1
|
Ma MQ, Yang C, Jin SY, Yang Y, Pan YY, Lin XH. Eburicoic acid inhibits endothelial cell pyroptosis and retards the development of atherosclerosis through the Keap1/Nrf2/HO‑1/ROS pathway. Mol Med Rep 2025; 32:186. [PMID: 40314092 PMCID: PMC12076288 DOI: 10.3892/mmr.2025.13551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 03/12/2025] [Indexed: 05/03/2025] Open
Abstract
Atherosclerosis (AS)‑related coronary artery disease is the main cause of morbidity and mortality around the globe. Eburicoic acid, a triterpenoid compound from Antrodia camphorata, exerts anti‑inflammatory and anti‑hyperlipidemic effects, although its role in atherogenesis remains unknown. Endothelial cell pyroptosis‑caused chronic inflammatory response within vessel walls is a critical initial event in atherogenesis, making it a promising target to prevent AS. The present study was designed to investigate the effects of eburicoic acid on endothelial cell pyroptosis, AS progression and the underlying mechanisms. The results showed that with dose and time increased, treatment of human umbilical vascular endothelial cells (HUVECs) with eburicoic acid markedly decreased the expression of Kelch‑like ECH‑associated protein 1 (Keap1), NF‑E2‑related factor 2 (Nrf2), reactive oxygen species (ROS), NLR family pyrin domain‑containing protein 3 (NLRP3), cleaved caspase‑1, apoptosis‑associated speck‑like protein containing CARD (ASC), N‑terminal gasdermin‑D (GSDMD‑N), downregulated the secretion levels of pro‑inflammatory cytokines interleukin (IL) 1β, IL‑6 and IL‑18, inhibited caspase‑1 activity and lactate dehydrogenase release and improved plasma membrane integrity. By contrast, the expression of nuclear Nrf2, total Nrf2 and heme oxygenase‑1 (HO‑1) were increased by eburicoic acid treatment in HUVECs dose‑ and time‑dependently. Moreover, the inhibitory effects of eburicoic acid on HUVEC pyroptosis were mainly compromised by pre‑treatment with ROS agonist, HO‑1 small interfering (si)RNA, or Nrf2 siRNA. Finally, it was observed that administering high‑fat‑diet fed ApoE‑/‑ mice with eburicoic acid markedly increased Nrf2 and HO‑1 levels and reduced the expression of Keap1, NLRP3, cleaved caspase‑1, ASC and GSDMD‑N in aortas and ameliorated hyperlipidemia and inflammation in the serum, leading to smaller atherosclerotic plaques, less lipid accumulation and high content of collagen fiber within plaques. These findings identified eburicoic acid as a potent anti‑atherogenic natural product by suppressing endothelial cell pyroptosis via the Keap1/Nrf2/HO‑1/ROS pathway. Eburicoic acid may be considered an effective phytomedicine for treating AS.
Collapse
Affiliation(s)
- Meng-Qing Ma
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Chun Yang
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Shi-Yu Jin
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yu Yang
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yan-Yan Pan
- Department of Cardiology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230022, P.R. China
| | - Xian-He Lin
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
2
|
Tang X, Estau D, Huang X, Li Z. Edaravone targets PDGFRβ to attenuate VSMC phenotypic transition. Life Sci 2025; 370:123568. [PMID: 40113075 DOI: 10.1016/j.lfs.2025.123568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/06/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
AIMS PDGFRβ-driven phenotypic transition of vascular smooth muscle cells (VSMCs) is a pathological hallmark in various cardiovascular diseases, yet effective interventions are lacking. Here, we explored a promising drug targeting PDGFRβ against VSMC phenotypic transition. MATERIALS AND METHODS Connectivity map (CMAP) analysis was employed to identify the promising drug targeting PDGFRβ against VSMC phenotypic transition. A cell model stimulated with PDGF-BB and a mouse model of femoral artery injury were used to study the effects of edaravone (EDA) on VSMC phenotypic transition and PDGFRβ signaling. Molecular docking, drug affinity responsive target stability (DRATS) and cellular thermal shift assay (CETSA) were used to investigate whether EDA targeted PDGFRβ, which was further validated by a titration experiment. KEY FINDINGS Our study revealed that an approved drug EDA might target PDGFRβ against VSMC phenotypic transition. CMAP analysis unraveled EDA as a potential drug related to PDGFRβ. EDA markedly suppressed PDGFRβ-mediated VSMC transition from a contractile to a dedifferentiated phenotype, and reduced neointimal formation in wire-injured arteries. Mechanistically, molecular docking studies showed that EDA interacted with PDGFRβ, which was further confirmed by DRATS and CETSA. Consequently, EDA significantly suppressed PDGFRβ downstream signaling, including AKT and ERK1/2. Furthermore, EDA inhibited VSMC phenotypic transition in a PDGFRβ-dependent manner. SIGNIFICANCE Our work identifies EDA as a repurposed drug targeting PDGFRβ to attenuate VSMC phenotypic transition and provide new intervention measures for cardiovascular diseases associated with VSMC phenotypic transition.
Collapse
MESH Headings
- Animals
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Mice
- Edaravone/pharmacology
- Molecular Docking Simulation
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Male
- Mice, Inbred C57BL
- Phenotype
- Humans
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Xueqing Tang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Dannya Estau
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China; Department of Pharmaceutical Management and Clinical Pharmacy, College of Pharmacy, Peking University, Beijing, China
| | - Xiaoru Huang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China; Department of Pharmaceutical Management and Clinical Pharmacy, College of Pharmacy, Peking University, Beijing, China
| | - Zijian Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China; Department of Pharmaceutical Management and Clinical Pharmacy, College of Pharmacy, Peking University, Beijing, China.
| |
Collapse
|
3
|
Steffensen LB, Kavan S, Jensen PS, Pedersen MK, Bøttger SM, Larsen MJ, Dembic M, Bergman O, Matic L, Hedin U, Andersen LVB, Lindholt JS, Houlind KC, Riber LP, Thomassen M, Rasmussen LM. Mutational landscape of atherosclerotic plaques reveals large clonal cell populations. JCI Insight 2025; 10:e188281. [PMID: 40198128 DOI: 10.1172/jci.insight.188281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/04/2025] [Indexed: 04/10/2025] Open
Abstract
The notion of clonal cell populations in human atherosclerosis has been suggested but not demonstrated. Somatic mutations are used to define cellular clones in tumors. Here, we characterized the mutational landscape of human carotid plaques through whole-exome sequencing to explore the presence of clonal cell populations. Somatic mutations were identified in 12 of 13 investigated plaques, while no mutations were detected in 11 non-atherosclerotic arteries. Mutated clones often constituted over 10% of the sample cell population, with genes related to the contractile apparatus enriched for mutations. In carriers of clonal hematopoiesis of indeterminate potential (CHIP), hematopoietic clones had infiltrated the plaque tissue and constituted substantial fractions of the plaque cell population alongside locally expanded clones. Our findings establish somatic mutations as a common feature of human atherosclerosis and demonstrate the existence of mutated clones expanding locally, as well as CHIP clones invading from the circulation. While our data do not support plaque monoclonality, we observed a pattern suggesting the coexistence of multiple mutated clones of considerable size spanning different regions of plaques. Mutated clones are likely to be relevant to disease development, and somatic mutations will serve as a convenient tool to uncover novel pathological processes of atherosclerosis in future studies.
