1
|
Zhang WZ, Wu CY, Lai H. A Review on the Role of DNA Methylation in Aortic Disease Associated With Marfan Syndrome. Cardiol Res 2025; 16:169-177. [PMID: 40370619 PMCID: PMC12074684 DOI: 10.14740/cr2033] [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: 12/23/2024] [Accepted: 03/08/2025] [Indexed: 05/16/2025] Open
Abstract
Marfan syndrome (MFS) is a genetic disorder primarily affecting the connective tissue, with cardiovascular complications as the leading cause of mortality. While mutations in the FBN1 gene are the primary cause, the severity and progression of the disease can vary significantly among individuals. DNA methylation, a key epigenetic regulatory mechanism, has garnered attention in MFS research, particularly regarding methylation changes in the FBN1 locus and their effects on fibrillin-1 expression. Differential methylation and expression of genes related to inflammation (e.g., interleukin (IL)-10, IL-17) and oxidative stress (e.g., PON2, TP53INP1) have been linked to MFS aortic pathology. These alterations likely contribute to disease progression by influencing inflammatory responses, smooth muscle cell apoptosis, and biomechanical properties of the aorta. The transforming growth factor-beta (TGF-β) signaling pathway plays a central role in MFS pathology, with aberrant methylation of related genes potentially elevating active TGF-β levels and exacerbating aortic lesions. Notably, tissue-specific methylation patterns, especially in smooth muscle cells of the aorta, remain poorly understood. A deeper understanding of DNA methylation's role in MFS could pave the way for early interventions and epigenetic-targeted therapies.
Collapse
Affiliation(s)
- Wei Ze Zhang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Ye Wu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Berger T, Dumfarth J, Kreibich M, Minatoya K, Ziganshin BA, Czerny M. Thoracic aortic aneurysm. Nat Rev Dis Primers 2025; 11:34. [PMID: 40341396 DOI: 10.1038/s41572-025-00617-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 05/10/2025]
Abstract
Aortic medicine has undergone remarkable progress in recent decades with regard to our understanding and treatment of aortic disease. In the past decade, the scientific community has called for the aorta to be viewed as an independent organ, advocating for a holistic approach to understanding thoracic aortic disease, integrating its embryological development, wall composition, pathophysiological mechanisms, surveillance and treatment. Thoracic aortic aneurysm (TAA) is a potentially fatal disease characterized by abnormal dilation of the thoracic aorta, whereby the structural integrity of the vessel wall is compromised. Although epidemiological studies of TAA are confounded by its asymptomatic nature and diagnostic challenges, available evidence suggests that TAA prevalence and treatment outcomes vary according to race, sex and socioeconomic factors. Pathophysiological mechanisms involve interactions between vascular smooth muscle cells and the extracellular matrix, influenced by genetic predisposition and embryological factors as well as arterial hypertension. Diagnosis relies on advanced imaging techniques, with CT angiography considered to be the gold standard diagnostic tool and with genetic screening recommended for heritable conditions. Preventive measures focus on managing cardiovascular risk factors, whereas treatment includes medical management, as well as endovascular and open surgical repair. TAA has a major effect on quality of life, particularly in younger, female and genetically predisposed patients, necessitating further research and tailored interventions.
Collapse
Affiliation(s)
- Tim Berger
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, University Medical Center Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Dumfarth
- University Clinic for Cardiac Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Maximilian Kreibich
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, University Medical Center Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Bulat A Ziganshin
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA
| | - Martin Czerny
- Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, University Medical Center Freiburg, Freiburg, Germany.
- University Clinic for Cardiac Surgery, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
3
|
Yu G, Li J, Zhang H, Zi H, Liu M, An Q, Qiu T, Li P, Song J, Liu P, Quan K, Li S, Liu Y, Zhu W, Du J. Single-cell analysis reveals the implication of vascular endothelial cell-intrinsic ANGPT2 in human intracranial aneurysm. Cardiovasc Res 2025; 121:658-673. [PMID: 39187926 DOI: 10.1093/cvr/cvae186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/04/2024] [Accepted: 06/13/2024] [Indexed: 08/28/2024] Open
Abstract
AIMS While previous single-cell RNA sequencing (scRNA-seq) studies have attempted to dissect intracranial aneurysm (IA), the primary molecular mechanism for IA pathogenesis remains unknown. Here, we uncovered the alterations of cellular compositions, especially the transcriptome changes of vascular endothelial cells (ECs), in human IA. METHODS AND RESULTS We performed scRNA-seq to compare the cell atlas of sporadic IA and the control artery. The transcriptomes of 43 462 cells were profiled for further analysis. In general, IA had increased immune cells (T/NK cells, B cells, myeloid cells, mast cells, neutrophils) and fewer vascular cells (ECs, vascular smooth muscle cells, and fibroblasts). Based on the obtained high-quantity and high-quality EC data, we found genes associated with angiogenesis in ECs from IA patients. By EC-specific expression of candidate genes in vivo, we observed the involvement of angpt2a in causing cerebral vascular abnormality. Furthermore, an IA zebrafish model mimicking the main features of human IA was generated through targeting pdgfrb gene, and knockdown of angpt2a alleviated the vascular dilation in the IA zebrafish model. CONCLUSION By performing a landscape view of the single-cell transcriptomes of IA and the control artery, we contribute to a deeper understanding of the cellular composition and the molecular changes of ECs in IA. The implication of angiogenic regulator ANGPT2 in IA formation and progression, provides a novel potential therapeutical target for IA interventions.
Collapse
Affiliation(s)
- Guo Yu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Jia Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Hongfei Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Huaxing Zi
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, Beijing 100049, China
| | - Mingjian Liu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Qingzhu An
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Tianming Qiu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Peiliang Li
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Jianping Song
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Peixi Liu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Kai Quan
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Sichen Li
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Yingjun Liu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- National Center for Neurological Disorders, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Neurosurgical Institute of Fudan University, 12 Middle Wulumuqi Road,Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery,12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, Beijing 100049, China
- School of Life Science and Technology, ShanghaiTech University, 319 Yue-Yang Road, Shanghai 200031, China
| |
Collapse
|
4
|
Das AA, Waldeck-Weiermair M, Yadav S, Spyropoulos F, Pandey A, Dutta T, Covington TA, Michel T. Differential aortic aneurysm formation provoked by chemogenetic oxidative stress. J Clin Invest 2025; 135:e188743. [PMID: 40100302 PMCID: PMC12043099 DOI: 10.1172/jci188743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
Aortic aneurysms are potentially fatal focal enlargements of the aortic lumen; the disease burden is increasing as the human population ages. Pathological oxidative stress is implicated in the development of aortic aneurysms. We pursued a chemogenetic approach to create an animal model of aortic aneurysm formation using a transgenic mouse line, DAAO-TGTie2, that expresses yeast d-amino acid oxidase (DAAO) under control of the endothelial Tie2 promoter. In DAAO-TGTie2 mice, DAAO generated the ROS hydrogen peroxide (H2O2) in endothelial cells only when provided with d-amino acids. When DAAO-TGTie2 mice were chronically fed d-alanine, the animals became hypertensive and developed abdominal, but not thoracic, aortic aneurysms. Generation of H2O2 in the endothelium led to oxidative stress throughout the vascular wall. Proteomics analyses indicated that the oxidant-modulated protein kinase JNK1 was dephosphorylated by the phosphoprotein phosphatase DUSP3 (dual specificity phosphatase 3) in abdominal, but not thoracic, aorta, causing activation of Kruppel-like Factor 4 (KLF4)-dependent transcriptional pathways that triggered phenotypic switching and aneurysm formation. Pharmacological DUSP3 inhibition completely blocked the aneurysm formation caused by chemogenetic oxidative stress. These studies establish that regional differences in oxidant-modulated signaling pathways lead to differential disease progression in discrete vascular beds and identify DUSP3 as a potential pharmacological target for the treatment of aortic aneurysms.
