotic lesions, suggesting translational relevance. Phenome-wide association studies indicated that genetic variation that results in reduced expression of ATF3 is correlated with an increased risk for atherosclerosis, and the expression of ATF3 was significantly down-regulated in humans with advanced vascular disease. CONCLUSION Our study indicates that the plasticity of atherosclerotic SMCs may in part be explained by dynamic changes in their chromatin architecture, which in turn may contribute to their maladaptive response to inflammation-induced stress.
Collapse
Affiliation(s)
- Ying Wang
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur drive, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Hua Gao
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur drive, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Fudi Wang
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur drive, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Zhongde Ye
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur drive, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Michal Mokry
- Department of Cardiology and Laboratory of Clinical Chemistry, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands
| | - Adam W Turner
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, 1335 Lee St, Charlottesville, VA 22908, USA
| | - Jianqin Ye
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur drive, Stanford, CA 94305, USA
| | - Simon Koplev
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - Lingfeng Luo
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur drive, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Tom Alsaigh
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur drive, Stanford, CA 94305, USA
- Department of Cardiovascular Medicine, Stanford University School of Medicine, 870 Quarry Road Extension, Stanford, CA 94305, USA
| | - Shaunak S Adkar
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur drive, Stanford, CA 94305, USA
| | - Maria Elishaev
- Department of Pathology and Laboratory Medicine, Centre for Heart Lung Innvoation, University of British Columbia, 166-1081 Burrard St, Vancouver, BC V6Z 1Y6, Canada
| | - Xiangyu Gao
- Department of Pathology and Laboratory Medicine, Centre for Heart Lung Innvoation, University of British Columbia, 166-1081 Burrard St, Vancouver, BC V6Z 1Y6, Canada
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, and the German Center for Cardiovascular Research (DZHK partner site), Biedersteiner Str. 29, Munich 80802, Germany
- Department of Internal Medicine, Center for Molecular Medicine, Karolinska Institute, Visionsgatan 18, Stockholm 171 76, Sweden
| | - Johan L M Björkegren
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY 10029, USA
| | - Gerard Pasterkamp
- Department of Cardiology and Laboratory of Clinical Chemistry, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands
| | - Clint L Miller
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, 1335 Lee St, Charlottesville, VA 22908, USA
| | - Elsie G Ross
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur drive, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Nicholas J Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur drive, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| |
Collapse
|
276
|
Decoding the transcriptome of calcified atherosclerotic plaque at single-cell resolution. Commun Biol 2022; 5:1084. [PMID: 36224302 PMCID: PMC9556750 DOI: 10.1038/s42003-022-04056-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 09/30/2022] [Indexed: 11/30/2022] Open
Abstract
Atherogenesis involves an interplay of inflammation, tissue remodeling and cellular transdifferentiation (CTD), making it especially difficult to precisely delineate its pathophysiology. Here we use single-cell RNA sequencing and systems-biology approaches to analyze the transcriptional profiles of vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) in calcified atherosclerotic core (AC) plaques and patient-matched proximal adjacent (PA) portions of carotid artery tissue from patients undergoing carotid endarterectomy. Our results reveal an anatomic distinction whereby PA cells express inflammatory mediators, while cells expressing matrix-secreting genes occupy a majority of the AC region. Systems biology analysis indicates that inflammation in PA ECs and VSMCs may be driven by TNFa signaling. Furthermore, we identify POSTN, SPP1 and IBSP in AC VSMCs, and ITLN1, SCX and S100A4 in AC ECs as possible candidate drivers of CTD in the atherosclerotic core. These results establish an anatomic framework for atherogenesis which forms the basis for exploration of a site-specific strategy for disruption of disease progression. Single-cell RNA sequencing and systems biology are used to profile the human vascular cell populations in calcified atherosclerotic core plaques from carotid endarterectomy samples, showing an anatomic distinction between gene expression of inflammatory versus matrix-secreting factors.
Collapse
|
277
|
Lu Y, Sun Y, Xu K, Shao Y, Saaoud F, Snyder NW, Yang L, Yu J, Wu S, Hu W, Sun J, Wang H, Yang X. Editorial: Endothelial cells as innate immune cells. Front Immunol 2022; 13:1035497. [PMID: 36268030 PMCID: PMC9577408 DOI: 10.3389/fimmu.2022.1035497] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yifan Lu
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Yu Sun
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Keman Xu
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Ying Shao
- Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Fatma Saaoud
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Nathaniel W. Snyder
- Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Ling Yang
- Department of Medical Genetics and Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Jun Yu
- Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Sheng Wu
- Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Wenhui Hu
- Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Jianxin Sun
- Center for Translational Medicine, Department of Medicine, Simmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Hong Wang
- Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
278
|
Wang J, Kang Z, Liu Y, Li Z, Liu Y, Liu J. Identification of immune cell infiltration and diagnostic biomarkers in unstable atherosclerotic plaques by integrated bioinformatics analysis and machine learning. Front Immunol 2022; 13:956078. [PMID: 36211422 PMCID: PMC9537477 DOI: 10.3389/fimmu.2022.956078] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/02/2022] [Indexed: 12/04/2022] Open
Abstract
Objective The decreased stability of atherosclerotic plaques increases the risk of ischemic stroke. However, the specific characteristics of dysregulated immune cells and effective diagnostic biomarkers associated with stability in atherosclerotic plaques are poorly characterized. This research aims to investigate the role of immune cells and explore diagnostic biomarkers in the formation of unstable plaques for the sake of gaining new insights into the underlying molecular mechanisms and providing new perspectives for disease detection and therapy. Method Using the CIBERSORT method, 22 types of immune cells between stable and unstable carotid atherosclerotic plaques from RNA-sequencing and microarray data in the public GEO database were quantitated. Differentially expressed genes (DEGs) were further calculated and were analyzed for enrichment of GO Biological Process and KEGG pathways. Important cell types and hub genes were screened using machine learning methods including least absolute shrinkage and selection operator (LASSO) regression and random forest. Single-cell RNA sequencing and clinical samples were further used to validate critical cell types and hub genes. Finally, the DGIdb database of gene–drug interaction data was utilized to find possible therapeutic medicines and show how pharmaceuticals, genes, and immune cells interacted. Results A significant difference in immune cell infiltration was observed between unstable and stable plaques. The proportions of M0, M1, and M2 macrophages were significantly higher and that of CD8+ T cells and NK cells were significantly lower in unstable plaques than that in stable plaques. With respect to DEGs, antigen presentation genes (CD74, B2M, and HLA-DRA), inflammation-related genes (MMP9, CTSL, and IFI30), and fatty acid-binding proteins (CD36 and APOE) were elevated in unstable plaques, while the expression of smooth muscle contraction genes (TAGLN, ACAT2, MYH10, and MYH11) was decreased in unstable plaques. M1 macrophages had the highest instability score and contributed to atherosclerotic plaque instability. CD68, PAM, and IGFBP6 genes were identified as the effective diagnostic markers of unstable plaques, which were validated by validation datasets and clinical samples. In addition, insulin, nivolumab, indomethacin, and α-mangostin were predicted to be potential therapeutic agents for unstable plaques. Conclusion M1 macrophages is an important cause of unstable plaque formation, and CD68, PAM, and IGFBP6 could be used as diagnostic markers to identify unstable plaques effectively.