Collapse
Affiliation(s)
- Lasse Bach Steffensen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
| | - Stephanie Kavan
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Clinical Biochemistry and Pharmacology, and
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Pia Søndergaard Jensen
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Clinical Biochemistry and Pharmacology, and
| | - Matilde Kvist Pedersen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
| | - Steffen Møller Bøttger
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
| | - Martin Jakob Larsen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
- Department of Clinical Research, and
| | - Maja Dembic
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
- Department of Clinical Research, and
- Department of Mathematics and Computer Science (IMADA), University of Southern Denmark, Odense, Denmark
| | - Otto Bergman
- Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Lars van Brakel Andersen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
| | - Jes Sanddal Lindholt
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Cardiothoracic Surgery, Odense University Hospital, Odense, Denmark
| | | | - Lars Peter Riber
- Department of Cardiothoracic Surgery, Odense University Hospital, Odense, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
| | - Lars Melholt Rasmussen
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Clinical Biochemistry and Pharmacology, and
| |
Collapse
|
4
|
Liang X, Zhang J, Yu J, Zhao J, Yang S. Quercetin ameliorates ox-LDL-induced cellular senescence of aortic endothelial cells and macrophages by p16/p21, p53/SERPINE1, and AMPK/mTOR pathways. Eur J Med Res 2025; 30:359. [PMID: 40319296 PMCID: PMC12049051 DOI: 10.1186/s40001-025-02562-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/06/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Atherosclerosis (AS), a chronic inflammatory disease of the arterial wall, remains a dominant cause of death and disability globally. Quercetin has been evidenced to be effective against AS, but the exact mechanisms are still largely unclear. METHODS Oxidized low-density lipoprotein (ox-LDL)-induced human aortic endothelial cells (HAECs) and mouse RAW264.7 macrophages were established, with quercetin treatment or p16, p21 or SERPINE1 siRNA transfection. Cellular senescence was assessed by SA-β-gal staining and detection of cellular senescence markers. Cell cycle, apoptosis and intracellular ROS were detected by flow cytometry, with cell proliferation by CCK-8. Lipid accumulation was assessed utilizing oil red O staining. Through transmission electron microscope, autophagosomes and mitochondria were investigated, with detection of autophagy markers. Finally, AS models of ApoE-/- mice were established through feeding high-fat diet, and the effect of quercetin on alleviating AS progression was investigated. RESULTS Quercetin protected HAECs from ox-LDL-elicited senescent phenotype, growth arrest and apoptosis and promoted cell viability in a concentration-dependent fashion. Furthermore, quercetin alleviated ox-LDL-elicited cellular senescence, ROS and lipid accumulation in macrophages. In ox-LDL-induced HAECs or/and macrophages, quercetin down-regulated the expression of p16, p21, p53 and SERPINE1, elevated p-AMPK/AMPK levels and decreased p-mTOR/mTOR levels, and these effects of quercetin were ameliorated by SERPINE1 knockdown. In AS mouse models, quercetin treatment alleviated AS progression. CONCLUSION Our findings proposed a novel anti-atherosclerotic mechanism of quercetin by mitigating ox-LDL-elicited senescent phenotype of aortic endothelial cells and macrophages by regulating p16/p21, p53/SERPINE1, and AMPK/mTOR pathways.
Collapse
Affiliation(s)
- Xiao Liang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167, Mannheim, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, 68167, Mannheim, Germany
| | - Jingyuan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167, Mannheim, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, 68167, Mannheim, Germany
| | - Jiangbo Yu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Jiyi Zhao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Shusen Yang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
5
|
Guo B, Wen X, Yu S, Yang J. Single-cell sequencing reveals PHLDA1-positive smooth muscle cells promote local invasion in head and neck squamous cell carcinoma. Transl Oncol 2025; 55:102301. [PMID: 40132389 PMCID: PMC11985064 DOI: 10.1016/j.tranon.2025.102301] [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: 11/13/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Smooth muscle cells within the tumor microenvironment play a crucial role in cancer progression. However, their involvement in the local invasion of head and neck squamous cell carcinoma remains poorly understood. In this research, we aim to investigate the role of smooth muscle cells-mediated cell interactions in facilitating the local invasion of head and neck squamous cell carcinoma. METHODS Single-cell sequencing data from the public databases GSE164690 and GSE181919 were utilized to identify a specific smooth muscle cells cluster. Smooth muscle cells were isolated from tumor microenvironment of head and neck squamous cell carcinoma. PHLDA1 expression in smooth muscle cells was assessed through immunofluorescence staining. The role of THBS1 was investigated through in vitro studies. RESULTS PHLDA1-positive smooth muscle cells were significantly enriched in head and neck squamous cell carcinoma. PHLDA1 promoted the expression of THBS1 in smooth muscle cells. In vitro, THBS1 facilitated head and neck squamous cell carcinoma migration and invasion through SDC1 receptor. CONCLUSION PHLDA1-positive smooth muscle cells play a critical role in head and neck squamous cell carcinoma invasion through THBS1. Targeting PHLDA1-positive smooth muscle cells or THBS1 may offer a promising therapeutic approach for head and neck squamous cell carcinoma treatment.
Collapse
Affiliation(s)
- Bing Guo
- Department of Burns and Plastic Surgery, Institute of Traumatic Medicine and Department of Plastic Surgery and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xutao Wen
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, School of Medicine, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Jiao Tong University, Shanghai, China
| | - Shun Yu
- Department of Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Jun Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Wang SY, Chen YS, Jin BY, Bilal A. The cGAS-STING pathway in atherosclerosis. Front Cardiovasc Med 2025; 12:1550930. [PMID: 40351606 PMCID: PMC12062000 DOI: 10.3389/fcvm.2025.1550930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Atherosclerosis (AS), a chronic inflammatory disease, remains a leading contributor to cardiovascular morbidity and mortality. Recent studies highlight the critical role of the cGAS-STING pathway-a key innate immune signaling cascade-in driving AS progression. This pathway is activated by cytoplasmic DNA from damaged cells, thereby triggering inflammation and accelerating plaque formation. While risk factors such as aging, obesity, smoking, hypertension, and diabetes are known to exacerbate AS, emerging evidence suggests that these factors may also enhance cGAS-STING pathway, which amplifies inflammatory responses. Targeting this pathway offers a promising therapeutic strategy to reduce the burden of cardiovascular diseases (CVD). In this review, we summarize the mechanisms of the cGAS-STING pathway, explore its role in AS, and evaluate potential inhibitors as future therapeutic candidates. By integrating current knowledge, we aim to provide insights for developing novel treatments to mitigate AS and CVD burden.
Collapse
Affiliation(s)
- Si-yu Wang
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Yu-shan Chen
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Bo-yuan Jin
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Ahmad Bilal
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
7
|
Du X, Huang J, Zhao C, Hu Z, Zhang L, Xu Z, Liu X, Li X, Zhang Z, Guo S, Yin T, Wang G. Retrospective perspectives and future trends in nanomedicine treatment: from single membranes to hybrid membranes. NANOSCALE 2025; 17:9738-9763. [PMID: 40136036 DOI: 10.1039/d4nr04999c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
At present, various diseases seriously threaten human life and health, and the development of nanodrug delivery systems has brought about a turnaround for traditional drug treatments, with nanoparticles being precisely targeted to improve bioavailability. Surface modification of nanoparticles can prolong blood circulation time and enhance targeting ability. The application of cell membrane-coated nanoparticles further improves their biocompatibility and active targeting ability, providing new hope for the treatment of various diseases. Various types of cell membrane biomimetic nanoparticles have gradually attracted increasing attention due to their unique advantages. However, the pathological microenvironment of different diseases is complex and varied, and the single-cell membrane has several limitations because a single functional property cannot fully meet the requirements of disease treatment. Hybrid cell membranes integrate the advantages of multiple biological membranes and have become an emerging research hotspot. This review summarizes the application of cell membrane biomimetic nanoparticles in the treatment of various diseases and discusses the advantages, challenges and future development of biomimetic nanoparticles. We propose that the fusion of multiple membranes may be a reasonable trend in the future to provide some ideas and directions for the treatment of various diseases.