Collapse
Affiliation(s)
- Apabrita Ayan Das
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Markus Waldeck-Weiermair
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Harvard University, Boston, Massachusetts, USA
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Shambhu Yadav
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Fotios Spyropoulos
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Harvard University, Boston, Massachusetts, USA
- Department of Pediatrics, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Arvind Pandey
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Tanoy Dutta
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Taylor A. Covington
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Thomas Michel
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
5
|
He J, Huang K, Fan X, Chang G. Sulfur Dioxide Alleviates Aortic Dissection Through Inhibiting Vascular Smooth Muscle Cell Phenotype Switch, Migration, and Proliferation via miR-184-3p/Cyp26b1 Axis. Antioxid Redox Signal 2025; 42:672-686. [PMID: 39834177 DOI: 10.1089/ars.2023.0471] [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] [Indexed: 01/22/2025]
Abstract
Aims: Abnormal migration and proliferation of vascular smooth muscle cells (VSMCs) are considered early events in the onset of thoracic aortic dissection (TAD). Endogenous sulfur dioxide (SO2), primarily produced by aspartate aminotransferase (AAT1) in mammals, has been reported to inhibit the migration and proliferation of VSMCs. However, the role of SO2 in the development of TAD remains unclear. Results: Endogenous SO2 production was decreased in aortic samples from patients with TAD. Supplementation with SO2 ameliorated β-aminopropionitrile-induced vascular injury in mice. Increasing the expression of SO2 pathway might reverse the abnormal migration, proliferation, and phenotypic switching in VSMCs. MicroRNA sequencing revealed miR-184-3p as the miRNA with the most significant increased expression level after AAT1 knockdown, and Cyp26b1 was predicted to be its potential target. A decrease in the SO2 pathway resulted in reduced Cyp26b1 expression, impairing VSMCs function, while restoring Cyp26b1 expression with miR-184-3p inhibitors could improve the VSMCs function. Innovation: This research extends the application of endogenous SO2 to the aortic diseases and elucidates the role of miRNA in endogenous SO2 regulatory network, highlighting its potential as a target for clinical practice. Conclusion: Endogenous SO2 inhibits the migration and proliferation of VSMCs in TAD progression via the miR-184-3p/Cyp26b1 axis. Antioxid. Redox Signal. 42, 672-686.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Animals
- Sulfur Dioxide/pharmacology
- Sulfur Dioxide/metabolism
- Mice
- Humans
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/drug therapy
- Aortic Dissection/genetics
- Phenotype
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Male
- Cytochrome P-450 Enzyme System/metabolism
- Cytochrome P-450 Enzyme System/genetics
- Signal Transduction
- Oxygenases
Collapse
Affiliation(s)
- Jie He
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation (Sun Yat-Sen University), The First Affiliated Hospital, Sun Yat Sen University, Guangzhou, China
- Department of Cardiovascular Surgery, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kan Huang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation (Sun Yat-Sen University), The First Affiliated Hospital, Sun Yat Sen University, Guangzhou, China
| | - Xiaoping Fan
- Department of Cardiovascular Surgery, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangqi Chang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation (Sun Yat-Sen University), The First Affiliated Hospital, Sun Yat Sen University, Guangzhou, China
| |
Collapse
|
6
|
Zhang Q, Cai Z, Yu Z, Di C, Qiu Y, Qi R. Agathis dammara Extract and its Monomer Araucarone Attenuate Abdominal Aortic Aneurysm in Mice. Cardiovasc Drugs Ther 2025; 39:239-257. [PMID: 37979015 DOI: 10.1007/s10557-023-07518-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/22/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a chronic vascular disease wherein the inflammation of vascular smooth muscle cells (VSMCs) plays a pivotal role in its development. Effectively mitigating AAA involves inhibiting VSMC inflammation. Agathis dammara (Lamb.) Rich, recognized for its robust anti-inflammatory and antioxidant attributes, has been employed as a traditional medicinal resource. Nonetheless, there is a dearth of information regarding the potential of Agathis dammara extract (AD) in attenuating AAA, specifically by diminishing vascular inflammation, notably VSMC inflammation. Furthermore, the active constituents of AD necessitate identification. AIM OF THE STUDY This study sought to ascertain the efficacy of AD in reducing AAA, evaluate its impact on VSMC inflammation, and elucidate whether the monomer araucarone (AO) in AD acts as an active component against AAA. MATERIALS AND METHODS The extraction of AD was conducted and subjected to analysis through High-Performance Liquid Chromatography (HPLC) and mass spectrometry. The isolation of the AO monomer followed, involving the determination of its content and purity. Subsequently, the effects of AD and AO on VSMC inflammation were assessed in vitro, encompassing an examination of inflammatory factors such as IL-6 and IL-18, as well as the activation of matrix metalloproteinase 9 (MMP9) in tumor necrosis factor-alpha (TNF-α)-stimulated VSMCs. To explore the inhibitory effects of AD/AO on AAA, C57BL/6J male mice were subjected to oral gavage (100 mg/kg) or intraperitoneal injection (50 mg/kg) of AD and AO in a porcine pancreatic elastase (PPE)-induced AAA model (14 days). This facilitated the observation of abdominal aorta dilatation, remodeling, elastic fiber disruption, and macrophage infiltration. Additionally, a three-day PPE mouse model was utilized to assess the effects of AD and AO (administered at 100 mg/kg via gavage) on acute inflammation and MMP9 expression in blood vessels. The mechanism by which AD/AO suppresses the inflammatory response was probed through the examination of NF-κB/NLRP3 pathway activation in VSMCs and aortas. RESULTS Liquid Chromatography-Mass Spectrometry (LC-MS) revealed that AO constituted 15.36% of AD's content, with a purity of 96%. Subsequent pharmacological investigations of AO were conducted in parallel with AD. Both AD and AO exhibited the ability to inhibit TNF-α-induced VSMC inflammation and MMP production in vitro. Furthermore, both substances effectively prevented PPE-induced AAA in mice, whether administered through gavage or intraperitoneal injection, evidenced by decreased vascular diameter dilation, disruption of elastin fiber layers, and infiltration of inflammatory cells. In the three-day PPE mouse model, AD and AO mitigated the heightened expression of inflammatory factors and the elevated expression of MMP9 induced by PPE. The activation of the NF-κB/NLRP3 pathway in both VSMCs and aortas was significantly suppressed by treatment with AD or AO. CONCLUSIONS Through suppressing NF-κB/NLRP3 pathway activation, AD effectively mitigates the inflammatory response in VSMCs, mitigates inflammation in aortas, prevents extracellular matrix degradation, and consequently impedes the progression of AAA. AO emerges as one of the active compounds in AD responsible for inhibiting VSMC inflammation and inhibiting AAA development.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/prevention & control
- Plant Extracts/pharmacology
- Plant Extracts/isolation & purification
- Male
- Disease Models, Animal
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/isolation & purification
- Mice, Inbred C57BL
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/pathology
- Aorta, Abdominal/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mice
- Inflammation Mediators/metabolism
- Diterpenes/pharmacology
- Diterpenes/isolation & purification
- Cells, Cultured
- Signal Transduction
- NF-kappa B/metabolism
Collapse
Affiliation(s)
- Qingyi Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhewei Yu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Chang Di
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Yingkun Qiu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.
| | - Rong Qi
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China.
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing, China.
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China.
| |
Collapse
|
7
|
Zheng Z, Wang W, Chen B, Huang M, Wang T, Xu Z, Dai X. LncRNA BANCR/miR-15a/MAPK1 Induces Apoptosis and Increases Proliferation of Vascular Smooth Muscle Cells in Aortic Dissection by Enhancing MMP2 Expression. Cell Biochem Biophys 2025:10.1007/s12013-025-01738-x. [PMID: 40156764 DOI: 10.1007/s12013-025-01738-x] [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] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
Aortic dissection is associated with a high mortality rate, contributing to an unfavorable prognosis. Preventive measures are more effective than therapeutic interventions for aortic dissection. While LncRNA BANCR is recognized as a functional translational regulator in various diseases, its role in aortic dissection remains unexplored. This study aims to elucidate the functions and molecular mechanisms of BANCR in aortic dissection. Vascular smooth muscle cells were isolated from dissected aortic tunica media samples and their phenotypes were compared with those of commercial vascular smooth muscle cells. BANCR expression was modulated via transient transfection (overexpression) and small interfering RNA (knockdown). The involvement of the p38 MAPK pathway was examined using the inhibitor SB202190. The competing endogenous RNA network was validated through a dual luciferase assay. Cellular phenotypes were assessed using the CCK-8 assay, scratch assay, and flow cytometry. BANCR was overexpressed in dissected aortic tissues and isolated vascular smooth muscle cells. MiR-15a-5p exhibited binding affinity to both BANCR and MAPK1. Overexpression of BANCR activated p38 phosphorylation, enhanced cell proliferation and migration, and increased apoptosis. SB202190 mitigated these BANCR-induced phenotypes by inhibiting p38 phosphorylation. Additionally, MMP2 upregulation was linked to BANCR overexpression via the p38 MAPK pathway. Suppression of BANCR expression or inhibition of p38 phosphorylation reduced MMP2 levels, thereby reversing BANCR-induced phenotypes. The LncRNA BANCR/miR-15a-5p/MAPK1 axis forms a ceRNA network that modulates MMP2 expression through the p38 MAPK signaling pathway in vascular smooth muscle cells. BANCR overexpression activates p38 MAPK phosphorylation, leading to enhanced MMP2 expression and subsequent increases in cell proliferation, migration, and apoptosis.