Collapse
Affiliation(s)
- Jing Wang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zijian Kang
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Critical Care Medicine, Naval Medical Center of People's Liberation Army of China (PLA), Shanghai, China
| | - Yandong Liu
- Department of Geriatrics, Navy 905th Hospital, Shanghai, China
| | - Zifu Li
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Jianmin Liu, ; Yang Liu, ; Zifu Li,
| | - Yang Liu
- Department of Critical Care Medicine, Naval Medical Center of People's Liberation Army of China (PLA), Shanghai, China
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Jianmin Liu, ; Yang Liu, ; Zifu Li,
| | - Jianmin Liu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Jianmin Liu, ; Yang Liu, ; Zifu Li,
| |
Collapse
|
279
|
The adventitia in arterial development, remodeling, and hypertension. Biochem Pharmacol 2022; 205:115259. [PMID: 36150432 DOI: 10.1016/j.bcp.2022.115259] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/20/2022]
Abstract
The adventitia receives input signals from the vessel wall, the immune system, perivascular nerves and from surrounding tissues to generate effector responses that regulate structural and mechanical properties of blood vessels. It is a complex and dynamic tissue that orchestrates multiple functions for vascular development, homeostasis, repair, and disease. The purpose of this review is to highlight recent advances in our understanding of the origins, phenotypes, and functions of adventitial and perivascular cells with particular emphasis on hypertensive vascular remodeling.
Collapse
|
280
|
Besler KJ, Blanchard V, Francis GA. Lysosomal acid lipase deficiency: A rare inherited dyslipidemia but potential ubiquitous factor in the development of atherosclerosis and fatty liver disease. Front Genet 2022; 13:1013266. [PMID: 36204319 PMCID: PMC9530988 DOI: 10.3389/fgene.2022.1013266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Lysosomal acid lipase (LAL), encoded by the gene LIPA, is the sole neutral lipid hydrolase in lysosomes, responsible for cleavage of cholesteryl esters and triglycerides into their component parts. Inherited forms of complete (Wolman Disease, WD) or partial LAL deficiency (cholesteryl ester storage disease, CESD) are fortunately rare. Recently, LAL has been identified as a cardiovascular risk gene in genome-wide association studies, though the directionality of risk conferred remains controversial. It has also been proposed that the low expression and activity of LAL in arterial smooth muscle cells (SMCs) that occurs inherently in nature is a likely determinant of the propensity of SMCs to form the majority of foam cells in atherosclerotic plaque. LAL also likely plays a potential role in fatty liver disease. This review highlights the nature of LAL gene mutations in WD and CESD, the association of LAL with prediction of cardiovascular risk from genome-wide association studies, the importance of relative LAL deficiency in SMC foam cells, and the need to further interrogate the pathophysiological impact and cell type-specific role of enhancing LAL activity as a novel treatment strategy to reduce the development and induce the regression of ischemic cardiovascular disease and fatty liver.
Collapse
|
281
|
Xu S, Lyu QR, Ilyas I, Tian XY, Weng J. Vascular homeostasis in atherosclerosis: A holistic overview. Front Immunol 2022; 13:976722. [PMID: 36172381 PMCID: PMC9512393 DOI: 10.3389/fimmu.2022.976722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis refers to the deposition of lipids and the co-existence of inflammation and impaired inflammation resolution in pan-vasculature, which causes lumen narrowing, hardening, plaque formation, and the manifestation of acute cardiovascular events. Emerging evidence has suggested that vascular circulation can be viewed as a complex homeostatic system analogous to a mini-ecosystem which consists of the vascular microenvironment (niche) and the crosstalk among phenotypically and functionally diverse vascular cell types. Here, we elucidate how cell components in the vascular wall affect vascular homeostasis, structure, function, and atherosclerosis in a holistic perspective. Finally, we discuss the potential role of vascular-stabilizing strategies including pharmacotherapies, natural substances and lifestyle modifications, in preventing cardiovascular diseases by preserving vascular integrity and homeostasis.
Collapse
Affiliation(s)
- Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China (USTC), Hefei, China
- *Correspondence: Suowen Xu, ; Jianping Weng,
| | - Qing Rex Lyu
- Medical Research Institute, Chongqing General Hospital, Chongqing, China
| | - Iqra Ilyas
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China (USTC), Hefei, China
| | - Xiao-Yu Tian
- School of Biomedical Sciences, Chinese University of Hong Kong, NT, Hong Kong SAR, China
| | - Jianping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China (USTC), Hefei, China
- *Correspondence: Suowen Xu, ; Jianping Weng,
| |
Collapse
|
282
|
Gogiraju R, Gachkar S, Velmeden D, Bochenek ML, Zifkos K, Hubert A, Münzel T, Offermanns S, Schäfer K. Protein Tyrosine Phosphatase 1B Deficiency in Vascular Smooth Muscle Cells Promotes Perivascular Fibrosis following Arterial Injury. Thromb Haemost 2022; 122:1814-1826. [PMID: 36075234 PMCID: PMC9512587 DOI: 10.1055/s-0042-1755329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background
Smooth muscle cell (SMC) phenotype switching plays a central role during vascular remodeling. Growth factor receptors are negatively regulated by protein tyrosine phosphatases (PTPs), including its prototype PTP1B. Here, we examine how reduction of PTP1B in SMCs affects the vascular remodeling response to injury.
Methods
Mice with inducible PTP1B deletion in SMCs (SMC.PTP1B-KO) were generated by crossing mice expressing Cre.ER
T2
recombinase under the
Myh11
promoter with PTP1B
flox/flox
mice and subjected to FeCl
3
carotid artery injury.
Results
Genetic deletion of PTP1B in SMCs resulted in adventitia enlargement, perivascular SMA
+
and PDGFRβ
+
myofibroblast expansion, and collagen accumulation following vascular injury. Lineage tracing confirmed the appearance of
Myh11
-Cre reporter cells in the remodeling adventitia, and SCA1
+
CD45
-
vascular progenitor cells increased. Elevated mRNA expression of transforming growth factor β (TGFβ) signaling components or enzymes involved in extracellular matrix remodeling and TGFβ liberation was seen in injured SMC.PTP1B-KO mouse carotid arteries, and mRNA transcript levels of contractile SMC marker genes were reduced already at baseline. Mechanistically, Cre recombinase (mice) or siRNA (cells)-mediated downregulation of PTP1B or inhibition of ERK1/2 signaling in SMCs resulted in nuclear accumulation of KLF4, a central transcriptional repressor of SMC differentiation, whereas phosphorylation and nuclear translocation of SMAD2 and SMAD3 were reduced. SMAD2 siRNA transfection increased protein levels of PDGFRβ and MYH10 while reducing ERK1/2 phosphorylation, thus phenocopying genetic PTP1B deletion.