Collapse
Affiliation(s)
- Xinya Du
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Junyang Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Chuanrong Zhao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Ziqiu Hu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | | | - Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xiaoying Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xinglei Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Zhengcai Zhang
- Lepu Medical Technology (Beijing) Co., Ltd, Beijing, China
| | - Songtao Guo
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| |
Collapse
|
8
|
Zaina S. The functional significance of vascular DNA hypermethylation in atherosclerosis: a historical perspective. Front Pharmacol 2025; 16:1562674. [PMID: 40303930 PMCID: PMC12037548 DOI: 10.3389/fphar.2025.1562674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
A decade ago, independent mechanistic and descriptive epigenomics data demonstrated for the first time that vascular DNA hypermethylation is a landmark of and causal factor in human and murine atherosclerosis. Since then, a flurry of converging evidence has assigned a prominent role to vascular DNA hypermethylation across the natural history of cardiovascular disease (CVD), from the exposure to risk factors, to the onset and progression of the atheroma. DNA hypermethylation is induced by and mediates the metabolic outcomes of high-fat diets and CVD risk-enhancing lipids in several models. Early-stage atheroma DNA is hypermethylated compared to normal adjacent tissue, and that trend is amplified as the atheroma progresses. That evidence has resulted in a strong interest for epigenetic drugs in CVD. Crucially, the DNA methylation inhibitor azacytidine has been singled out as a potent guardian of the contractile, anti-atherogenic phenotype of smooth muscle cells (SMC). Those findings are gaining relevance, as the antiatherogenic effects of the anticancer drugs azacytidine and decitabine fit into the recently revived hypothesis that the atheroma is a SMC-driven cancer-like mass. Finally, this 10-year anniversary has been marked by the first report that nanoparticles loaded with a DNA methyltransferase inhibitor drug are anti-inflammatory and inhibit murine atherosclerosis. Exciting work lies ahead to assess whether DNA hypermethylation is a practical and effective target to prevent or cure human atherosclerosis.
Collapse
Affiliation(s)
- Silvio Zaina
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, Leon, Mexico
| |
Collapse
|
9
|
Lu W, Zhang Z, Qiao G, Zou G, Li G. Immune Regulation and Disulfidptosis in Atherosclerosis Influence Disease Progression and Therapy. Biomedicines 2025; 13:926. [PMID: 40299531 PMCID: PMC12025079 DOI: 10.3390/biomedicines13040926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Atherosclerosis is a progressive and complex vascular pathology characterized by cellular heterogeneity, metabolic dysregulation, and chronic inflammation. Despite extensive research, the intricate molecular mechanisms underlying its development and progression remain incompletely understood. Methods: Single-cell RNA sequencing (scRNA-seq) was employed to conduct a comprehensive mapping of immune cell enrichment and interactions within atherosclerotic plaques, aiming to investigate the cellular and molecular complexities of these structures. This approach facilitated a deeper understanding of the heterogeneities present in smooth muscle cells, which were subsequently analyzed using pseudotime trajectory analysis to monitor the developmental trajectories of smooth muscle cell (SMC) subpopulations. An integrative bioinformatics approach, primarily utilizing Weighted Gene Co-expression Network Analysis (WGCNA) and machine learning techniques, identified Cathepsin C (CTSC), transforming growth factor beta-induced protein (TGFBI), and glia maturation factor-γ (GMFG) as critical biomarkers. A diagnostic risk score model was developed and rigorously tested through Receiver Operating Characteristic analysis. To illustrate the functional impact of CTSC on the regulation of plaque formation and SMC viability, both in vitro and in vivo experimental investigations were conducted. Results: An analysis revealed SMCs identified as the most prominent cellular type, exhibiting the highest density of disulfidptosis. Pseudotime trajectory analysis illuminated the dynamic activation pathways in SMCs, highlighting their significant role in plaque development and instability. Further characterization of macrophage subtypes demonstrated intercellular communication with SMCs, which exhibited specific signaling pathways, particularly between the proximal and core areas of plaques. The integrated diagnostic risk score model, which incorporates CTSC, TGFBI, and GMFG, proved to be highly accurate in distinguishing high-risk patients with elevated immune responses and systemic inflammation. Knockdown experiments of CTSC conducted in vitro revealed enhanced SMC survival rates, reduced oxidative stress, and inhibited apoptosis, while in vivo experiments confirmed a decrease in plaque burden and improvement in lipid profiles. Conclusions: This study emphasizes the significance of disulfidptosis in the development of atherosclerosis and identifies CTSC as a potential therapeutic target for stabilizing plaques by inhibiting SMC apoptosis and oxidative damage. Additionally, the risk score model serves as a valuable diagnostic tool for identifying high-risk patients and guiding precision treatment strategies.
Collapse
Affiliation(s)
| | - Zhidong Zhang
- Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 451460, China; (W.L.); (G.Q.); (G.Z.); (G.L.)
| | | | | | | |
Collapse
|
10
|
Zhang BC, Zhu WY, Wang SN, Zhu MM, Ma H, Dong L, Yang XX, Ma CR, Ma LK, Chen YL. Colchicine reduces neointima formation and VSMC phenotype transition by modulating SRF-MYOCD activation and autophagy. Acta Pharmacol Sin 2025; 46:951-963. [PMID: 39663419 PMCID: PMC11950430 DOI: 10.1038/s41401-024-01438-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/17/2024] [Accepted: 11/17/2024] [Indexed: 12/13/2024]
Abstract
Vascular smooth muscle cell (VSMC) phenotype transformation significantly contributes to vascular intimal hyperplasia. However, effective preventive and therapeutic measures are lacking. Colchicine, a binary alkaloid derived from Colchicum autumnale, is traditionally used for treating inflammatory diseases. Its role in neointima formation is not fully understood. Here, we investigated the role of colchicine in vascular intimal hyperplasia. We found that colchicine significantly reduced vascular intimal hyperplasia in an animal model at 7, 14, and 28 days post carotid artery ligation and increased the number of contractile-phenotype VSMCs (SMA-positive cells) in the neointimal areas. In vitro experiments demonstrated that colchicine facilitated the transition of VSMCs from a proliferative phenotype to a contractile phenotype. Additionally, colchicine attenuated PDGF-BB-induced phenotypic conversion and upregulated the expression of serum response factor (SRF) and myocardin (MYOCD). Further molecular mechanistic studies revealed that colchicine inhibited the expression of forkhead box protein O3A (FOXO3A) to increase the activation of the SRF‒MYOCD complex. FOXO3A can bind to MSX1/2, thereby inhibiting the expression of SRF-MYOCD and contractile genes. Moreover, colchicine maintains vascular homeostasis and stabilizes the contractile phenotype by affecting the expression of autophagy-related genes (LC3II, p62, and Beclin-1) induced by FOXO3A. Additionally, colchicine inhibited monocyte/macrophage infiltration and inflammatory cytokine expression. In summary, this study suggests that colchicine inhibits vascular intimal hyperplasia by modulating FOXO3A-mediated SRF-MYOCD activation and autophagy, providing new insights for future therapeutic approaches targeting occlusive vascular diseases.
Collapse
Affiliation(s)
- Bu-Chun Zhang
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Wen-Ya Zhu
- Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Sheng-Nan Wang
- Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Meng-Meng Zhu
- Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Hui Ma
- Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Liang Dong
- Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China
| | - Xiao-Xiao Yang
- Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China.
| | - Chuan-Rui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China.
| | - Li-Kun Ma
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Yuan-Li Chen
- Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China.