Collapse
Affiliation(s)
- Zihe Zheng
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Wei Wang
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Bo Chen
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
- Department of Cardiovascular Surgery, Gaozhou People's Hospital, Gaozhou, China
| | - Ming Huang
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Tao Wang
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zheng Xu
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaofu Dai
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China.
| |
Collapse
|
8
|
Bhatt N, Seo H, Hanneman K, Burris N, Simmons CA, Chung JCY. Imaging-based biomechanical parameters for assessing risk of aortic dissection and rupture in thoracic aortic aneurysms. Eur J Cardiothorac Surg 2025; 67:ezaf128. [PMID: 40234250 DOI: 10.1093/ejcts/ezaf128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/23/2025] [Accepted: 04/13/2025] [Indexed: 04/17/2025] Open
Abstract
OBJECTIVES Imaging-based methods of measuring aortic biomechanics may provide superior and a more personalized in vivo risk assessment of patients with thoracic aortic aneurysms compared to traditional aortic size criteria such as maximal aortic diameter. We aim to summarize the data on in vivo imaging techniques for evaluation of aortic biomechanics. METHODS A thorough search of literature was conducted in MEDLINE, EMBASE and Google Scholar for evidence of various imaging-based biomechanics techniques. All imaging modalities were included. Data involving preclinical/animal models or exclusively focussed on abdominal aortic aneurysms were excluded. RESULTS The various imaging-based biomechanical parameters can be divided into categories of increasing complexity: strain-based, stiffness-based and computational modelling-derived. Strain-based and stiffness-based parameters are more simply calculated and can be derived using multiple imaging modalities. Initial studies are promising towards linking these parameters with clinically relevant end-points, including aortic dissection, though work is required for standardization. Computationally derived parameters provide detail of stress exerted on the aortic wall with great spatial resolution. However, they are highly dependent on the assumptions applied to the models, such as material properties of the aortic wall. CONCLUSIONS Imaging-based aortic biomechanics represent a major technical advancement for personalized in vivo risk stratification of patients with ascending thoracic aortic aneurysm. The next steps in clinical translation require large-scale validation of these markers towards predicting aortic dissections and comparison against the gold standard ex vivo aortic biomechanics as well as development of a user-friendly, low-cost algorithm that can be widely adopted.
Collapse
Affiliation(s)
- Nitish Bhatt
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Hijun Seo
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Kate Hanneman
- Department of Medical Imaging, University Medical Imaging Toronto, University of Toronto, Toronto, ON, Canada
| | - Nicholas Burris
- Department of Radiology, University of Michigan, Ann Arbor, MI, United States
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Jennifer C-Y Chung
- Division of Cardiovascular Surgery, University Health Network, Toronto, ON, Canada
| |
Collapse
|
9
|
Wang H, Kazaleh M, Gioscia-Ryan R, Millar J, Temprano-Sagrera G, Wood S, Van Den Bergh F, Blin MG, Wragg KM, Luna A, Hawkins RB, Soleimanpour SA, Sabater-Lleal M, Shu C, Beard DA, Ailawadi G, Deng JC, Goldstein DR, Salmon M. Deficiency of mitophagy mediator Parkin in aortic smooth muscle cells exacerbates abdominal aortic aneurysm. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.30.621201. [PMID: 39554010 PMCID: PMC11565987 DOI: 10.1101/2024.10.30.621201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Abdominal aortic aneurysms (AAAs) are a degenerative aortic disease and associated with hallmarks of aging, such as mitophagy. Despite this, the exact associations among mitophagy, aging, and AAA progression remain unknown. In our study, gene expression analysis of human AAA tissue revealed downregulation of mitophagy pathways, mitochondrial structure, and function-related proteins. Human proteomic analyses identified decreased levels of mitophagy mediators PINK1 and Parkin. Aged mice and, separately, a murine AAA model showed reduced mitophagy in aortic vascular smooth muscle cells (VSMCs) and PINK1 and Parkin expression. Parkin knockdown in VSMCs aggravated AAA dilation in murine models, with elevated mitochondrial ROS and impaired mitochondrial function. Importantly, inhibiting USP30, an antagonist of the PINK1/Parkin pathway, increased mitophagy in VSMCs, improved mitochondrial function, and reduced AAA incidence and growth. Our study elucidates a critical mechanism that proposes AAAs as an age-associated disease with altered mitophagy, introducing new potential therapeutic approaches.
Collapse
|
10
|
He W, Yu S, Li J, Li S, Chen Z, Zhang J, Liu Y, Zhou M, Yang T, Cheng W, Dai SS. From inflammation to remodelling: A novel BASP1 + monocyte subset as a catalyst for acute aortic dissection. J Adv Res 2025:S2090-1232(25)00144-4. [PMID: 40057028 DOI: 10.1016/j.jare.2025.03.003] [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: 11/12/2024] [Revised: 02/10/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025] Open
Abstract
INTRODUCTION Monocytes comprise heterogeneous cell populations. However, beyond traditionally considered as precursors of tissue macrophages, heterogeneity and detailed effects of monocytes in acute aortic dissection (AAD) are largely unknown. OBJECTIVES To investigate the role of brain soluble acid protein 1 positive (BASP1+) monocyte subset in promoting AAD development as well as the underlying mechanism. METHODS Monocyte/macrophage heterogeneity in both human peripheral blood and aortic tissues were assayed by scRNA-seq. Monocyte trafficking and lineage tracing were detected by immunofluorescence and using BASP1-CreER/Lyz2-DreER-tdT reporter mice with AAD. The effects and underlying mechanism were investigated by laser speckle image, ultrasound imaging, Co-IP, ChIP-sequencing. Conditional knockout of BASP1 on monocyte and BASP1 siRNA were used to observe BASP1+ monocyte subset-targeted AAD intervention. RESULTS "PIP2-SP1-ACTN1/VAV3" and "ITGB1-Rac1-GSN" signalling mediated BASP1+ monocyte subset as the first line immune cells infiltrating aortic tissues in AAD induction and partial of them transformed to BASP1+ macrophages to amplify the inflammation. Meanwhile, BASP1+ monocyte subset induced an inflammatory phenotype vascular smooth muscle cell (VSMC) subset and a ROS-enriched endothelial cell (EC) subset accompanied with promoting the apoptosis of normal VSMC and EC, contributing to vascular remodelling and dampening the myo-endothelial gap junction. Selective deletion of BASP1+ monocyte subset and interference with BASP1 expression in monocytes both inhibited the development of AAD in mice. CONCLUSION Interpretation BASP1+ monocyte subset and its regulatory network provides deep insight into AAD pathogenesis and a novel potential target for early intervention in AAD formation.