Conclusion
Chronic reduction of PTP1B in SMCs promotes dedifferentiation, perivascular fibrosis, and adverse remodeling following vascular injury by mechanisms involving an ERK1/2 phosphorylation-driven shift from SMAD2 to KLF4-regulated gene transcription.
Collapse
Affiliation(s)
- Rajinikanth Gogiraju
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - Sogol Gachkar
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - David Velmeden
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - Magdalena L Bochenek
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Konstantinos Zifkos
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Astrid Hubert
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Rhine-Main Site, Mainz, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, JW Goethe University Frankfurt, Frankfurt, Germany.,Cardiopulmonary Institute (CPI), Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK e.V.), Rhine-Main Site, Frankfurt and Bad Nauheim, Germany
| | - Katrin Schäfer
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Rhine-Main Site, Mainz, Germany
| |
Collapse
|
283
|
Shen Y, Xu LR, Yan D, Zhou M, Han TL, Lu C, Tang X, Lin CP, Qian RZ, Guo DQ. BMAL1 modulates smooth muscle cells phenotypic switch towards fibroblast-like cells and stabilizes atherosclerotic plaques by upregulating YAP1. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166450. [PMID: 35598770 DOI: 10.1016/j.bbadis.2022.166450] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/03/2022] [Accepted: 05/13/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Ischemic heart diseases and ischemic stroke are closely related to circadian clock and unstable atherosclerotic plaques. Vascular smooth muscle cells (VSMCs) can stabilize or destabilize an atherosclerotic lesion through phenotypic switch. BMAL1 is not only an indispensable core component in circadian clock but also an important regulator in atherosclerosis and VSMCs proliferation. However, little is known about the modulation mechanisms of BMAL1 in VSMCs phenotypic switch and atherosclerotic plaque stability. METHODS We integrated histological analysis of human plaques, in vivo experiments of VSMC-specific Bmal1-/- mice, in vitro experiments, and gene set enrichment analysis (GSEA) of public datasets of human plaques to explore the function of BMAL1 in VSMCs phonotypic switch and plaque stability. FINDINGS Comparing to human unstable plaques, BMAL1 was higher in stable plaques, accompanied by elevated YAP1 and fibroblast maker FSP1 which were positively correlated with BMAL1. In response to Methyl-β-cyclodextrin-cholesterol, oxidized-low-density-lipoprotein and platelet-derived-growth-factor-BB, VSMCs embarked on phenotypic switch and upregulated BMAL, YAP1 and FSP1. Besides, BMAL1 overexpression promoted VSMCs phonotypic switch towards fibroblast-like cells by transcriptionally upregulating the expression of YAP1. BMAL1 or YAP1 knock-down inhibited VSMCs phonotypic switch and downregulated FSP1. Furthermore, VSMC-specific Bmal1-/- mice exhibited VSMCs with lower YAP1 and FSP1 levels, and more vulnerable plaques with less collagen content. In addition, BMAL1 suppressed the migration of VSMCs. The GSEA results of public datasets were consistent with our laboratory findings. INTERPRETATION Our results highlight the importance of BMAL1 as a major regulator in VSMCs phenotypic switch towards fibroblast-like cells which stabilize an atherosclerotic plaque.
Collapse
Affiliation(s)
- Yang Shen
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Li-Rong Xu
- Department of Pathology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dong Yan
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Min Zhou
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Tong-Lei Han
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Chao Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Rd., Shanghai 200032, China
| | - Xiao Tang
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Chang-Po Lin
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China.
| | - Rui-Zhe Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Rd., Shanghai 200032, China.
| | - Da-Qiao Guo
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China.
| |
Collapse
|
284
|
Tian W, Zhang T, Wang X, Zhang J, Ju J, Xu H. Global research trends in atherosclerosis: A bibliometric and visualized study. Front Cardiovasc Med 2022; 9:956482. [PMID: 36082127 PMCID: PMC9445883 DOI: 10.3389/fcvm.2022.956482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIncreasing evidence has spurred a considerable evolution of concepts related to atherosclerosis, prompting the need to provide a comprehensive view of the growing literature. By retrieving publications in the Web of Science Core Collection (WoSCC) of Clarivate Analytics, we conducted a bibliometric analysis of the scientific literature on atherosclerosis to describe the research landscape.MethodsA search was conducted of the WoSCC for articles and reviews serving exclusively as a source of information on atherosclerosis published between 2012 and 2022. Microsoft Excel 2019 was used to chart the annual productivity of research relevant to atherosclerosis. Through CiteSpace and VOSviewer, the most prolific countries or regions, authors, journals, and resource-, intellectual-, and knowledge-sharing in atherosclerosis research, as well as co-citation analysis of references and keywords, were analyzed.ResultsA total of 20,014 publications were retrieved. In terms of publications, the United States remains the most productive country (6,390, 31,93%). The most publications have been contributed by Johns Hopkins Univ (730, 3.65%). ALVARO ALONSO produced the most published works (171, 0.85%). With a betweenness centrality of 0.17, ERIN D MICHOS was the most influential author. The most prolific journal was identified as Atherosclerosis (893, 4.46%). Circulation received the most co-citations (14,939, 2.79%). Keywords with the ongoing strong citation bursts were “nucleotide-binding oligomerization (NOD), Leucine-rich repeat (LRR)-containing protein (NLRP3) inflammasome,” “short-chain fatty acids (SCFAs),” “exosome,” and “homeostasis,” etc.ConclusionThe research on atherosclerosis is driven mostly by North America and Europe. Intensive research has focused on the link between inflammation and atherosclerosis, as well as its complications. Specifically, the NLRP3 inflammasome, interleukin-1β, gut microbiota and SCFAs, exosome, long non-coding RNAs, autophagy, and cellular senescence were described to be hot issues in the field.