| |
Collapse
|
11
|
Ma Y, Shi Y, Lun J, Wang M, Zhang C, Li X, Yi Q, Cao X, Hu Z, Yang W, Cai H. Isoforskolin, adenylate cyclase agonist, inhibits endothelial-to-mesenchymal transition in atherosclerosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156520. [PMID: 39986229 DOI: 10.1016/j.phymed.2025.156520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/04/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Atherosclerosis is a common chronic disease characterized by the formation of atheromatous plaques and endothelial dysfunction. Endothelial-to-mesenchymal transition (EndMT) has been identified as a crucial driver of atherosclerosis, with TGF-β serving as a pivotal mediator of EndMT. Isoforskolin (ISOF), derived from the plant Coleus forskohlii, is an effective activator of adenylyl cyclase (AC). AC can catalyze the production of cyclic adenosine monophosphate (cAMP), mediating various biological functions. Several phosphodiesterase (PDE) inhibitors that degrade cAMP have been clinically utilized in the treatment of atherosclerosis. However, the evidence regarding the efficacy and mechanisms of AC agonists in the treatment of atherosclerosis remains inadequate. PURPOSE In this study, our primary objective was to examine the therapeutic impact of ISOF on atherosclerosis and elucidate its potential mechanisms. METHODS Male ApoE-/- rats were fed a high-fat diet for 18 weeks and then administered ISOF by gavage continuously for 12 weeks. A cell model was established by injuring mouse aortic endothelial cells (MAECs) with ox-LDL. Oil Red O staining and Masson staining were used to assess the plaque area and content of collagen. Aortic vasodilatory function was analyzed using the DMT Myograph system. Immunofluorescence was used to determine the localization of CD31 and α-SMA. Proteomics analysis was utilized to identify potential pharmacological mechanisms of ISOF. Quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to detect the mRNA expression levels of AC (1-10) in aortic tissue. AC activity and cAMP concentration were detected using specific kits. The CCK-8 assay, wound-healing, and transwell assays were used to measure cell viability and proliferation. Mechanistically, western blot analysis was used to detect candidate protein expression levels. Finally, the pharmacological knockdown of AC5 was employed to clarify the potential mechanism of ISOF. RESULTS ISOF effectively inhibited atherosclerotic plaque progression and improved aortic vasodilatory function in ApoE-/- rats fed a high-fat diet. At the cellular level, ISOF enhanced cell viability and proliferation of MAECs compromised by ox-LDL. These phenotypic improvements were attributed to the inhibitory effect of ISOF on endothelial-to-mesenchymal transition (EndMT); ISOF increased the expression of endothelial markers such as CD31 and E-cadherin while decreasing the expression of mesenchymal markers, including N-cadherin and α-SMA. Mechanistically, and consistent with the results of proteomic analysis, ISOF markedly inhibited the TGF-β/Smad3 signaling pathway both in vivo and in vitro. Furthermore, the expression levels of adenylyl cyclase 5 (AC5) were significantly higher than those of other AC isoforms in rat aorta. ISOF might upregulate the expression of AC5, subsequently activating downstream protein kinase A (PKA) but not cAMP-activated exchange protein-1 (EPAC1). However, when AC5 was silenced, the effects of ISOF on EndMT and the cAMP/PKA/TGF-β pathway were effectively abolished. CONCLUSION The study showed that ISOF effectively combats atherosclerosis by inhibiting EndMT through the regulation of the AC5-dependent cAMP/PKA/TGF-β axis. This finding suggests a potential promising therapeutic strategy for treating atherosclerosis.
Collapse
Affiliation(s)
- Yiming Ma
- Heart Center, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Yunke Shi
- Heart Center, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Jinping Lun
- Heart Center, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Mingqiang Wang
- Heart Center, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Chaoyue Zhang
- Heart Center, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Xianbin Li
- Heart Center, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Qian Yi
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Xingyu Cao
- Heart Center, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Zhao Hu
- Geriatric Cardiology Department, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China.
| | - Weimin Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China.
| | - Hongyan Cai
- Heart Center, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China.
| |
Collapse
|
12
|
Hu Y, Zhou Y, Dai N, Song S, Zhao X, Zhao Y, Cheng L, Lu H, Ge J. Enhancing panvascular medicine: unveiling the nexus of pan-cardio-oncology and expanding therapeutic frontiers. Sci Bull (Beijing) 2025; 70:798-800. [PMID: 39828464 DOI: 10.1016/j.scib.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Affiliation(s)
- Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Cardiology, Shanghai Geriatric Medical Center, Shanghai 201100, China
| | - Neng Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| | - Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| | - Xin Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| | - Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China.
| | - Leilei Cheng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China.
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Cardiology, Shanghai Geriatric Medical Center, Shanghai 201100, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
13
|
Sakuta K, Iguchi Y. Acute Ischemic Stroke: Significance of Multimodal Pre-operative Prediction of Intracranial Atherosclerosis-Related Large Vessel Occlusion. Intern Med 2025:5350-25. [PMID: 40159166 DOI: 10.2169/internalmedicine.5350-25] [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] [Indexed: 04/02/2025] Open
Abstract
When large-vessel occlusion (LVO) occurs in acute ischemic stroke, accurate differentiation of intracranial atherosclerosis-related occlusion (ICAD-O) from embolism-related occlusion (EMB-O) is critical for guiding treatment decisions. This review focuses on the preoperative prediction of ICAD-O and emphasizes the practical clinical and imaging factors. ICAD-O is often associated with younger age, male sex, and vascular risk factors, such as hypertension and diabetes, and the absence of atrial fibrillation is a key characteristic of its differentiation. Imaging findings, including truncal-type occlusion and non-culprit stenosis, further aid in the identification of ICAD-O. Predictive scales integrating clinical and imaging data, such as the ISAT, REMIT, ABC2D, and ATHE scores, provide structured approaches for distinguishing ICAD-O from EMB-O. By reviewing these predictive tools and findings, this review aims to enhance the accuracy and efficiency of the preoperative diagnosis, supporting better-informed clinical decision-making for stroke patients with LVO.
Collapse
Affiliation(s)
- Kenichi Sakuta
- Department of Neurology, The Jikei University School of Medicine, Japan
| | - Yasuyuki Iguchi
- Department of Neurology, The Jikei University School of Medicine, Japan
| |
Collapse
|
14
|
Li S, Du Y, Chen G, Mao Y, Zhang W, Kang M, Zhu S, Wang D. Protocatechuic Acid Attenuates Inflammation in Macrophage-like Vascular Smooth Muscle Cells in ApoE -/- Mice. Nutrients 2025; 17:1090. [PMID: 40292571 PMCID: PMC11944442 DOI: 10.3390/nu17061090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Non-resolving inflammation in macrophage-like cells (MLCs) transdifferentiated from vascular smooth muscle cells and monocyte-derived macrophages aggravates atherosclerosis. We previously showed that polyphenolic protocatechuic acid (PCA) could reduce inflammation burden in monocyte-derived macrophages; however, it remains unknown how this compound affects MLCs inflammation. Methods: MLCs from the transdifferentiation of vascular smooth muscle cells induced by cholesterol and 30-week-old male ApoE-/- mice fed a semi-purified AIN-93G diet containing either 0.003% (wt:wt) of PCA for a duration of 20 weeks were used to examine the impact of PCA on the inflammatory response of MLCs. Results: Physiologically achievable doses of PCA (0.25-1 μM) dose-dependently inhibited lipopolysaccharide-induced NF-κB activation and simultaneously reduced pro-inflammatory cytokine levels. Mechanistically, this effect was mediated by effecting exportin-1 function, promoting nuclear export of phosphorylated-p65, independent of NF-κB kinase inhibitor α/β/γ, NF-κB inhibitor α, or importin-mediated nuclear import of p-p65. PCA reduced the nucleocytoplasmic ratio of exportin-1 (44%) without altering its abundance. Importantly, dietary supplementation with PCA reduced interleukin-1β content within MLCs in atherosclerotic plaques of ApoE-/- mice. In addition, dietary PCA reduced MLCs content in atherosclerotic plaques. Conclusions: PCA could attenuate inflammatory response in MLCs by targeting exportin-1 and also could inhibit the transdifferentiation of vascular smooth muscle cells into MLCs within atherosclerotic plaques, which might promote the translation from preclinical studies to clinical trials in patients with atherosclerosis.