Collapse
Affiliation(s)
- Wenhui He
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China
| | - Sanjiu Yu
- Department of Cardiac Surgery, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Jun Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Siyu Li
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China
| | - Zongtao Chen
- Health Management Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Jingyu Zhang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yangwuyue Liu
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China
| | - Mi Zhou
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China
| | - Teng Yang
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China
| | - Wei Cheng
- Department of Cardiac Surgery, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China.
| | - Shuang-Shuang Dai
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
11
|
Ma L, Xie L, Wu Q, Jin L, Li J, Tang L, Zhang L, Chen L, Qiu Z. Targeting the S100A9/P38 MAPK/HSPB1 axis as a novel approach for aortic dissection therapy. Int Immunopharmacol 2025; 149:114225. [PMID: 39904041 DOI: 10.1016/j.intimp.2025.114225] [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: 11/28/2024] [Revised: 01/02/2025] [Accepted: 02/01/2025] [Indexed: 02/06/2025]
Abstract
INTRODUCTION Aortic dissection (AD) is caused by inflammatory responses and extracellular matrix (ECM) degradation processes, in which S100A9, a proinflammatory protein, may play a role. This study explored the role S100A9/P38 MAPK/HSPB1 signaling axis in AD pathogenesis and the therapeutic potential of targeting this pathway. METHODS S100A9 expression in the aortic tissues of patients with AD/healthy controls were analyzed using bioinformatics, ELISA, qPCR, western blotting, and immunohistochemistry. In an AD mouse model induced by β-aminopropionitrile and angiotensin II (Ang-II), S100A9 expression was inhibited using specific inhibitors to assess its relationship with AD, and proteomics were performed to explore the pathways related to S100A9 expression. Human aortic vascular smooth muscle cells (HVSMC) were treated with Ang-II, S100A9 knockdown, P38 MAPK inhibitors, and HSPB1 knockdown, and experimental methods were used to assess changes in inflammatory cytokines, ECM remodeling, cell proliferation, and apoptosis. Rescue experiments validated the role of the S100A9/P38 MAPK/HSPB1 axis. RESULTS S100A9 was significantly upregulated in patients with AD, while levels of inflammatory cytokines and matrix metalloproteinases (MMPs) were elevated. S100a9 inhibition reduced the incidence of AD, improved survival, and stabilized the aortic structure in mice, with reduced collagen deposition and SMC apoptosis in vitro. S100A9 knockdown reduces Ang-II-induced HVSMC proliferation, apoptosis resistance, and ECM degradation. Mechanistic studies revealed that the S100A9/P38 MAPK/HSPB1 axis regulates inflammatory cytokine and MMPs release. CONCLUSION S100A9 regulates inflammation and ECM degradation through the P38 MAPK/HSPB1 axis, influencing HVSMC proliferation and apoptosis and promoting AD development. This pathway may be a promising therapeutic target for AD treatment.
Collapse
Affiliation(s)
- Likang Ma
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Linfeng Xie
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Qingsong Wu
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Lei Jin
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Jiakang Li
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences Fujian Medical University Fuzhou China
| | - Lele Tang
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Li Zhang
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences Fujian Medical University Fuzhou China.
| | - Liangwan Chen
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China.
| | - Zhihuang Qiu
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China.
| |
Collapse
|
12
|
Kucher AN, Koroleva IA, Nazarenko MS. Exploring Disparities in Atherosclerosis Comorbidity with Aortic Aneurysm. Biomedicines 2025; 13:593. [PMID: 40149570 PMCID: PMC11940622 DOI: 10.3390/biomedicines13030593] [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: 11/25/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 03/29/2025] Open
Abstract
Aortic aneurysm (AA) and atherosclerosis (AS) of various vascular beds are asymptomatic for a long time and are relatively common pathological conditions that lead to life-threatening and disabling complications. In this review, we discuss the current understanding of the high variation in direct and inverse comorbidity of AA and AS as presented in scientific publications. Estimates of AA and AS comorbidity depend on several factors, such as the location of AA (ascending or descending thoracic aorta or abdominal aorta), familial or sporadic cases of AA, syndromic forms of AA, and/or aortic valve pathology (bicuspid aortic valve [BAV]). To identify the causes of the comorbidity of AA and AS, it is important to consider and characterise many factors in detail. These factors include clinical characteristics of the patients included in a study (age, sex) and risk factors (mainly the presence of monogenic forms and BAV, hypertension, hypercholesterolaemia, diabetes mellitus, and cigarette smoking). Additionally, it is essential to consider characteristics of the disease course and the nature of multimorbidity and to take into account pathologies not only of the cardiovascular system but also of other organ systems, with special attention to metabolic and endocrine disorders.
Collapse
Affiliation(s)
| | | | - Maria S. Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| |
Collapse
|
13
|
Schröder LC, Hüttermann L, Kliesow Remes A, Voran JC, Hille S, Sommer W, Lutter G, Warnecke G, Frank D, Schade D, Müller OJ. AAV library screening identifies novel vector for efficient transduction of human aorta. Gene Ther 2025; 32:154-162. [PMID: 39695327 PMCID: PMC11946879 DOI: 10.1038/s41434-024-00511-8] [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: 08/05/2024] [Revised: 11/16/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024]
Abstract
Targeted gene delivery to vascular smooth muscle cells (VSMCs) could prevent or improve a variety of diseases affecting the vasculature and particularly the aorta. Thus, we aimed to develop a delivery vector that efficiently targets VSMCs. We selected engineered adeno-associated virus (AAV) capsids from a random AAV capsid library and tested the top enriched motifs in parallel screening through individual barcoding. This approach allowed us to distinguish capsids that only transduce cells based on genomic DNA (gDNA) from those also mediating transgene expression based on transcribed cDNA reads. After three rounds of selection on primary murine VSMCs (mVSMCs), we identified a novel targeting motif (RFTEKPA) that significantly improved transduction and gene expression efficiency over AAV9-wild type (WT) and increased expression in mVSMCs by 70% compared to the previously identified SLRSPPS peptide. Further analysis showed that the novel motif also improved expression in human aortic smooth muscle cells (HAoSMCs) and human aortic tissue ex vivo up to threefold compared to SLRSPPS and approximately 70-fold to AAV9-WT. This high cross-species transduction efficiency makes the novel capsid motif a potential candidate for future clinical application in vascular diseases.
Collapse
Affiliation(s)
- Lena C Schröder
- Department of Internal Medicine V, University of Kiel, Kiel, Germany
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Leonard Hüttermann
- Department of Internal Medicine V, University of Kiel, Kiel, Germany
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Anca Kliesow Remes
- Department of Internal Medicine V, University of Kiel, Kiel, Germany
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Jakob C Voran
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
- Department of Internal Medicine III, University of Kiel, Kiel, Germany
| | - Susanne Hille
- Department of Internal Medicine V, University of Kiel, Kiel, Germany
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Wiebke Sommer
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
- Department of Cardiac and Vascular Surgery, University of Kiel, Kiel, Germany
| | - Georg Lutter
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
- Department of Cardiac and Vascular Surgery, University of Kiel, Kiel, Germany
| | - Gregor Warnecke
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
- Department of Cardiac and Vascular Surgery, University of Kiel, Kiel, Germany
| | - Derk Frank
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
- Department of Internal Medicine III, University of Kiel, Kiel, Germany
| | - Dennis Schade
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
- Department of Pharmaceutical & Medicinal Chemistry, Institute of Pharmacy, University of Kiel, Kiel, Germany
| | - Oliver J Müller
- Department of Internal Medicine V, University of Kiel, Kiel, Germany.
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany.
| |
Collapse
|
14
|
Liu Y, Tan X, Wang L, Jin W, He Y, Yan Y, Hu K, Wang H, Xiang C, Hou M, Lai Y. Identification of the CeRNA axis of circ_0000006/miR-483-5p/KDM2B in the progression of aortic aneurysm to aorta dissection. BMC Cardiovasc Disord 2025; 25:141. [PMID: 40021969 PMCID: PMC11869725 DOI: 10.1186/s12872-025-04598-8] [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: 06/07/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Aortic aneurysm (AA) and aortic dissection (AD) are serious cardiovascular disorders with a high risk of mortality. The molecular mechanisms underlying the progression from AA to AD are not well understood. This study aimed to identify the key circular RNA (circRNA)-microRNA (miRNA)-messenger RNA (mRNA) regulatory axis involved in this disease progression. METHODS CircRNA microarray, miRNA microarray, and mRNA sequencing were performed on plasma samples from healthy controls, AA patients, and AD patients. Bioinformatics analysis integrated the expression profiles to identify dysregulated circRNA-miRNA-mRNA networks. Key molecules were validated in vascular smooth muscle cells (VSMCs) and an AD mouse model. Cell proliferation, migration, and phenotypic transition assays were conducted after modulating the identified circRNA. The impact on AD progression was evaluated in mice upon circRNA knockdown. RESULTS A total of 12 circRNAs were found upregulated in AD compared to AA samples. miR-483-5p was downregulated while its targets KDM2B and circ_0000006 were upregulated in AD. Silencing circ_0000006 in VSMCs inhibited PDGF-induced phenotypic switching, proliferation, and migration by increasing miR-483-5p and decreasing KDM2B levels. In the AD mouse model, knockdown of circ_0000006 alleviated disease progression with similar molecular changes. CONCLUSION The study identified a novel circ_0000006/miR-483-5p/KDM2B axis dysregulated during AD progression. Targeting this axis, especially circ_0000006, could be a potential strategy to mitigate the transition from AA to AD by modulating VSMC phenotype and function.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Animals
- Humans
- Aortic Dissection/genetics
- Aortic Dissection/pathology
- Aortic Dissection/enzymology
- Aortic Dissection/metabolism
- Disease Progression
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- Cell Movement
- Disease Models, Animal
- Cell Proliferation
- Male
- Cells, Cultured
- Aortic Aneurysm/genetics
- Aortic Aneurysm/pathology
- Aortic Aneurysm/enzymology
- Aortic Aneurysm/metabolism
- Case-Control Studies
- Female
- Middle Aged
- Mice, Inbred C57BL
- Signal Transduction
- Gene Regulatory Networks
- Gene Expression Regulation
- Mice
- Phenotype
- Aged
- RNA, Competitive Endogenous
Collapse
Affiliation(s)
- Yong Liu
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China.