Collapse
Affiliation(s)
- Wende Tian
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tai Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Gastroenterology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyi Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jianqing Ju
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jianqing Ju,
| | - Hao Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Hao Xu,
| |
Collapse
|
285
|
Kawai K, Vozenilek AE, Kawakami R, Sato Y, Ghosh SKB, Virmani R, Finn AV. Understanding the role of alternative macrophage phenotypes in human atherosclerosis. Expert Rev Cardiovasc Ther 2022; 20:689-705. [PMID: 35942866 DOI: 10.1080/14779072.2022.2111301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
INTRODUCTION Atherosclerosis-based ischemic heart disease is still the primary cause of death throughout the world. Over the past decades there has been no significant changes in the therapeutic approaches to atherosclerosis, which are mainly based on lipid lowering therapies and management of comorbid conditions such as diabetes and hypertension. The involvement of macrophages in atherosclerosis has been recognized for decades. More recently, a more detailed and sophisticated understanding of their various phenotypes and roles in the atherosclerotic process has been recognized. This new data is revealing how specific subtypes of macrophage-induced inflammation may have distinct effects on atherosclerosis progression and may provide new approaches for treatment, based upon targeting of specific macrophage subtypes. AREAS COVERED We will comprehensively review the spectrum of macrophage phenotypes and how they contribute to atherosclerotic plaque development and progression. EXPERT OPINION Various signals derived from atherosclerotic lesions drive macrophages into complex subsets with different gene expression profiles, phenotypes, and functions, not all of which are understood. Macrophage phenotypes include those that enhance, heal, and regress the atherosclerotic lesions though various mechanisms. Targeting of specific macrophage phenotypes may provide a promising and novel approach to prevent atherosclerosis progression.
Collapse
Affiliation(s)
- Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aimee E Vozenilek
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Yu Sato
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | | | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aloke V Finn
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA.,University of Maryland, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
286
|
Wang Y, Wang Q, Xu D. New insights into macrophage subsets in atherosclerosis. J Mol Med (Berl) 2022; 100:1239-1251. [PMID: 35930063 DOI: 10.1007/s00109-022-02224-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 05/27/2022] [Accepted: 06/15/2022] [Indexed: 12/11/2022]
Abstract
Macrophages in atherosclerotic patients are notably plastic and heterogeneous. Single-cell RNA sequencing (Sc RNA-seq) can provide information about all the RNAs in individual cells, and it is used to identify cell subpopulations in atherosclerosis (AS) and reveal the heterogeneity of these cells. Recently, some findings from Sc RNA-seq experiments have suggested the existence of multiple macrophage subsets in atherosclerotic plaque lesions, and these subsets exhibit significant differences in their gene expression levels and functions. These cells affect various aspects of plaque lesion development, stabilization, and regression, as well as plaque rupture. This article aims to review the content and results of current studies that used RNA-seq to explore the different types of macrophages in AS and the related molecular mechanisms as well as to identify the potential roles of these macrophage types in the pathogenesis of atherosclerotic plaques. Also, this review listed some new therapeutic targets for delaying atherosclerotic lesion progression and treatment based on the experimental results.
Collapse
Affiliation(s)
- Yurong Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qiong Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
287
|
Integrating multiplex immunofluorescent and mass spectrometry imaging to map myeloid heterogeneity in its metabolic and cellular context. Cell Metab 2022; 34:1214-1225.e6. [PMID: 35858629 DOI: 10.1016/j.cmet.2022.06.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 02/28/2022] [Accepted: 06/23/2022] [Indexed: 12/24/2022]
Abstract
Cells often adopt different phenotypes, dictated by tissue-specific or local signals such as cell-cell and cell-matrix contacts or molecular micro-environment. This holds in extremis for macrophages with their high phenotypic plasticity. Their broad range of functions, some even opposing, reflects their heterogeneity, and a multitude of subsets has been described in different tissues and diseases. Such micro-environmental imprint cannot be adequately studied by single-cell applications, as cells are detached from their context, while histology-based assessment lacks the phenotypic depth due to limitations in marker combination. Here, we present a novel, integrative approach in which 15-color multispectral imaging allows comprehensive cell classification based on multi-marker expression patterns, followed by downstream analysis pipelines to link their phenotypes to contextual, micro-environmental cues, such as their cellular ("community") and metabolic ("local lipidome") niches in complex tissue. The power of this approach is illustrated for myeloid subsets and associated lipid signatures in murine atherosclerotic plaque.
Collapse
|
288
|
Chattopadhyay A, Guan (关蒲骏) P, Majumder S, Kaw K, Zhou (周桢) Z, Zhang C, Prakash SK, Kaw A, Buja LM, Kwartler CS, Milewicz DM. Preventing Cholesterol-Induced Perk (Protein Kinase RNA-Like Endoplasmic Reticulum Kinase) Signaling in Smooth Muscle Cells Blocks Atherosclerotic Plaque Formation. Arterioscler Thromb Vasc Biol 2022; 42:1005-1022. [PMID: 35708026 PMCID: PMC9311463 DOI: 10.1161/atvbaha.121.317451] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/31/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Vascular smooth muscle cells (SMCs) undergo complex phenotypic modulation with atherosclerotic plaque formation in hyperlipidemic mice, which is characterized by de-differentiation and heterogeneous increases in the expression of macrophage, fibroblast, osteogenic, and stem cell markers. An increase of cellular cholesterol in SMCs triggers similar phenotypic changes in vitro with exposure to free cholesterol due to cholesterol entering the endoplasmic reticulum, triggering endoplasmic reticulum stress and activating Perk (protein kinase RNA-like endoplasmic reticulum kinase) signaling. METHODS We generated an SMC-specific Perk knockout mouse model, induced hyperlipidemia in the mice by AAV-PCSK9DY injection, and subjected them to a high-fat diet. We then assessed atherosclerotic plaque formation and performed single-cell transcriptomic studies using aortic tissue from these mice. RESULTS SMC-specific deletion of Perk reduces atherosclerotic plaque formation in male hyperlipidemic mice by 80%. Single-cell transcriptomic data identify 2 clusters of modulated SMCs in hyperlipidemic mice, one of which is absent when Perk is deleted in SMCs. The 2 modulated SMC clusters have significant overlap of transcriptional changes, but the Perk-dependent cluster uniquely shows a global decrease in the number of transcripts. SMC-specific Perk deletion also prevents migration of both contractile and modulated SMCs from the medial layer of the aorta. CONCLUSIONS Our results indicate that hypercholesterolemia drives both Perk-dependent and Perk-independent SMC modulation and that deficiency of Perk significantly blocks atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Abhijnan Chattopadhyay
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School The University of Texas Health Science Center at Houston (A.C., P.G., S.M., K.K., Z.Z., S.K.P.‚ A.K., C.S.K., D.M.M.)
| | - Pujun Guan (关蒲骏)
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School The University of Texas Health Science Center at Houston (A.C., P.G., S.M., K.K., Z.Z., S.K.P.‚ A.K., C.S.K., D.M.M.)
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and UTHealth, Houston (P.G.)
| | - Suravi Majumder
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School The University of Texas Health Science Center at Houston (A.C., P.G., S.M., K.K., Z.Z., S.K.P.‚ A.K., C.S.K., D.M.M.)
| | - Kaveeta Kaw
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School The University of Texas Health Science Center at Houston (A.C., P.G., S.M., K.K., Z.Z., S.K.P.‚ A.K., C.S.K., D.M.M.)
| | - Zhen Zhou (周桢)
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School The University of Texas Health Science Center at Houston (A.C., P.G., S.M., K.K., Z.Z., S.K.P.‚ A.K., C.S.K., D.M.M.)
| | - Chen Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (C.Z.)