Collapse
MESH Headings
- Animals
- Hydroxybenzoates/pharmacology
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/cytology
- Apolipoproteins E/genetics
- Mice
- Macrophages/drug effects
- Macrophages/metabolism
- Inflammation/drug therapy
- Atherosclerosis/drug therapy
- NF-kappa B/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Mice, Knockout, ApoE
- Mice, Knockout
- Cytokines/metabolism
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Shuangshuang Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Northern Campus, Guangzhou 510080, China; (S.L.); (Y.D.); (G.C.); (Y.M.); (W.Z.); (M.K.); (S.Z.)
| | - Yushi Du
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Northern Campus, Guangzhou 510080, China; (S.L.); (Y.D.); (G.C.); (Y.M.); (W.Z.); (M.K.); (S.Z.)
| | - Guanyu Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Northern Campus, Guangzhou 510080, China; (S.L.); (Y.D.); (G.C.); (Y.M.); (W.Z.); (M.K.); (S.Z.)
| | - Yihui Mao
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Northern Campus, Guangzhou 510080, China; (S.L.); (Y.D.); (G.C.); (Y.M.); (W.Z.); (M.K.); (S.Z.)
| | - Wenyu Zhang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Northern Campus, Guangzhou 510080, China; (S.L.); (Y.D.); (G.C.); (Y.M.); (W.Z.); (M.K.); (S.Z.)
| | - Mengxi Kang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Northern Campus, Guangzhou 510080, China; (S.L.); (Y.D.); (G.C.); (Y.M.); (W.Z.); (M.K.); (S.Z.)
| | - Shasha Zhu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Northern Campus, Guangzhou 510080, China; (S.L.); (Y.D.); (G.C.); (Y.M.); (W.Z.); (M.K.); (S.Z.)
| | - Dongliang Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Northern Campus, Guangzhou 510080, China; (S.L.); (Y.D.); (G.C.); (Y.M.); (W.Z.); (M.K.); (S.Z.)
- Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Sun Yat-sen University, Guangzhou 518107, China
| |
Collapse
|
15
|
Liu Z, Wang Y, Li L, Liu L, Li Y, Li Z, Xie Y, Yu F. SNAI2, a potential crossing point between cancer and cardiovascular disease. FASEB J 2025; 39:e70459. [PMID: 40059450 DOI: 10.1096/fj.202500198r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 05/13/2025]
Abstract
Cancer and cardiovascular disease remain the leading causes of morbidity and mortality worldwide, and the two separate disease entities share several similarities and possible interactions. Patients with cancer may have underlying cardiovascular disease, which is often exacerbated by the stress of tumor growth or treatment. At the same time, cardiotoxicity induced by anti-cancer therapies or the malignant process itself can lead to new cardiovascular diseases. Efforts have been made to find a rational explanation for this phenomenon. As a classical tumor-promoting factor, we notice that SNAI2 simultaneously plays an important pathogenic role in cardiovascular diseases. Moreover, there are several striking parallels in the mechanisms of cancer and CVD, such as shared risk factors (e.g., smoking and diabetes), cellular phenotypic switching, and metabolic remodeling, all of which are mediated by SNAI2. This review aims to summarize SNAI2's role in the core mechanisms linking cancer and CVD, as well as explore therapeutic approaches targeting SNAI2 and also seeks to provide insights into the common mechanisms underlying both cancer and CVD.
Collapse
Affiliation(s)
- Zihao Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yingzi Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lei Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Linlu Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuhao Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhixin Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yucheng Xie
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fengxu Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular Remodeling and Dysfunction Key Laboratory of Luzhou, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
16
|
Miao M, Liu X, Zhang H, Dai H. Immuno-inflammatory mechanisms in cardio-oncology: new hopes for immunotargeted therapies. Front Oncol 2025; 15:1516977. [PMID: 40182041 PMCID: PMC11966441 DOI: 10.3389/fonc.2025.1516977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
Cardio-oncology is an emerging interdisciplinary field concerned with cancer treatment-related cardiovascular toxicities (CTR-CVT) and concomitant cardiovascular diseases (CVD) in patients with cancer. Inflammation and immune system dysregulation are common features of tumors and cardiovascular disease (CVD). In addition to the mutual exacerbating effect through inflammation, tumor treatments, including immunotherapy, chemotherapy, radiation therapy, and targeted therapy, may induce immune inflammatory reactions leading to cardiovascular damage. Cancer immunotherapy is currently a new method of cancer treatment. Immunotherapeutic agents, such as immune checkpoint inhibitors (ICIs), chimeric antigen receptor T cell immunotherapy (CAR-T), mRNA vaccines, etc., can induce anti-tumor effects by enhancing the host immune response to eliminate tumor cells. They have achieved remarkable therapeutic efficacy in clinical settings but lead to many immune-related adverse events (irAEs), especially CTR-CVT. Establishing specific evaluation, diagnostic, and monitoring criteria (e.g., inflammatory biomarkers) for both immunotherapy and anti-inflammatory therapy-related cardiovascular toxicity is vital to guide clinical practice. This article explores the role of immune response and inflammation in tumor cardiology, unravels the underlying mechanisms, and provides improved methods for monitoring and treating in CTR-CVT in the field of cardio-oncology.
Collapse
Affiliation(s)
- Meiqi Miao
- Department of Cardiology, Kunshan Hospital of Chinese Medicine, Kunshan, China
| | - Xinxin Liu
- Postdoctoral Mobile Station, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- The Innovation Base, Mudanjiang Collaborative Innovation Center for the Development and Application of Northern Medicinal Resources, Mudanjiang, China
| | - Han Zhang
- Department of Cardiology, Yan’an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hailong Dai
- Department of Cardiology, Yan’an Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
17
|
Wang Z, Sun W, Zhang K, Ke X, Wang Z. New insights into the relationship of mitochondrial metabolism and atherosclerosis. Cell Signal 2025; 127:111580. [PMID: 39732307 DOI: 10.1016/j.cellsig.2024.111580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Atherosclerotic cardiovascular and cerebrovascular diseases are the number one killer of human health. In view of the important role of mitochondria in the formation and evolution of atherosclerosis, our manuscript aims to comprehensively elaborate the relationship between mitochondria and the formation and evolution of atherosclerosis from the aspects of mitochondrial dynamics, mitochondria-organelle interaction (communication), mitochondria and cell death, mitochondria and vascular smooth muscle cell phenotypic switch, etc., which is combined with genome, transcriptome and proteome, in order to provide new ideas for the pathogenesis of atherosclerosis and the diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Zexun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang 212001, China
| | - Wangqing Sun
- Department of Radiology, Yixing Tumor Hospital, Yixing 214200, China
| | - Kai Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Xianjin Ke
- Department of Neurology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
18
|
Qin YS, Yi J, Chen YJ, Zhang W, Tang SF. Recent Advances in Micro/Nanomotor for the Therapy and Diagnosis of Atherosclerosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11443-11468. [PMID: 39648908 DOI: 10.1021/acsami.4c15165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Atherosclerotic cardiovascular disease poses a significant global public health threat with a high incidence that can result in severe mortality and disability. The lack of targeted effects from traditional therapeutic drugs on atherosclerosis may cause damage to other organs and tissues, necessitating the need for a more focused approach to address this dilemma. Micro/nanomotors are self-propelled micro/nanoscale devices capable of converting external energy into autonomous movement, which offers advantages in enhancing penetration depth and retention while increasing contact area with abnormal sites, such as atherosclerotic plaque, inflammation, and thrombosis, within blood vessel walls. Recent studies have demonstrated the crucial role micro/nanomotors play in treating atherosclerotic cardiovascular disease. Hence, this review highlights the recent progress of micro/nanomotor technology in atherosclerotic cardiovascular disease, including the effective promotion of micro/nanomotors in the circulatory system, overcoming hemorheological barriers, targeting the atherosclerotic plaque microenvironment, and targeting intracellular drug delivery, to facilitate atherosclerotic plaque localization and therapy. Furthermore, we also describe the potential application of micro/nanomotors in the imaging of vulnerable plaque. Finally, we discuss key challenges and prospects for treating atherosclerotic cardiovascular disease while emphasizing the importance of designing individualized management strategies specific to its causes and microenvironmental factors.