- , No. 63, Culture Road, Nanchong, Sichuan, 637000, China.
| | - Xiong Tan
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Liang Wang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Weitao Jin
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yangchen He
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yu Yan
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Kai Hu
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Hao Wang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Chaohu Xiang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Ming Hou
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yinglong Lai
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| |
Collapse
|
15
|
Kumari K, Verma K, Sahu M, Dwivedi J, Paliwal S, Sharma S. Emerging role of mesenchymal cells in cardiac and cerebrovascular diseases: Physiology, pathology, and therapeutic implications. Vascul Pharmacol 2025:107473. [PMID: 39993517 DOI: 10.1016/j.vph.2025.107473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/11/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025]
Abstract
In recent years, the therapeutic utility of mesenchymal stem cells (MSCs) has received substantial attention from investigators, owing to their pleiotropic properties. The emerging insights from the developments in tissue engineering provide perspectives for the repair of damaged tissue and the replacement of failing organs. Perivascular cells including MSC-like pericytes, vascular smooth muscles, and other cells located around blood vessels, have been acknowledged to contribute to in situ angiogenesis and repair process. MSCs offer a wide array of therapeutic applications in different pathological states. However, in the current article, we have highlighted the recent updates on MSCs and their key applications in cardiac and cerebrovascular diseases, evident in different preclinical and clinical studies. We believe the present article would assist the investigators in understanding the recent advances of MSCs and exploring their therapeutic potential in varied ailments, especially cardiac and cerebrovascular diseases.
Collapse
Affiliation(s)
- Kajal Kumari
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Kanika Verma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; Department of Internal Medicine, Division of Cardiology, LSU Health Sciences Center - Shreveport, LA, USA
| | - Meenal Sahu
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; Department of Bioscience & Biotechnology, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Sarvesh Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India.
| |
Collapse
|
16
|
Wang W, Chen J, Lai S, Zeng R, Fang M, Wan L, Li Y. METTL14 promotes ferroptosis in smooth muscle cells during thoracic aortic aneurysm by stabilizing the m 6A modification of ACSL4. Am J Physiol Cell Physiol 2025; 328:C387-C399. [PMID: 39672203 DOI: 10.1152/ajpcell.00577.2024] [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: 08/09/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/15/2024]
Abstract
Thoracic aortic aneurysm (TAA) is a vascular disease associated with high mortality rates. Ferroptosis has been shown to mediate the transformation of vascular smooth muscle cells (VSMCs). However, the regulatory mechanisms by which ferroptosis influences TAA remain unclear. In this study, we induced TAA mouse models using angiotensin II (Ang II) and evaluated the impact of ferroptosis on the pathological changes observed in TAA mice, employing liproxstatin-1 as a treatment. In addition, we assessed the regulatory effect of METTL14 on the ferroptosis of VSMCs after treating them with a ferroptosis activator (imidazole ketone erastin, IKE). RNA binding protein immunoprecipitation (RIP) and RNA pull-down assays were conducted to investigate the interaction between acyl-CoA synthase long-chain family member 4 (ACSL4) mRNA and the proteins METTL14 or IGF2BP2. The results indicated that the level of ferroptosis was elevated in the thoracic aorta of TAA mice, and METTL14 was upregulated in TAA models and IKE-induced VSMCs. Knockdown of METTL14 was found to inhibit the progression of TAA by reducing the ferroptosis of VSMCs. Furthermore, IGF2BP2 recognized METTL14-modified ACSL4 mRNA and regulated its stability, thereby mediating the ferroptosis of VSMCs. Collectively, the effects of METTL14 on VSMC ferroptosis present therapeutic potential for the treatment of TAA.NEW & NOTEWORTHY Our study confirmed that METTL14 can induce ferroptosis in vascular smooth muscle cells during the progression of thoracic aortic aneurysm by mediating the m6A modification of ACSL4 mRNA.
Collapse
MESH Headings
- Animals
- Ferroptosis/physiology
- Ferroptosis/drug effects
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Coenzyme A Ligases/metabolism
- Coenzyme A Ligases/genetics
- Mice
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Methyltransferases/metabolism
- Methyltransferases/genetics
- Male
- Mice, Inbred C57BL
- Disease Models, Animal
- Humans
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Angiotensin II
- Aorta, Thoracic/pathology
Collapse
Affiliation(s)
- Wenjun Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Jiayi Chen
- The First Clinical Medical College of Nanchang University, Nanchang, People's Republic of China
| | - Songqing Lai
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Ruiyuan Zeng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Ming Fang
- Department of Emergency, Gaoxin Branch of The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Li Wan
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Yiying Li
- Department of Prenatal Diagnostic Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
17
|
Leng S, Li H, Zhang P, Dang Z, Shao B, Xue S, Ning Y, Teng X, Zhang L, Wang H, Li N, Zhang F, Yu W. SGK1-Mediated Vascular Smooth Muscle Cell Phenotypic Transformation Promotes Thoracic Aortic Dissection Progression. Arterioscler Thromb Vasc Biol 2025; 45:238-259. [PMID: 39633576 PMCID: PMC11748913 DOI: 10.1161/atvbaha.124.321421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND The occurrence of thoracic aortic dissection (TAD) is closely related to the transformation of vascular smooth muscle cells (VSMCs) from a contractile to a synthetic phenotype. The role of SGK1 (serum- and glucocorticoid-regulated kinase 1) in VSMC phenotypic transformation and TAD occurrence is unclear. METHODS Four-week-old male Sgk1F/F (Sgk1 floxed) and Sgk1F/F;TaglnCre (smooth muscle cell-specific Sgk1 knockout) mice were administered β-aminopropionitrile monofumarate for 4 weeks to model TAD. The SGK1 inhibitor GSK650394 was administered daily via intraperitoneal injection to treat the mouse model of TAD. Immunopurification and mass spectrometry were used to identify proteins that interact with SGK1. Immunoprecipitation, immunofluorescence colocalization, and GST (glutathione S-transferase) pull-down were used to detect molecular interactions between SGK1 and SIRT6 (sirtuin 6). RNA-sequencing analysis was performed to evaluate changes in the SIRT6 transcriptome. Quantitative chromatin immunoprecipitation was used to determine the target genes regulated by SIRT6. Functional experiments were also conducted to investigate the role of SGK1-SIRT6-MMP9 (matrix metalloproteinase 9) in VSMC phenotypic transformation. The effect of SGK1 regulation on target genes was evaluated in human and mouse TAD samples. RESULTS Sgk1F/F;TaglnCre or pharmacological blockade of Sgk1 inhibited the formation and rupture of β-aminopropionitrile monofumarate-induced TADs in mice and reduced the degradation of the ECM (extracellular matrix) in vessels. Mechanistically, SGK1 promoted the ubiquitination and degradation of SIRT6 by phosphorylating SIRT6 at Ser338, thereby reducing the expression of the SIRT6 protein. Furthermore, SIRT6 transcriptionally inhibits the expression of MMP9 through epigenetic modification, forming the SGK1-SIRT6-MMP9 regulatory axis, which participates in the ECM signaling pathway. Additionally, our data showed that the lack of SGK1-mediated inhibition of ECM degradation and VSMC phenotypic transformation is partially dependent on the regulatory effect of SIRT6-MMP9. CONCLUSIONS These findings highlight the key role of SGK1 in the pathogenesis of TAD. A lack of SGK1 inhibits VSMC phenotypic transformation by regulating the SIRT6-MMP9 axis, providing insights into potential epigenetic strategies for TAD treatment.