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston (C.Z.)
| | - Siddharth K. Prakash
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School The University of Texas Health Science Center at Houston (A.C., P.G., S.M., K.K., Z.Z., S.K.P.‚ A.K., C.S.K., D.M.M.)
| | - Anita Kaw
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School The University of Texas Health Science Center at Houston (A.C., P.G., S.M., K.K., Z.Z., S.K.P.‚ A.K., C.S.K., D.M.M.)
| | - L. Maximillian Buja
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston (L.M.B.)
| | - Callie S. Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School The University of Texas Health Science Center at Houston (A.C., P.G., S.M., K.K., Z.Z., S.K.P.‚ A.K., C.S.K., D.M.M.)
| | - Dianna M. Milewicz
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School The University of Texas Health Science Center at Houston (A.C., P.G., S.M., K.K., Z.Z., S.K.P.‚ A.K., C.S.K., D.M.M.)
| |
Collapse
|
289
|
Owsiany KM, Deaton RA, Soohoo KG, Nguyen AT, Owens GK. Dichotomous Roles of Smooth Muscle Cell-Derived MCP1 (Monocyte Chemoattractant Protein 1) in Development of Atherosclerosis. Arterioscler Thromb Vasc Biol 2022; 42:942-956. [PMID: 35735018 PMCID: PMC9365248 DOI: 10.1161/atvbaha.122.317882] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Smooth muscle cells (SMCs) in atherosclerotic plaque take on multiple nonclassical phenotypes that may affect plaque stability and, therefore, the likelihood of myocardial infarction or stroke. However, the mechanisms by which these cells affect stability are only beginning to be explored. METHODS In this study, we investigated the contribution of inflammatory MCP1 (monocyte chemoattractant protein 1) produced by both classical Myh11 (myosin heavy chain 11)+ SMCs and SMCs that have transitioned through an Lgals3 (galectin 3)+ state in atherosclerosis using smooth muscle lineage tracing mice that label all Myh11+ cells and a dual lineage tracing system that targets Lgals3-transitioned SMC only. RESULTS We show that loss of MCP1 in all Myh11+ smooth muscle results in a paradoxical increase in plaque size and macrophage content, driven by a baseline systemic monocytosis early in atherosclerosis pathogenesis. In contrast, knockout of MCP1 in Lgals3-transitioned SMCs using a complex dual lineage tracing system resulted in lesions with an increased Acta2 (actin alpha 2, smooth muscle)+ fibrous cap and decreased investment of Lgals3-transitioned SMCs, consistent with increased plaque stability. Finally, using flow cytometry and single-cell RNA sequencing, we show that MCP1 produced by Lgals3-transitioned SMCs influences multiple populations of inflammatory cells in late-stage plaques. CONCLUSIONS MCP1 produced by classical SMCs influences monocyte levels beginning early in disease and was atheroprotective, while MCP1 produced by the Lgals3-transitioned subset of SMCs exacerbated plaque pathogenesis in late-stage disease. Results are the first to determine the function of Lgals3-transitioned inflammatory SMCs in atherosclerosis and highlight the need for caution when considering therapeutic interventions involving MCP1.
Collapse
Affiliation(s)
- Katherine M. Owsiany
- University of Virginia School of Medicine, Charlottesville VA 22903,Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908, USA
| | - Rebecca A. Deaton
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908, USA
| | | | | | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908, USA.,Corresponding author: Univ. of Virginia School of Medicine, Robert M. Berne Cardiovascular Research Center, PO Box 801394, MR5 Building, Charlottesville, Virginia 22908-1394, Phone: 434-924-5993,
| |
Collapse
|
290
|
Pan D, Wu W, Zuo G, Xie X, Li H, Ren X, Kong C, Zhou W, Zhang Z, Waterfall M, Chen S. Sphingosine 1-phosphate receptor 2 promotes erythrocyte clearance by vascular smooth muscle cells in intraplaque hemorrhage through MFG-E8 production. Cell Signal 2022; 98:110419. [PMID: 35905868 DOI: 10.1016/j.cellsig.2022.110419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/13/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
Intraplaque hemorrhage (IPH) accelerates atherosclerosis progression. To scavenge excessive red blood cells (RBCs), vascular smooth muscle cells (VSMCs) with great plasticity may function as phagocytes. Here, we investigated the erythrophagocytosis function of VSMCs and possible regulations involved. Based on transcriptional microarray analysis, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis showed that genes up-regulated in human carotid atheroma with IPH were enriched in functions of phagocytic activities, while those down-regulated were enriched in VSMCs contraction function. Transcriptional expression of Milk fat globule-epidermal growth factor 8 (MFG-E8) was also down-regulated in atheroma with IPH. In high-fat diet-fed apolipoprotein E-deficient mice, erythrocytes were present in cells expressing VSMC markers αSMA in the brachiocephalic artery, suggesting VSMCs play a role in erythrophagocytosis. Using immunofluorescence and flow cytometry, we also found that eryptotic RBCs were bound to and internalized by VSMCs in a phosphatidylserine/MFG-E8/integrin αVβ3 dependent manner in vitro. Inhibiting S1PR2 signaling with specific inhibitor JTE-013 or siRNA decreased Mfge8 expression and impaired the erythrophagocytosis of VSMCs in vitro. Partial ligation was performed in the left common carotid artery (LCA) followed by intra-intimal injection of isolated erythrocytes to observe their clearance in vivo. Interfering S1PR2 expression in VSMCs with Adeno-associated virus 9 inhibited MFG-E8 expression inside LCA plaques receiving RBCs injection and attenuated erythrocytes clearance. Erythrophagocytosis by VSMCs increased vascular endothelial growth factor-a secretion and promoted angiogenesis. The present study revealed that VSMCs act as phagocytes for RBC clearance through S1PR2 activation induced MFG-E8 release.