Collapse
Affiliation(s)
- Yu-Sheng Qin
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| | - Juan Yi
- Department of Laboratory Medicine, Liuzhou Traditional Chinese Medical Hospital, The Third Affiliated Hospital of Guangxi University of Chinese Medicine, Liuzhou 545006, China
| | - Yan-Jun Chen
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei Zhang
- Department of Radiology, Liuzhou People's Hospital, Liuzhou 545006, China
| | - Shi-Fu Tang
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| |
Collapse
|
19
|
Fang R, Yang Q, Wu D, Zhao J, Xu S. Fibrinopeptide a promotes the proliferation and migration of vascular smooth muscle cells by regulating the integrin αVβ3/PI3K/AKT signaling pathway. Mol Biol Rep 2025; 52:205. [PMID: 39907881 DOI: 10.1007/s11033-025-10314-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Atherosclerosis is characterized by subintimal proliferation and migration of vascular smooth muscle cells (VSMCs) in response to stimuli such as coagulation and inflammatory factors. Fibrinopeptide A (FPA), a biomarker for coagulation system activation, is elevated in patients with ischemic heart disease. However, its role in the pathophysiology of cardiovascular disorders remains unclear. This study aimed to investigate the impact of FPA on VSMCs proliferation and migration and elucidate the underlying molecular mechanisms. METHODS AND RESULTS Transcriptome sequencing and bioinformatics analysis were employed to elucidate molecular pathways. Scratch wound and Transwell assays were performed to evaluate cell migration capacity. Molecular expression patterns were assessed using immunofluorescence, real-time quantitative PCR, and Western blot assays. The differentially expressed genes (DEGs) in the FPA-treated VSMCs were enriched in the cell cycle and PI3K/AKT signaling pathway. FPA treatment enhanced VSMCs' migratory capacity and upregulated integrin αVβ3, PI3K, P-AKT, AKT, Cyclin D1, and PCNA expression. The integrin αVβ3 inhibitor Cyclo-RGDfk effectively suppressed VSMCs migration and reduced the expression levels of these genes in FPA-stimulated VSMCs. CONCLUSIONS These results suggested that FPA promotes the proliferation and migration of VSMCs by regulating the PI3K/AKT signaling pathway through integrin αVβ3.
Collapse
Affiliation(s)
- Rourou Fang
- Department of School of Public Health, Shaanxi University of Chinese Medicine, Xixian New Area, 712046, China
| | - Qifan Yang
- Department of School of Public Health, Shaanxi University of Chinese Medicine, Xixian New Area, 712046, China
| | - Dongdong Wu
- Department of School of Public Health, Shaanxi University of Chinese Medicine, Xixian New Area, 712046, China
| | - Jing Zhao
- College of Acupuncture and Tuina, Shaanxi University of Chinese Medicine, Xixian New Area, 712046, China.
| | - Shouzhu Xu
- Department of School of Public Health, Shaanxi University of Chinese Medicine, Xixian New Area, 712046, China.
| |
Collapse
|
20
|
Lambert J, Jørgensen HF. Epigenetic regulation of vascular smooth muscle cell phenotypes in atherosclerosis. Atherosclerosis 2025; 401:119085. [PMID: 39709233 DOI: 10.1016/j.atherosclerosis.2024.119085] [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: 07/16/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/23/2024]
Abstract
Vascular smooth muscle cells (VSMCs) in adult arteries maintain substantial phenotypic plasticity, which allows for the reversible cell state changes that enable vascular remodelling and homeostasis. In atherosclerosis, VSMCs dedifferentiate in response to lipid accumulation and inflammation, resulting in loss of their characteristic contractile state. Recent studies showed that individual, pre-existing VSMCs expand clonally and can acquire many different phenotypes in atherosclerotic lesions. The changes in gene expression underlying this phenotypic diversity are mediated by epigenetic modifications which affect transcription factor access and thereby gene expression dynamics. Additionally, epigenetic mechanisms can maintain cellular memory, potentially facilitating reversion to the contractile state. While technological advances have provided some insight, a comprehensive understanding of how VSMC phenotypes are governed in disease remains elusive. Here we review current literature in light of novel insight from studies at single-cell resolution. We also discuss how lessons from epigenetic studies of cellular regulation in other fields could help in translating the potential of targeting VSMC phenotype conversion into novel therapies in cardiovascular disease.
Collapse
Affiliation(s)
- Jordi Lambert
- Section of Cardiorespiratory Medicine, University of Cambridge, VPD Heart and Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
| | - Helle F Jørgensen
- Section of Cardiorespiratory Medicine, University of Cambridge, VPD Heart and Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
| |
Collapse
|
21
|
Chandler HL, Wheeler J, Escott‐Price V, Murphy K, Lancaster TM. Non-APOE variants predominately expressed in smooth muscle cells contribute to the influence of Alzheimer's disease genetic risk on white matter hyperintensities. Alzheimers Dement 2025; 21:e14455. [PMID: 39737667 PMCID: PMC11848156 DOI: 10.1002/alz.14455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 01/01/2025]
Abstract
INTRODUCTION White matter hyperintensity volumes (WMHVs) are disproportionally prevalent in individuals with Alzheimer's disease (AD), potentially reflecting neurovascular injury. We quantify the association between AD polygenic risk score (AD-PRS) and WMHV, exploring single-nucleotide polymorphisms (SNPs) that are proximal to genes overexpressed in cerebrovascular cell species. METHODS In a UK-Biobank sub-sample (mean age = 64, range = 45-81 years), we associate WMHV with (1) AD-PRS estimated via SNPs across the genome (minus apolipoprotein E [APOE] locus) and (2) AD-PRS estimated with SNPs proximal to specific genes that are overexpressed in cerebrovascular cell species. RESULTS We observed a positive association between non-APOE-AD-PRS and WMHVs. We further demonstrate an association between WMHVs and AD-PRS constructed with SNPs that are proximal to genes over-represented in smooth muscles cells (SMCs; β = 0.135, PFWE < 0.01) and internally replicated (PDISCOVERY+REPLICATION < 0.01). DISCUSSION Common AD genetic risk could explain physiological processes underlying vascular pathology in AD. SMC function may offer a treatment target to prevent WMHV-related AD pathophysiology prior to the onset of symptoms. HIGHLIGHTS Alzheimer's disease (AD) risk factors such as apolipoprotein E (APOE) ε4, link to increased white matter hyperintensity volume (WMHV). WMHVs indicate vascular risk and neurovascular injury in AD. The broader genetic link between AD risk and WMHV is not fully understood. We quantify AD polygenic risk score (PRS) associations with WMHV, excluding APOE. AD-PRS in smooth muscle cells (SMCs) shows a significant association with increased WMHV.
Collapse
Affiliation(s)
- Hannah Louise Chandler
- School of Physics and AstronomyCardiff University Brain Research Imaging Centre (CUBRIC)Cardiff UniversityCardiffUK
| | - Joshua Wheeler
- School of Clinical SciencesUniversity of BristolBristolUK
- Department of PsychologyUniversity of BathBathUK
| | - Valentina Escott‐Price
- Centre for Neuropsychiatric Genetics and GenomicsDepartment of Psychological Medicine and Clinical NeurosciencesCardiff UniversityCardiffUK
| | - Kevin Murphy
- School of Physics and AstronomyCardiff University Brain Research Imaging Centre (CUBRIC)Cardiff UniversityCardiffUK
| | | |
Collapse
|
22
|
Zeng Y, Wu Z, Xiong M, Liang Z, Chen Z, Huang H, Yang H, Chen Q. Piezo1 promotes vibration-induced vascular smooth muscle injury by regulating the NF-κB/p65 axis. Commun Biol 2025; 8:96. [PMID: 39833492 PMCID: PMC11747106 DOI: 10.1038/s42003-025-07524-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Vibration induced damage to the peripheral circulatory system is thought to be an early stage of hand-arm vibration syndrome (HAVS) caused by occupational exposure to hand-transmitted vibration (HTV). This study investigated the mechanisms underlying vibration-induced vascular injury, focusing on the role of Piezo1, a mechanosensitive channel, and its association with the NF-κB/p65 signaling pathway. We demonstrated that vibration exposure leads to Piezo1-mediated upregulation of angiogenic chemokines, including CCL2, CCL5, CXCL1, CXCL2, and CXCL10, through the NF-κB/p65 pathway. To mimic the effects of vibration, a rat vibration model and a cellular vibration model were used. Animal and cellular models showed that vibration-induced vascular dysfunction while increasing Piezo1 expression. Piezo1 knockdown or p65 inhibition attenuated these effects, suggesting a crucial role for the Piezo1-NF-κB/p65 axis in vascular dysfunction. Furthermore, chemokines were identified as potential biomarkers for early diagnosis of HAVS in occupationally exposed individuals. These results highlight Piezo1 and the NF-κB/p65 pathway as potential therapeutic targets for HAVS and underscore the need for further validation in human samples and exploration of additional signaling mechanisms involved in vibration-induced vascular injury.