Collapse
MESH Headings
- Animals
- Immediate-Early Proteins/genetics
- Immediate-Early Proteins/metabolism
- Immediate-Early Proteins/antagonists & inhibitors
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/drug effects
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/deficiency
- Aortic Dissection/pathology
- Aortic Dissection/genetics
- Aortic Dissection/enzymology
- Aortic Dissection/chemically induced
- Male
- Phenotype
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/drug effects
- Mice, Knockout
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/enzymology
- Aortic Aneurysm, Thoracic/chemically induced
- Disease Models, Animal
- Humans
- Mice
- Disease Progression
- Aorta, Thoracic/pathology
- Aorta, Thoracic/enzymology
- Cells, Cultured
- Signal Transduction
- Matrix Metalloproteinase 9/metabolism
- Matrix Metalloproteinase 9/genetics
- Vascular Remodeling
- Mice, Inbred C57BL
- Dissection, Thoracic Aorta
Collapse
Affiliation(s)
- Shuai Leng
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Center of Translational Medicine (S.L., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Haijie Li
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Pengfei Zhang
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhiqiao Dang
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Baowei Shao
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shishan Xue
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yansong Ning
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xilong Teng
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Leilei Zhang
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Honglu Wang
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Na Li
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fengquan Zhang
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wenqian Yu
- Department of Cardiac Surgery (S.L., H.L., P.Z., Z.D., B.S., S.X., Y.N., X.T., L.Z., H.W., N.L., F.Z., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Center of Translational Medicine (S.L., W.Y.), Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
18
|
Zong P, Li CX, Feng J, Yue Z, Nethramangalath T, Xie Y, Qin X, Cicchetti M, Cai Y, Jellison E, Matsushita M, Runnels LW, Yue L. TRPM7 channel activity promotes the pathogenesis of abdominal aortic aneurysms. NATURE CARDIOVASCULAR RESEARCH 2025; 4:197-215. [PMID: 39953276 DOI: 10.1038/s44161-024-00596-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 12/04/2024] [Indexed: 02/17/2025]
Abstract
Abdominal aortic aneurysms (AAAs) occur in 1-2% of the elderly. The rupture of an AAA usually causes uncontrollable lethal hemorrhage, and its risk increases with AAA size. However, there is no effective pharmacological therapy for hindering AAA growth. Here we show that global or vascular smooth muscle cell (VSMC)-specific transient receptor potential melastatin 7 (TRPM7) knockout in mice prevented AAA formation, as indicated by inhibited VSMC reprogramming, reduced inflammatory infiltration and suppressed matrix degradation. Mechanistically, we showed that TRPM7-mediated Ca2+ signaling promotes Kruppel-like factor 4 (KLF4) activation, driving VSMC reprogramming and accelerating AAA growth. By generating channel-dead and using kinase-inactive knockin mice, we found that it is the channel function, rather than kinase activity, that is required for TRPM7-mediated AAA pathogenesis. Importantly, TRPM7 inhibitor NS8593 suppressed VSMC reprogramming and protected mice against AAA formation. Our data suggest that TRPM7 is a promising therapeutic target for developing effective prophylactic medications to limit AAA progression. In addition, the channel-dead TRPM7 knockin mice will serve as a valuable tool for elucidating the roles of TRPM7 in other pathophysiological conditions.
Collapse
MESH Headings
- Animals
- TRPM Cation Channels/genetics
- TRPM Cation Channels/metabolism
- TRPM Cation Channels/deficiency
- TRPM Cation Channels/antagonists & inhibitors
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/prevention & control
- Mice, Knockout
- Disease Models, Animal
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Kruppel-Like Factor 4
- Male
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/deficiency
- Mice, Inbred C57BL
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Calcium Signaling/drug effects
- Aorta, Abdominal/pathology
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/drug effects
- Humans
- Cells, Cultured
- Vascular Remodeling/drug effects
- Mice
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, USA
| | - Cindy X Li
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
- Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, USA
| | - Jianlin Feng
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Zhichao Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | | | - Yangzhouyun Xie
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Xin Qin
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Mara Cicchetti
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Yujun Cai
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Evan Jellison
- Department of Immunology, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA
| | - Masayuki Matsushita
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Loren W Runnels
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA.
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT, USA.
| |
Collapse
|
19
|
Guo S, Li J, Pang S, Li J, Tian X. Exosome miR-199a-5p modulated vascular remodeling and inflammatory infiltration of Takayasu's arteritis. Arthritis Res Ther 2025; 27:11. [PMID: 39833857 PMCID: PMC11744942 DOI: 10.1186/s13075-025-03475-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Advances in treatment have swiftly alleviated systemic inflammation of Takayasu's arteritis (TAK), while subclinical vascular inflammation and the ensuing arterial remodeling continue to present unresolved challenges in TAK. The phenotypic switching of vascular smooth muscle cells (VSMC) is regarded as the first step in vascular pathology and contributes to arterial remodeling. Exosomes facilitate the transfer and exchange of proteins and specific nucleic acids, thereby playing a significant role in intercellular communication. Little is known about the modulatory role of serum exosomes in phenotypic switching of VSMC and vascular remodeling in TAK. METHODS Serum exosomes isolated from TAK patients were co-cultured with VSMC to identify the modulatory role of exosomes. VSMC were transfected with miR-199a-5p mimic and inhibitor. CCK8 assays and EdU assays were performed to measure proliferative ability. The migration of VSMC was evaluated by scratch assays and transwell migration assays. The flow cytometry was employed to identify apoptosis of VSMC. Dual-luciferase reporter assay, RNA immunoprecipitation assay and fluorescence in situ hybridization were utilized to validate the target gene of miR-199a-5p. The correlational analysis was conducted among exosome miRNA, serum MMP2, TIMP2 and clinical parameters in TAK patients. RESULTS The coculture of VSMC with serum exosome mediated dedifferentiation of VSMC. Through gain- and loss-of-function approaches, miR-199a-5p over-expression significantly increased expression of VSMC marker genes and inhibited VSMC proliferation and migration, whilst the opposite effect was observed when endogenous miR-199a-5p was knocked down. The overexpression of miR-199a-5p suppressed VSMC apoptosis. Further, MMP2 serves as functional target gene of miR-199a-5p. The correlation analyses revealed an inverse correlation between Vasculitis Damage Index and exosome miR-199a-5p level or serum MMP2, which requires validation in a larger cohort. CONCLUSION Our study indicated that the miR-199a-5p/MMP2 pathway played a role in inhibiting the migration, proliferation and apoptosis of VSMC. The decreased secretion of MMP2 may potentially prompt the intimal infiltration of inflammatory cells within the vascular wall, offering a novel therapeutic opportunity by tackling both inflammatory responses and the neointimal overgrowth associated with TAK arterial damage. Moreover, exosome miR-199a-5p and MMP2 derived from serum possess potential as future biomarkers for vascular injury.
Collapse
Affiliation(s)
- Shuning Guo
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Jiehan Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shurui Pang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China.
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China.
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| |
Collapse
|
20
|
Paz AA, Jiménez TA, Ibarra-Gonzalez J, Astudillo-Maya C, Beñaldo FA, Figueroa EG, Llanos AJ, Gonzalez-Candia A, Herrera EA. Gestational hypoxia elicits long-term cardiovascular dysfunction in female guinea pigs. Life Sci 2025; 361:123282. [PMID: 39615619 DOI: 10.1016/j.lfs.2024.123282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Gestational hypoxia (GH) has been implicated in the developmental programming of cardiovascular diseases (CVDs) in the offspring, with most studies focusing on males, conversely, the effects on female cardiovascular health remain understudied. We aimed to investigate the impact of GH on the cardiovascular system of female guinea pig offspring from the early postnatal period to adulthood. METHODS Pregnant guinea pigs were subjected to normoxic or hypoxic conditions from gestational day 30 until delivery (∼70 days). Female offspring were monitored with biometric parameters and peripheral vascular function (ultrasound) from birth to one year old. In addition, we assessed cardiovascular structure, oxidative stress, inflammatory state (IHC, qPCR, and immunoblot assays), and thoracic aorta reactivity (wire-myography) at one year of age. KEY FINDINGS GH increased heart rate and peripheral pulsatility index. At one year old, GH-exposed females exhibited cardiac remodeling, characterized by increased left ventricular luminal area and coronary artery muscle occupation. Furthermore, GH increased aortic vascular wall, intima-media thickness and contractile capacity. This was accompanied by reduced endothelium-dependent vasodilation and enhanced oxidative stress. Additionally, GH increased collagen deposition and oxidative stress in the right ventricle, accompanied by reduced antioxidant enzymes expression and reduced inflammatory mediator levels. SIGNIFICANCE GH exerts long-lasting effects on the cardiovascular health of female guinea pig offspring, contributing to cardiac remodeling, vascular dysfunction, oxidative stress, and inflammatory changes. These findings highlight the importance of GH as a risk factor for developing CVDs in female offspring and emphasize the need for sex-specific interventions to mitigate adverse long-term gestational effects.