Collapse
Affiliation(s)
- Daorong Pan
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Wen Wu
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Guangfeng Zuo
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Xiangrong Xie
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Hui Li
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Xiaomin Ren
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Chaohua Kong
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Wenying Zhou
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Zihan Zhang
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Martin Waterfall
- Institute of Immunology & Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH8 9JZ, United Kingdom
| | - Shaoliang Chen
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China.
| |
Collapse
|
291
|
Khachigian LM, Black BL, Ferdinandy P, De Caterina R, Madonna R, Geng YJ. Transcriptional regulation of vascular smooth muscle cell proliferation, differentiation and senescence: Novel targets for therapy. Vascul Pharmacol 2022; 146:107091. [PMID: 35896140 DOI: 10.1016/j.vph.2022.107091] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 10/16/2022]
Abstract
Vascular smooth muscle cells (SMC) possess a unique cytoplasticity, regulated by transcriptional, translational and phenotypic transformation in response to a diverse range of extrinsic and intrinsic pathogenic factors. The mature, differentiated SMC phenotype is physiologically typified transcriptionally by expression of genes encoding "contractile" proteins, such as SMα-actin (ACTA2), SM-MHC (myosin-11) and SM22α (transgelin). When exposed to various pathological conditions (e.g., pro-atherogenic risk factors, hypertension), SMC undergo phenotypic modulation, a bioprocess enabling SMC to de-differentiate in immature stages or trans-differentiate into other cell phenotypes. As recent studies suggest, the process of SMC phenotypic transformation involves five distinct states characterized by different patterns of cell growth, differentiation, migration, matrix protein expression and declined contractility. These changes are mediated via the action of several transcriptional regulators, including myocardin and serum response factor. Conversely, other factors, including Kruppel-like factor 4 and nuclear factor-κB, can inhibit SMC differentiation and growth arrest, while factors such as yin yang-1, can promote SMC differentiation whilst inhibiting proliferation. This article reviews recent advances in our understanding of regulatory mechanisms governing SMC phenotypic modulation. We propose the concept that transcription factors mediating this switching are important biomarkers and potential pharmacological targets for therapeutic intervention in cardiovascular disease.
Collapse
Affiliation(s)
- Levon M Khachigian
- Vascular Biology and Translational Research, Department of Pathology, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Brian L Black
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States of America
| | - Péter Ferdinandy
- Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| | - Raffaele De Caterina
- Cardiovascular Division, Pisa University Hospital & University of Pisa, Via Paradisa, 2, Pisa 56124, Italy
| | - Rosalinda Madonna
- Cardiovascular Division, Pisa University Hospital & University of Pisa, Via Paradisa, 2, Pisa 56124, Italy; Division of Cardiovascular Medicine, Department of Internal Medicine, The Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Yong-Jian Geng
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| |
Collapse
|
292
|
Huang J, Huang Y, Shi X, Lyu Y, Wu M, Chen Y, Zhou L, Yu H, Xie H, Chen F. Phenotypic modulation of vascular smooth muscle cells in the corpus spongiosum surrounding the urethral plate in hypospadias. Andrologia 2022; 54:e14540. [PMID: 35866316 DOI: 10.1111/and.14540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/08/2022] [Accepted: 07/11/2022] [Indexed: 11/28/2022] Open
Abstract
Hypospadias is an abnormal ventral development of the penis caused by incomplete virilization of the male genital tubercle. This study investigated the phenotypic modulation of vascular smooth muscle cells (VSMCs) in the corpus spongiosum surrounding the urethral plate in hypospadias. The urethral corpus spongiosum tissue was collected for HE, Masson and α-SMA immunohistochemical staining. Spongiosum VSMCs were cultured and identified by α-SMA fluorescence. qRT-PCR and Western blotting and fluorescence were performed. The results showed that the vascular lumen of the corpus spongiosum around the urethral plate was larger and that the vascular smooth muscle layer was thicker in hypospadias. The expression of the contractile markers α-SMA and Calponin 1 in VSMCs was decreased, the expression of the synthetic marker OPN was increased, and the transcription of the phenotypic switching factors SRF and MYOCD was decreased. The expression of Ki67, PCNA and BAX was increased, and the expression of Bcl-2 was decreased. The phenotype of corpus spongiosum VSMCs in hypospadias changed from the contractional type to the synthetic type. This phenotypic modulation was associated with increased proliferation and apoptosis rates. SRF and MYOCD may be the main factors mediating the phenotypic modulation of urethral corpus spongiosum VSMCs.
Collapse
Affiliation(s)
- Jiayao Huang
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yichen Huang
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiujuan Shi
- School of Medicine, Tongji University, Shanghai, China
| | - Yiqing Lyu
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Min Wu
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Chen
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lijun Zhou
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huan Yu
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hua Xie
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fang Chen
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
293
|
Feng M, Zhou Q, Tu W, Wang Y, Du Y, Xu K. ATF4 promotes brain vascular smooth muscle cells proliferation, invasion and migration by targeting miR-552-SKI axis. PLoS One 2022; 17:e0270880. [PMID: 35857794 PMCID: PMC9299314 DOI: 10.1371/journal.pone.0270880] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Background Studies have indicated vascular smooth muscle cells (VSMCs) played a crucial role in atherosclerosis and microRNAs (miRNAs) played key roles in biological functions of VSMCs. Whereas, the potential function and mechanism of miR-552 in VSMCs remains unclear. Our aim was to explore the role of miR-552 on VSMCs and underlying mechanism. Material/Methods MTT assay and transwell assay were used to measure the proliferation, invasion, and migration of human brain VSMCs (HBVSMCs) and mice VSMCs (mVSMCs), respectively. Bioinformatics tools and luciferase assay were adopted to verify the association between miR-552 and SKI. Rescue experiments were employed to assess the interaction of miR-552 and SKI in modulating biological functions in HBVSMCs and mVSMCs. The expression level of transcription factors (TFs)was measured via qRT-PCR assay. The effect of ATF4 on miR-552 and SKI expression was tested by qRT-PCR or western blot assay. Finally, chromatin immunoprecipitation (ChIP) assay and JASPAR databases were used to analyze the regulatory linkage between ATF4 and miR-552. Results We found that miR-552 was upregulated in HBVSMCs treated with PDGF-bb and miR-552 overexpression could promote proliferation, invasion, and migration of HBVSMCs and mVSMCs, whereas, miR-552 knockdown had the opposite impact. In addition, we also found that SKI was a direct target of miR-552, which reversed miR-552-mediated proliferation, invasion, and migration in HBVSMCs and mVSMCs. Furthermore, we also discovered that miR-552 overexpression promoted the effects of ATF4 elevation on proliferation, migration and invasion of HBVSMCs and mVSMCs, but, miR-552 decline had the opposite impact. Conclusions ATF4-miR-552-SKI axis played critical roles in the proliferation and migration of HBVSMCs and mVSMCs, which were closely involved in atherosclerosis (AS). Therefore, our findings might offer a novel therapeutic target for AS.