Collapse
Affiliation(s)
- Yingshan Zeng
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Zhiquan Wu
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Mengtian Xiong
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Zhishan Liang
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Ziyu Chen
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Huimin Huang
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Hongyu Yang
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China.
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China.
| | - Qingsong Chen
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China.
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China.
| |
Collapse
|
23
|
Mey L, Bonaterra GA, Hoffmann J, Schwarzbach H, Schwarz A, Eiden LE, Weihe E, Kinscherf R. PAC1 Agonist Maxadilan Reduces Atherosclerotic Lesions in Hypercholesterolemic ApoE-Deficient Mice. Int J Mol Sci 2024; 25:13245. [PMID: 39769009 PMCID: PMC11675839 DOI: 10.3390/ijms252413245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
A possible involvement of immune- and vasoregulatory PACAP signaling at the PAC1 receptor in atherogenesis and plaque-associated vascular inflammation has been suggested. Therefore, we tested the PAC1 receptor agonist Maxadilan and the PAC1 selective antagonist M65 on plaque development and lumen stenosis in the ApoE-/- atherosclerosis model for possible effects on atherogenesis. Adult male ApoE-/- mice were fed a cholesterol-enriched diet (CED) or standard chow (SC) treated with Maxadilan, M65 or Sham. Effects of treatment on atherosclerotic plaques, lumen stenosis, apoptosis and pro-inflammatory signatures were analyzed in the brachiocephalic trunk (BT). The percentage of Maxadilan treated mice exhibiting plaques under SC and CED was lower than that of Sham or M65 treatment indicating opposite effects of Maxadilan and M65. Maxadilan application inhibited lumen stenosis in SC and CED mice compared to the Sham mice. In spite of increased cholesterol levels, lumen stenosis of Maxadilan-treated mice was similar under CED and SC. In contrast, M65 under SC or CED did not reveal a significant influence on lumen stenosis. Maxadilan significantly reduced the TNF-α-immunoreactive (TNF-α+) area in the plaques under CED, but not under SC. In contrast, the IL-1β+ area was reduced after Maxadilan treatment in SC mice but remained unchanged in CED mice compared to Sham mice. Maxadilan reduced caspase-3 immunoreactive (caspase-3+) in the tunica media under both, SC and CED without affecting lipid content in plaques. Despite persistent hypercholesterolemia, Maxadilan reduces lumen stenosis, apoptosis and TNF-α driven inflammation. Our data suggest that Maxadilan provides atheroprotection by acting downstream of hypercholesterolemia-induced vascular inflammation. This implicates the potential of PAC1-specific agonist drugs against atherosclerosis even beyond statins and PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitors.
Collapse
Affiliation(s)
- Lilli Mey
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany (H.S.); (A.S.); (E.W.); (R.K.)
| | - Gabriel A. Bonaterra
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany (H.S.); (A.S.); (E.W.); (R.K.)
| | - Joy Hoffmann
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany (H.S.); (A.S.); (E.W.); (R.K.)
| | - Hans Schwarzbach
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany (H.S.); (A.S.); (E.W.); (R.K.)
| | - Anja Schwarz
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany (H.S.); (A.S.); (E.W.); (R.K.)
| | - Lee E. Eiden
- Section on Molecular Neuroscience, National Institute of Mental Health Intramural Research Program, 49 Convent Drive, Room 5A38, Bethesda, MD 20892, USA;
| | - Eberhard Weihe
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany (H.S.); (A.S.); (E.W.); (R.K.)
| | - Ralf Kinscherf
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany (H.S.); (A.S.); (E.W.); (R.K.)
| |
Collapse
|
24
|
Song J, Cao C, Wang Z, Li H, Yang L, Kang J, Meng H, Li L, Liu J. Mechanistic insights into the regression of atherosclerotic plaques. Front Physiol 2024; 15:1473709. [PMID: 39628943 PMCID: PMC11611857 DOI: 10.3389/fphys.2024.1473709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
Atherosclerosis is a major contributor to cardiovascular diseases and mortality globally. The progression of atherosclerotic disease results in the expansion of plaques and the development of necrotic cores. Subsequent plaque rupture can lead to thrombosis, occluding blood vessels, and end-organ ischemia with consequential ischemic injury. Atherosclerotic plaques are formed by the accumulation of lipid particles overloaded in the subendothelial layer of blood vessels. Abnormally elevated blood lipid levels and impaired endothelial function are the initial factors leading to atherosclerosis. The atherosclerosis research has never been interrupted, and the previous view was that the pathogenesis of atherosclerosis is an irreversible and chronic process. However, recent studies have found that the progression of atherosclerosis can be halted when patients' blood lipid levels are reversed to normal or lower. A large number of studies indicates that it can inhibit the progression of atherosclerosis lesions and promote the regression of atherosclerotic plaques and necrotic cores by lowering blood lipid levels, improving the repair ability of vascular endothelial cells, promoting the reverse cholesterol transport in plaque foam cells and enhancing the ability of macrophages to phagocytize and clear the necrotic core of plaque. This article reviews the progress of research on the mechanism of atherosclerotic plaque regression. Our goal is to provide guidance for developing better therapeutic approaches to atherosclerosis by reviewing and analyzing the latest scientific findings.
Collapse
Affiliation(s)
- Jianshu Song
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
- Research Institute of Traditional Chinese Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Ce Cao
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Ziyan Wang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Haoran Li
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
- Research Institute of Traditional Chinese Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Lili Yang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Jing Kang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Hongxu Meng
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Lei Li
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Jianxun Liu
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
25
|
Zhu J, Wang Z, Liu C, Shi M, Guo Z, Li Y, Yu R, Wei J. Study on the Anti-Atherosclerotic Mechanisms of Xin-Tong-Tai Granule Through Network Pharmacology, Molecular Docking, and Experimental Validation. J Inflamm Res 2024; 17:8147-8164. [PMID: 39525320 PMCID: PMC11545721 DOI: 10.2147/jir.s490815] [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: 09/11/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background Xin-Tong-Tai Granule (XTTG), a Chinese medicine (CM) formula, has demonstrated significant therapeutic effects on atherosclerosis (AS) in both clinical and experimental settings. Nonetheless, the mechanisms underlying XTTG's efficacy remain largely unexplored. This study aimed to elucidate the mechanisms through which XTTG acts against AS, employing network pharmacology, molecular docking, and experimental validation techniques. Methods Initially, target identification for the main chemical components of XTTG was conducted using database, followed by determining the intersection targets between these compounds and disease. Protein-protein interaction (PPI) network analysis, Gene Ontology (GO) enrichment, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were subsequently utilized to investigate the potential pathways through which XTTG exerts its effects on AS. Molecular docking was done to confirm the binding efficacy of XTTG's active components. Additionally, the effects of XTTG were evaluated in vitro using oxidized low-density lipoprotein (ox-LDL) induced human aortic vascular smooth muscle cells (HAVSMCs) and in vivo in apolipoprotein E gene knockout (ApoE-/-) mice fed a high-fat diet (HFD). Results 229 therapeutic targets were screened for PPI network and enrichment analysis. Notably, the nuclear factor kappa-B (NF-κB) signaling pathway, along with processes related to inflammation and autophagy, were significantly enriched, highlighting their importance. In vitro studies showed that XTTG repressed cell proliferation and lipid droplet aggregation in ox-LDL-induced HAVSMCs. It also decreased the ratio of phosphorylated NF-κB p65/ NF-κB p65, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels, and elevated microtubule-associated protein light chain 3 (LC3) and decreased p62 protein expression. In vivo, XTTG ameliorated blood lipid profiles and aortic pathology in HFD-fed ApoE-/- mice, reduced NF-κB p65 expression and serum levels of TNF-α and IL-6, increased the ratio of LC3II/LC3I while decreasing p62 protein expression. Conclusion XTTG mitigates AS primarily through anti-inflammatory and autophagy-modulating mechanisms, particularly via inhibition of NF-κB p65 expression. These findings underscore the potential of CM in treating AS and support its further clinical exploration.