Collapse
Affiliation(s)
- Adolfo A Paz
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Tamara A Jiménez
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Julieta Ibarra-Gonzalez
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Cristian Astudillo-Maya
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Felipe A Beñaldo
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Esteban G Figueroa
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile
| | - Aníbal J Llanos
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Putre, Chile
| | | | - Emilio A Herrera
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Putre, Chile.
| |
Collapse
|
21
|
Wang Y, Zhang J, Wang Y, Wu F, Song B, Li J, Lin Q, Xie Y, Xia Y, An X, Liao J. Ubiquitin-like modifier-activating enzyme 1 as a potential therapeutic target for aortic dissection. Int Immunopharmacol 2025; 145:113742. [PMID: 39637577 DOI: 10.1016/j.intimp.2024.113742] [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: 09/29/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Aortic dissection (AD) is a life-threatening aortopathy with no specific pharmacological therapy. Ubiquitination, a highly orchestrated enzymatic cascade involving sequential E1-E2-E3 interactions, is suggested to contribute to the disease pathogenesis. However, the specific role of E1 enzymes in AD progression remains unknown. In this study, we analyzed the aortic transcriptional profiles of a human ascending dissection dataset (GSE52093) and identified ubiquitin-like modifier-activating enzyme 1 (UBA1) as a significantly up-regulated E1 enzyme in human AD. This finding was further corroborated by immunohistochemistry and RT-qPCR in a mouse model of AD induced by β-aminopropionitrile (BAPN). Treatment of TAK-243, a specific UBA1 inhibitor, prevented BAPN-induced AD formation in mice and attenuated aortic medial degeneration, as evidenced by decreased elastin fragmentation (evaluated by EVG scoring), reduced vascular smooth muscle cell loss (visualized by α-SMA immunohistochemistry), and less extracellular matrix degradation (indicated by diminished MMP2 and MMP9 expression in immunohistochemistry and RT-qPCR). Furthermore, TAK-243 treatment attenuated lesional macrophage accumulation and activation, as demonstrated by CD68 immunohistochemistry and RT-qPCR analysis of aortic pro-inflammatory cytokine expression. In vitro, UBA1 activation was observed in macrophages (RAW264.7 cells) treated with angiotensin II (AngII), and TAK-243 significantly reduced AngII-induced macrophage activation, at least partially through the inhibition of IκBα and NF-κB p65 phosphorylation. In conclusion, we demonstrate that UBA1 may facilitate AD progression by promoting macrophage activation via the NF-κB signaling pathway. These findings reveal a pathogenic role for the E1 enzyme UBA1 in AD and show a pharmacological potential of UBA1-targeted therapy against this disease.
Collapse
Affiliation(s)
- Yao Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jinjin Zhang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yunsong Wang
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Feng Wu
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Baoshen Song
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jiatian Li
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Qiuyue Lin
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yunpeng Xie
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yunlong Xia
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiangbo An
- Department of Interventional Therapy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Jiawei Liao
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
22
|
Li JP, He WB, Xu SW, Liu JZ, Huang ZQ, Li CP, Huang CL, Mai PB, Geng DF, Qiu XF, Liu ZY, Zhang K, Zhou SX. FSTL1 protects against acute aortic dissection by suppressing vascular smooth muscle cell phenotypic switching and degradation of the extracellular matrix. J Mol Cell Cardiol 2025; 198:60-73. [PMID: 39631359 DOI: 10.1016/j.yjmcc.2024.11.008] [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: 06/07/2024] [Revised: 11/10/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024]
Abstract
Acute aortic dissection (AAD) is a life-threatening cardiovascular emergency, which is closely related to the vascular smooth muscle cells (VSMCs) phenotypic switching and extracellular matrix (ECM) degradation. Previous studies have found that the secreted extracellular glycoprotein Follistatin-like 1 (FSTL1) is demonstrated as a protective factor for cardiovascular diseases. However, the role of FSTL1 in AAD remains elusive. We aimed to investigate whether FSTL1 could regulate VSMCs phenotypic switching and ECM degradation in AAD. Firstly, we found that FSTL1 expression in aorta was significantly decreased in human AAD examined by western blot and immunohistochemical staining. Then we established a mouse AAD model by administering β-aminopropionitrile (BAPN) dissolved in drinking water for 28 days. We found that FSTL1 expression in aorta was also decreased in mouse AAD. Exogenous supplement with recombinant human FSTL1 protein could rescue VSMCs phenotypic switching and ECM degradation to reduce the occurrence and progression of mouse AAD. In vitro, FSTL1 protein and adenovirus overexpressing FSTL1 (ad-FSTL1) reversed the primary VSMCs phenotypic switching and decreased the expression of MMP2 induced by PDGF-BB. Knocking down FSTL1 initiates VSMCs phenotypic switching and increases the expression of MMP2. In terms of mechanisms, AMPK phosphorylation was decreased and could be improved by FSTL1 protein in mouse AAD. FSTL1 protein and ad-FSTL1 reversed the decreased AMPK phosphorylation induced by PDGF-BB in primary VSMCs. These findings indicate that FSTL1 protects against VSMCs phenotypic switching and ECM degradation in AAD, and targeting FSTL1 may be a potential new strategy for prevention and treatment of AAD.
Collapse
Affiliation(s)
- Jun-Ping Li
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong 510317, PR China; Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China.
| | - Wan-Bing He
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China.
| | - Shu-Wan Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China.
| | - Juan-Zhang Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China; Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China.
| | - Zhao-Qi Huang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China; Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China.
| | - Chang-Ping Li
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China; Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China.
| | - Chun-Ling Huang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China.
| | - Pei-Biao Mai
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences (Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen, Guangdong 518000, China.
| | - Deng-Feng Geng
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China; Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China.
| | - Xiao-Fu Qiu
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong 510317, PR China
| | - Zhao-Yu Liu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China.
| | - Kun Zhang
- Department of Cardiology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Shu-Xian Zhou
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China; Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China.
| |
Collapse
|
23
|
Zhou S, Ju S, Li X, Ruan C, Dong Z. Revealing the roles of IL-7R in abdominal aortic aneurysm through integrated analysis of single-cell RNA-seq and bulk RNA-seq. Biochem Biophys Res Commun 2024; 741:151042. [PMID: 39586133 DOI: 10.1016/j.bbrc.2024.151042] [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: 09/12/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a common cardiovascular disease in the elderly, but there are still no therapeutic targets for this disease. In this study, we collected and analyzed bulk RNA sequencing (bulk RNA-seq) and single-cell RNA sequencing (scRNA-seq) data of AAA from the Gene Expression Omnibus (GEO) database. The immune infiltration-related genes were identified and categorized into various cell types, revealing potential key genes and pathways. Examination of three bulk RNA-seq datasets revealed a total of 4087 differentially expressed genes. The expression levels of the immune-related genes IL-7R were significantly elevated in AAA tissues across all three datasets. Furthermore, scRNA-Seq analysis revealed increased expression of IL-7R in CD4+ memory cells within AAA tissues. Immunofluorescence staining corroborated these findings, demonstrating increased expression of IL-7R in CD4+ T cells in AAA tissues. In vitro, activation of IL-7R elevated the activation of JAK/STAT pathway and phenotypic switching in SMCs, while inhibition of IL-7R abolished these effects and suppressed the secretion of IFN-γ. In conclusion, the activation of IL-7R in CD4+ T cells is a key contributor to the pathogenesis of AAA, as it can promote secretion of IFN-γ via JAK/STAT pathway and induce phenotypic switching of SMCs.