Collapse
Affiliation(s)
- Meina Feng
- Department of Neurology, Wuhan Brain Hospital, General Hospital of the YANGTZE River Shipping, Wuhan, China
- * E-mail: (MF); (QZ)
| | - Qin Zhou
- Department of Neurology, Wuhan Brain Hospital, General Hospital of the YANGTZE River Shipping, Wuhan, China
- * E-mail: (MF); (QZ)
| | - Wenxian Tu
- Department of Neurology, Wuhan Brain Hospital, General Hospital of the YANGTZE River Shipping, Wuhan, China
| | - Yunfeng Wang
- Department of Neurology, Wuhan Brain Hospital, General Hospital of the YANGTZE River Shipping, Wuhan, China
| | - Yuanmin Du
- Department of Neurology, Wuhan Brain Hospital, General Hospital of the YANGTZE River Shipping, Wuhan, China
| | - Kang Xu
- Department of Neurology, Wuhan Brain Hospital, General Hospital of the YANGTZE River Shipping, Wuhan, China
| |
Collapse
|
294
|
Peng M, Sun R, Hong Y, Wang J, Xie Y, Zhang X, Li J, Guo H, Xu P, Li Y, Wang X, Wan T, Zhao Y, Huang F, Wang Y, Ye R, Liu Q, Liu G, Liu X, Xu G. Extracellular vesicles carrying proinflammatory factors may spread atherosclerosis to remote locations. Cell Mol Life Sci 2022; 79:430. [PMID: 35851433 PMCID: PMC11071964 DOI: 10.1007/s00018-022-04464-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022]
Abstract
Most cells involved in atherosclerosis release extracellular vesicles (EVs), which can carry bioactive substances to downstream tissues via circulation. We hypothesized that EVs derived from atherosclerotic plaques could promote atherogenesis in remote locations, a mechanism that mimics the blood metastasis of cancer. Ldlr gene knockout (Ldlr KO) rats were fed on a high cholesterol diet and underwent partial carotid ligation to induce local atherosclerosis. EVs were separated from carotid artery tissues and downstream blood of carotid ligation by centrifugation. MiRNA sequencing and qPCR were then performed to detect miRNA differences in EVs from rats with and without induced carotid atherosclerosis. Biochemical analyses demonstrated that EVs derived from atherosclerosis could increase the expression of ICAM-1, VCAM-1, and E-selectin in endothelial cells in vitro. EVs derived from atherosclerosis contained a higher level of miR-23a-3p than those derived from controls. MiR-23a-3p could promote endothelial inflammation by targeting Dusp5 and maintaining ERK1/2 phosphorylation in vitro. Inhibiting EV release could attenuate atherogenesis and reduce macrophage infiltration in vivo. Intravenously administrating atherosclerotic plaque-derived EVs could induce intimal inflammation, arterial wall thickening and lumen narrowing in the carotids of Ldlr KO rats, while simultaneous injection of miR-23a-3p antagomir could reverse this reaction. The results suggested that EVs may transfer atherosclerosis to remote locations by carrying proinflammatory factors, particularly miR-23a-3p.
Collapse
Affiliation(s)
- Mengna Peng
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Rui Sun
- Department of Neurology, Shanghai Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Ye Hong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210002, Jiangsu, China
| | - Jia Wang
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Yi Xie
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Xiaohao Zhang
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Juanji Li
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Hongquan Guo
- Department of Neurology, Jinling Hospital, the First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, Jiangsu, China
| | - Pengfei Xu
- Division of Life Sciences and Medicine, Stroke Center & Department of Neurology, Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Yunzi Li
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Xiaoke Wang
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Ting Wan
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Ying Zhao
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Feihong Huang
- Department of Neurology, Guilin People's Hospital, Guilin, 541002, Guangxi, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191, China
- Institute of Cardiovascular Sciences, School of Basic Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Ruidong Ye
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Qian Liu
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - George Liu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191, China
- Institute of Cardiovascular Sciences, School of Basic Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Xinfeng Liu
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China.
- Department of Neurology, Jinling Hospital, the First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, Jiangsu, China.
- Division of Life Sciences and Medicine, Stroke Center & Department of Neurology, Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230036, Anhui, China.
| | - Gelin Xu
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China.
- Department of Neurology, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China.
| |
Collapse
|
295
|
Meneri M, Bonato S, Gagliardi D, Comi GP, Corti S. New Insights into Cerebral Vessel Disease Landscapes at Single-Cell Resolution: Pathogenetic and Therapeutic Perspectives. Biomedicines 2022; 10:1693. [PMID: 35884997 PMCID: PMC9313091 DOI: 10.3390/biomedicines10071693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/03/2022] [Accepted: 07/11/2022] [Indexed: 11/19/2022] Open
Abstract
Cerebrovascular diseases are a leading cause of death and disability globally. The development of new therapeutic targets for cerebrovascular diseases (e.g., ischemic, and hemorrhagic stroke, vascular dementia) is limited by a lack of knowledge of the cellular and molecular biology of health and disease conditions and the factors that cause injury to cerebrovascular structures. Here, we describe the role of advances in omics technology, particularly RNA sequencing, in studying high-dimensional, multifaceted profiles of thousands of individual blood and vessel cells at single-cell resolution. This analysis enables the dissection of the heterogeneity of diseased cerebral vessels and their atherosclerotic plaques, including the microenvironment, cell evolutionary trajectory, and immune response pathway. In animal models, RNA sequencing permits the tracking of individual cells (including immunological, endothelial, and vascular smooth muscle cells) that compose atherosclerotic plaques and their alteration under experimental settings such as phenotypic transition. We describe how single-cell RNA transcriptomics in humans allows mapping to the molecular and cellular levels of atherosclerotic plaques in cerebral arteries, tracking individual lymphocytes and macrophages, and how these data can aid in identifying novel immune mechanisms that could be exploited as therapeutic targets for cerebrovascular diseases. Single-cell multi-omics approaches will likely provide the unprecedented resolution and depth of data needed to generate clinically relevant cellular and molecular signatures for the precise treatment of cerebrovascular diseases.
Collapse
Affiliation(s)
- Megi Meneri
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Sara Bonato
- Stroke Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Delia Gagliardi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Giacomo P. Comi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
296
|
Wang P, Pan Y, Yang C, Zhang L, Zhao Z, Ye K, Li L, Xia S, Lu X, Shi H, Li W, Yin M. TNFα activation and TGFβ blockage act synergistically for smooth muscle cell calcification in patients with venous thrombosis via TGFβ/ERK pathway. J Cell Mol Med 2022; 26:4479-4491. [PMID: 35808901 PMCID: PMC9357635 DOI: 10.1111/jcmm.17472] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/12/2022] [Accepted: 06/16/2022] [Indexed: 11/26/2022] Open
Abstract
Venous calcification has been observed in post‐thrombotic syndrome (PTS) patients; yet, the cell types and possible mechanisms regulating this process are still unclear. We evaluated the calcium deposition within the venous wall, the cell type involved in the calcified remodelling of the venous wall after thrombosis and explored possible mechanisms in vitro. Calcium deposition was found in human specimens of superficial thrombotic veins and was co‐localized with VSMCs markers αSMA and TAGLN (also known as SM22α). Besides, the expression of osteogenesis‐related genes was dramatically changed in superficial thrombotic veins. Moreover, the inhibition of the TGFβ signalling pathway after TNFα treatment effectively induced the expression of osteogenic phenotype markers, the calcium salt deposits and the obvious phosphorylation of ERK1/2 and JNK2 in the VSMCs calcification model. Supplementing TGFβ2 or blocking the activation of the ERK/MAPK signalling pathway prevented the transformation of VSMCs into osteoblast‐like cells in vitro. Taken together, VSMCs have an important role in venous calcification after thrombosis. Supplementing TGFβ2 or inhibiting the ERK/MAPK signalling pathway can reduce the appearance of VSMCs osteogenic phenotype. Our findings may present a novel therapeutic approach to prevent of vascular calcification after venous thrombosis.