Collapse
Affiliation(s)
- Junping Zhu
- School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of TCM Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Ziyan Wang
- First Hospital and First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan Universities of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Chengxin Liu
- First Hospital and First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan Universities of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Min Shi
- School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan Universities of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Zhihua Guo
- School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- First Hospital and First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan Universities of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Ya Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Rong Yu
- School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of TCM Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Jiaming Wei
- School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan Universities of Chinese Medicine, Changsha, 410208, People’s Republic of China
| |
Collapse
|
26
|
Yang Q, Liu J, Zhang T, Zhu T, Yao S, Wang R, Wang W, Dilimulati H, Ge J, An S. Exploring shared biomarkers and shared pathways in insomnia and atherosclerosis using integrated bioinformatics analysis. Front Mol Neurosci 2024; 17:1477903. [PMID: 39439987 PMCID: PMC11493776 DOI: 10.3389/fnmol.2024.1477903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Background Insomnia (ISM) is one of the non-traditional drivers of atherosclerosis (AS) and an important risk factor for AS-related cardiovascular disease. Our study aimed to explore the shared pathways and diagnostic biomarkers of ISM-related AS using integrated bioinformatics analysis. Methods We download the datasets from the Gene Expression Omnibus database and the GeneCards database. Weighted gene co-expression network analysis and gene differential expression analysis were applied to screen the AS-related gene set. The shared genes of ISM and AS were obtained by intersecting with ISM-related genes. Subsequently, candidate diagnostic biomarkers were identified by constructing protein-protein interaction networks and machine learning algorithms, and a nomogram was constructed. Moreover, to explore potential mechanisms, a comprehensive analysis of shared genes was carried out, including enrichment analysis, protein interactions, immune cell infiltration, and single-cell sequencing analysis. Results We successfully screened 61 genes shared by ISM and AS, of which 3 genes (IL10RA, CCR1, and SPI1) were identified as diagnostic biomarkers. A nomogram with excellent predictive value was constructed (the area under curve of the model constructed by the biomarkers was 0.931, and the validation set was 0.745). In addition, the shared genes were mainly enriched in immune and inflammatory response regulation pathways. The biomarkers were associated with a variety of immune cells, especially myeloid immune cells. Conclusion We constructed a diagnostic nomogram based on IL10RA, CCR1, and SPI1 and explored the inflammatory-immune mechanisms, which indicated new insights for early diagnosis and treatment of ISM-related AS.
Collapse
Affiliation(s)
- Qichong Yang
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Juncheng Liu
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
- Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tingting Zhang
- Center for Clinical Single-Cell Biomedicine, Henan Province People’s Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tingting Zhu
- Department of Cardiopulmonary Functions Test, Henan Province People’s Hospital, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Siyu Yao
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Rongzi Wang
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Wenjuan Wang
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Haliminai Dilimulati
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Songtao An
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| |
Collapse
|
27
|
Yu Y, Cai Y, Yang F, Yang Y, Cui Z, Shi D, Bai R. Vascular smooth muscle cell phenotypic switching in atherosclerosis. Heliyon 2024; 10:e37727. [PMID: 39309965 PMCID: PMC11416558 DOI: 10.1016/j.heliyon.2024.e37727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/25/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Atherosclerosis (AS) is a complex pathology process involving intricate interactions among various cells and biological processes. Vascular smooth muscle cells (VSMCs) are the predominant cell type in normal arteries, and under atherosclerotic stimuli, VSMCs respond to altered blood flow and microenvironment changes by downregulating contractile markers and switching their phenotype. This review overviews the diverse phenotypes of VSMCs, including the canonical contractile VSMCs, synthetic VSMCs, and phenotypes resembling macrophages, foam cells, myofibroblasts, osteoblasts/chondrocytes, and mesenchymal stem cells. We summarize their presumed protective and pro-atherosclerotic roles in AS development. Additionally, we underscore the molecular mechanisms and regulatory pathways governing VSMC phenotypic switching, encompassing transcriptional regulation, biochemical factors, plaque microenvironment, epigenetics, miRNAs, and the cytoskeleton, emphasizing their significance in AS development. Finally, we outline probable future research directions targeting VSMCs, offering insights into potential therapeutic strategies for AS management.
Collapse
Affiliation(s)
- Yanqiao Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yajie Cai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Furong Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Yankai Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhuorui Cui
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dazhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Ruina Bai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| |
Collapse
|
28
|
Guan X, Hu Y, Hao J, Lu M, Zhang Z, Hu W, Li D, Li C. Stress, Vascular Smooth Muscle Cell Phenotype and Atherosclerosis: Novel Insight into Smooth Muscle Cell Phenotypic Transition in Atherosclerosis. Curr Atheroscler Rep 2024; 26:411-425. [PMID: 38814419 DOI: 10.1007/s11883-024-01220-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE OF REVIEW Our work is to establish more distinct association between specific stress and vascular smooth muscle cells (VSMCs) phenotypes to alleviate atherosclerotic plaque burden and delay atherosclerosis (AS) progression. RECENT FINDING In recent years, VSMCs phenotypic transition has received significant interests. Different stresses were found to be associated with VSMCs phenotypic transition. However, the explicit correlation between VSMCs phenotype and specific stress has not been elucidated clearly yet. We discover that VSMCs phenotypic transition, which is widely involved in the progression of AS, is associated with specific stress. We discuss approaches targeting stresses to intervene VSMCs phenotypic transition, which may contribute to develop innovative therapies for AS.
Collapse
Affiliation(s)
- Xiuya Guan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jiaqi Hao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenxian Hu
- Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, China.
| | - Dongxiao Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, China.
| |
Collapse
|
29
|
Martinez AN, Tortelote GG, Pascale CL, Ekanem UOI, Leite APDO, McCormack IG, Dumont AS. Dimethyl Fumarate Mediates Sustained Vascular Smooth Muscle Cell Remodeling in a Mouse Model of Cerebral Aneurysm. Antioxidants (Basel) 2024; 13:773. [PMID: 39061841 PMCID: PMC11274241 DOI: 10.3390/antiox13070773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/20/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Cerebral aneurysms (CA) are a type of vascular disease that causes significant morbidity and mortality with rupture. Dysfunction of the vascular smooth muscle cells (VSMCs) from circle of Willis (CoW) vessels mediates CA formation, as they are the major cell type of the arterial wall and play a role in maintaining vessel integrity. Dimethyl fumarate (DMF), a first-line oral treatment for relapsing-remitting multiple sclerosis, has been shown to inhibit VSMC proliferation and reduce CA formation in a mouse model. Potential unwanted side effects of DMF on VSMC function have not been investigated yet. The present study characterizes the impact of DMF on VSMC using single-cell RNA-sequencing (scRNA-seq) in CoW vessels following CA induction and further explores its role in mitochondrial function using in vitro VSMC cultures. Two weeks of DMF treatment following CA induction impaired the transcription of the glutathione redox system and downregulated mitochondrial respiration genes in VSMCs. In vitro, DMF treatment increased lactate formation and enhanced the mitochondrial production of reactive oxygen species (ROS). These effects rendered VSMCs vulnerable to oxidative stress and led to mitochondrial dysfunction and enhancement of apoptosis. Taken together, our data support the concept that the DMF-mediated antiproliferative effect on VSMCs is linked to disturbed antioxidative functions resulting in altered mitochondrial metabolism. This negative impact of DMF treatment on VSMCs may be linked to preexisting alterations of cerebrovascular function due to renal hypertension. Therefore, before severe adverse effects emerge, it would be clinically relevant to develop indices or biomarkers linked to this disturbed antioxidative function to monitor patients undergoing DMF treatment.
Collapse
Affiliation(s)
- Alejandra N. Martinez
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Giovane G. Tortelote
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | - Crissey L. Pascale
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Uduak-Obong I. Ekanem
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Ana Paula de O. Leite
- Department of Pharmacology, The Tulane Center for Sex-Based Biology and Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Isabella G. McCormack
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Aaron S. Dumont
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| |
Collapse
|