Collapse
Affiliation(s)
- Penghui Wang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yiqing Pan
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenghao Yang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Linjie Zhang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zhen Zhao
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Kaichuang Ye
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lei Li
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shoubing Xia
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Vascular Center of Shanghai JiaoTong University, Shanghai, China
| | - Huihua Shi
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Weimin Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Minyi Yin
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Vascular Center of Shanghai JiaoTong University, Shanghai, China
| |
Collapse
|
297
|
Lu S, Weiser-Evans MC. Lgals3-Transitioned Inflammatory Smooth Muscle Cells: Major Regulators of Atherosclerosis Progression and Inflammatory Cell Recruitment. Arterioscler Thromb Vasc Biol 2022; 42:957-959. [DOI: 10.1161/atvbaha.122.318009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sizhao Lu
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora. (S.L., M.C.M.W.-E.)
| | - Mary C.M. Weiser-Evans
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora. (S.L., M.C.M.W.-E.)
- Department of Medicine, School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado Anschutz Medical Campus, Aurora. (M.C.M.W.-E.)
- Department of Medicine, Cardiovascular Pulmonary Research Program, University of Colorado Anschutz Medical Campus, Aurora. (M.C.M.W.-E.)
| |
Collapse
|
298
|
Zhou H, Wang L, Liu S, Wang W. The role of phosphoinositide 3-kinases in immune-inflammatory responses: potential therapeutic targets for abdominal aortic aneurysm. Cell Cycle 2022; 21:2339-2364. [PMID: 35792922 DOI: 10.1080/15384101.2022.2094577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The pathogenesis of abdominal aortic aneurysm (AAA) includes inflammatory responses, matrix metalloproteinases (MMPs) degradation, VSMC apoptosis, oxidative stress, and angiogenesis, among which the inflammatory response plays a key role. At present, surgery is the only curing treatment, and no effective drug can delay AAA progression in clinical practice. Therefore, searching for a signaling pathway related to the immune-inflammatory response is an essential direction for developing drugs targeting AAA. Recent studies have confirmed that the PI3K family plays an important role in many inflammatory diseases and is involved in regulating various cellular functions, especially in the immune-inflammatory response. This review focuses on the role of each isoform of PI3K in each stage of AAA immune-inflammatory response, making available explorations for a deeper understanding of the mechanism of inflammation and immune response during the formation and development of AAA.
Collapse
Affiliation(s)
- Haiyang Zhou
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shuai Liu
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
299
|
Theofilis P, Sagris M, Antonopoulos AS, Oikonomou E, Tsioufis K, Tousoulis D. Non-Invasive Modalities in the Assessment of Vulnerable Coronary Atherosclerotic Plaques. Tomography 2022; 8:1742-1758. [PMID: 35894012 PMCID: PMC9326642 DOI: 10.3390/tomography8040147] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/04/2022] [Accepted: 07/04/2022] [Indexed: 12/26/2022] Open
Abstract
Coronary atherosclerosis is a complex, multistep process that may lead to critical complications upon progression, revolving around plaque disruption through either rupture or erosion. Several high-risk features are associated with plaque vulnerability and may add incremental prognostic information. Although invasive imaging modalities such as optical coherence tomography or intravascular ultrasound are considered to be the gold standard in the assessment of vulnerable coronary atherosclerotic plaques (VCAPs), contemporary evidence suggests a potential role for non-invasive methods in this context. Biomarkers associated with deleterious pathophysiologic pathways, including inflammation and extracellular matrix degradation, have been correlated with VCAP characteristics and adverse prognosis. However, coronary computed tomography (CT) angiography has been the most extensively investigated technique, significantly correlating with invasive method-derived VCAP features. The estimation of perivascular fat attenuation as well as radiomic-based approaches represent additional concepts that may add incremental information. Cardiac magnetic resonance imaging (MRI) has also been evaluated in clinical studies, with promising results through the various image sequences that have been tested. As far as nuclear cardiology is concerned, the implementation of positron emission tomography in the VCAP assessment currently faces several limitations with the myocardial uptake of the radiotracer in cases of fluorodeoxyglucose use, as well as with motion correction. Moreover, the search for the ideal radiotracer and the most adequate combination (CT or MRI) is still ongoing. With a look to the future, the possible combination of imaging and circulating inflammatory and extracellular matrix degradation biomarkers in diagnostic and prognostic algorithms may represent the essential next step for the assessment of high-risk individuals.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
- Correspondence:
| | - Marios Sagris
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| | - Alexios S. Antonopoulos
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| | - Evangelos Oikonomou
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
- 3rd Cardiology Department, Thoracic Diseases Hospital “Sotiria”, University of Athens Medical School, 11527 Athens, Greece
| | - Konstantinos Tsioufis
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| | - Dimitris Tousoulis
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| |
Collapse
|
300
|
Parnigoni A, Viola M, Karousou E, Rovera S, Giaroni C, Passi A, Vigetti D. ROLE OF HYALURONAN IN PATHOPHYSIOLOGY OF VASCULAR1 ENDOTHELIAL AND SMOOTH MUSCLE CELLS. Am J Physiol Cell Physiol 2022; 323:C505-C519. [PMID: 35759431 DOI: 10.1152/ajpcell.00061.2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the main components of the extracellular matrix (ECM) of the blood vessel is hyaluronic acid or hyaluronan (HA). It is a ubiquitous polysaccharide belonging to the family of glycosaminoglycans, but, differently from other proteoglycan-associated glycosaminoglycans, it is synthesized on the plasma membrane by a family of three HA synthases (HAS). HA can be released as a free polymer in the extracellular space or remain associated with the membrane in the pericellular space via HAS or via binding proteins. In fact, several cell surface proteins can interact with HA working as HA receptors like CD44, RHAMM, and LYVE-1. In physiological conditions, HA is localized in the glycocalyx and in the adventitia and is responsible for the loose and hydrated vascular structure favoring flexibility and allowing the stretching of vessels in response to mechanical forces. During atherogenesis, ECM undergoes dramatic alterations which have a crucial role in lipoprotein retention and in triggering multiple signaling cascades that wake up cells from their quiescent status. HA becomes highly present in the media and neointima favoring smooth muscle cells dedifferentiation, migration, and proliferation that strongly contribute to vessel wall thickening. Further, HA is able to modulate immune cell recruitment both within the vessel wall and on the endothelial cell layer. This review is focused on the effects of HA on vascular cell behavior.
Collapse
Affiliation(s)
- Arianna Parnigoni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Simona Rovera
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|