Collapse
Affiliation(s)
- Siyuan Zhou
- Jinshan Hospital, Fudan University, Shanghai, China; Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Shuai Ju
- Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Li
- Jinshan Hospital, Fudan University, Shanghai, China
| | - Chengchao Ruan
- Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Zhihui Dong
- Jinshan Hospital, Fudan University, Shanghai, China; Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
24
|
Doukas P, Hruschka B, Bassett C, Buhl EM, Simon F, Saraber P, Jacobs MJ, Uhl C, Schurgers LJ, Gombert A. Distribution and Maturity of Medial Collagen Fibers in Thoracoabdominal Post-Dissection Aortic Aneurysms: A Comparative Study of Marfan and Non-Marfan Patients. Int J Mol Sci 2024; 26:14. [PMID: 39795873 PMCID: PMC11720456 DOI: 10.3390/ijms26010014] [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: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/13/2025] Open
Abstract
Thoracoabdominal aortic aneurysms (TAAAs) are rare but serious conditions characterized by dilation of the aorta characterized by remodeling of the vessel wall, with changes in the elastin and collagen content. Individuals with Marfan syndrome have a genetic predisposition for elastic fiber fragmentation and elastin degradation and are prone to early aneurysm formation and progression. Our objective was to analyze the medial collagen characteristics through histological, polarized light microscopy, and electron microscopy methods across the thoracic and abdominal aorta in twenty-five patients undergoing open surgical repair, including nine with Marfan syndrome. While age at surgery differed significantly between the groups, maximum aortic diameter and aneurysm extent did not. Collagen content increased from thoracic to infrarenal segments in both cohorts, with non-Marfan patients exhibiting higher collagen percentages, notably in the infrarenal aorta (729.3 nm vs. 1068.3 nm, p = 0.02). Both groups predominantly displayed mature collagen fibers, with the suprarenal segment containing the highest proportion of less mature fibers. Electron microscopy revealed comparable collagen fibril diameters across segments irrespective of Marfan status. Our findings underscore non-uniform histological patterns in TAAAs and suggest that ECM remodeling involves mature collagen deposition, albeit with lower collagen content observed in the infrarenal aorta of Marfan patients.
Collapse
Affiliation(s)
- Panagiotis Doukas
- Department of Vascular Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (B.H.); (C.B.); (M.J.J.); (C.U.); (A.G.)
| | - Bernhard Hruschka
- Department of Vascular Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (B.H.); (C.B.); (M.J.J.); (C.U.); (A.G.)
| | - Cathryn Bassett
- Department of Vascular Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (B.H.); (C.B.); (M.J.J.); (C.U.); (A.G.)
| | - Eva Miriam Buhl
- Institute of Pathology, Electron Microscopy Facility, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Florian Simon
- Clinic for Vascular and Endovascular Surgery, University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, 40225 Düsseldorf, Germany;
| | - Pepijn Saraber
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (P.S.); (L.J.S.)
| | - Michael Johan Jacobs
- Department of Vascular Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (B.H.); (C.B.); (M.J.J.); (C.U.); (A.G.)
| | - Christian Uhl
- Department of Vascular Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (B.H.); (C.B.); (M.J.J.); (C.U.); (A.G.)
| | - Leon J. Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (P.S.); (L.J.S.)
| | - Alexander Gombert
- Department of Vascular Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (B.H.); (C.B.); (M.J.J.); (C.U.); (A.G.)
| |
Collapse
|
25
|
Wu H, Li Z, Yang L, He L, Liu H, Yang S, Xu Q, Li Y, Li W, Li Y, Gong Z, Shen Y, Yang X, Huang J, Yu F, Li L, Zhu J, Sun L, Fu Y, Kong W. ANK Deficiency-Mediated Cytosolic Citrate Accumulation Promotes Aortic Aneurysm. Circ Res 2024; 135:1175-1192. [PMID: 39513269 DOI: 10.1161/circresaha.124.325152] [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: 08/01/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Disturbed metabolism and transport of citrate play significant roles in various pathologies. However, vascular citrate regulation and its potential role in aortic aneurysm (AA) development remain poorly understood. METHODS Untargeted metabolomics by mass spectrometry was applied to identify upregulated metabolites of the tricarboxylic acid cycle in AA tissues of mice. To investigate the role of citrate and its transporter ANK (progressive ankylosis protein) in AA development, vascular smooth muscle cell (VSMC)-specific Ank-knockout mice were used in both Ang II (angiotensin II)- and CaPO4-induced AA models. RESULTS Citrate was abnormally increased in both human and murine aneurysmal tissues, which was associated with downregulation of ANK, a citrate membrane transporter, in VSMCs. The knockout of Ank in VSMCs promoted AA formation in both Ang II- and CaPO4-induced AA models, while its overexpression inhibited the development of aneurysms. Mechanistically, ANK deficiency in VSMCs caused abnormal cytosolic accumulation of citrate, which was cleaved into acetyl coenzyme A and thus intensified histone acetylation at H3K23, H3K27, and H4K5. Cleavage under target and tagmentation analysis further identified that ANK deficiency-induced histone acetylation activated the transcription of inflammatory genes in VSMCs and thus promoted a citrate-related proinflammatory VSMC phenotype during aneurysm diseases. Accordingly, suppressing citrate cleavage to acetyl coenzyme A downregulated inflammatory gene expression in VSMCs and restricted ANK deficiency-aggravated AA formation. CONCLUSIONS Our studies define the pathogenic role of ANK deficiency-induced cytosolic citrate accumulation in AA pathogenesis and an undescribed citrate-related proinflammatory VSMC phenotype. Targeting ANK-mediated citrate transport may emerge as a novel diagnostic and therapeutic strategy in AA.
Collapse
MESH Headings
- Animals
- Mice
- Citric Acid/metabolism
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice, Knockout
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/genetics
- Aortic Aneurysm/pathology
- Aortic Aneurysm/etiology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Inbred C57BL
- Cytosol/metabolism
- Male
- Cells, Cultured
- Acetylation
- Acetyl Coenzyme A/metabolism
- Disease Models, Animal
- Histones/metabolism
Collapse
Affiliation(s)
- Hao Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China (L.Y.)
| | - Lin He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Hao Liu
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, China (H.L., Q.X., J.Z.)
| | - Shiyu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Qinfeng Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yanjie Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wenqiang Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yiran Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
- Hwamei College of Life and Health Sciences, Zhejiang Wanli University, Ningbo, China (Z.G.)
| | - Yicong Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xueyuan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiaqi Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Li Li
- Department of Pathology, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.L.)
| | - Junming Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Luyang Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | | |
Collapse
|
26
|
Yong X, Hu X, Kang T, Deng Y, Li S, Yu S, Hou Y, You J, Dai X, Zhang J, Zhang J, Zhou J, Zhang S, Zheng J, Yang Q, Li J. Identification of CCR7 and CBX6 as key biomarkers in abdominal aortic aneurysm: Insights from multi-omics data and machine learning analysis. IET Syst Biol 2024; 18:250-260. [PMID: 39602349 DOI: 10.1049/syb2.12106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/30/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a severe vascular condition, marked by the progressive dilation of the abdominal aorta, leading to rupture if untreated. The objective of this study was to identify key biomarkers and decipher the immune mechanisms underlying AAA utilising multi-omics data analysis and machine learning techniques. Single-cell RNA sequencing disclosed a heightened presence of macrophages and CD8-positive alpha-beta T cells in AAA, highlighting their critical role in disease pathogenesis. Analysis of cell-cell communication highlighted augmented interactions between macrophages and dendritic cells derived from monocytes. Enrichment analysis of differential expression gene indicated substantial involvement of immune and metabolic pathways in AAA pathogenesis. Machine learning techniques identified CCR7 and CBX6 as key candidate biomarkers. In AAA, CCR7 expression is upregulated, whereas CBX6 expression is downregulated, both showing significant correlations with immune cell infiltration. These findings provide valuable insights into the molecular mechanisms underlying AAA and suggest potential biomarkers for diagnosis and therapeutic intervention.
Collapse
Affiliation(s)
- Xi Yong
- Department of Vascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- The First Affiliated Hospital, Jinan University, Guangzhou, China
- Hepatobiliary, Pancreatic and Intestinal Research Institute of North Sichuan Medical College, Nanchong, China
| | - Xuerui Hu
- Department of Endocrine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tengyao Kang
- Department of Vascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Yanpiao Deng
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Sixuan Li
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Shuihan Yu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Yani Hou
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Jin You
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Xiaohe Dai
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Jialin Zhang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Junjia Zhang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Junlin Zhou
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Siyu Zhang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Jianghua Zheng
- Department of Vascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Qin Yang
- Department of Infectious Diseases, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jingdong Li
- Hepatobiliary, Pancreatic and Intestinal Research Institute of North Sichuan Medical College, Nanchong, China
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
27
|
Han Y, Cui Y, Liu J, Wang D, Zou G, Qi X, Meng J, Huang X, He H, Li X. Single-Cell RNA-Seq Reveals Injuries in Aortic Dissection and Identifies PDGF Signalling Pathway as a Potential Therapeutic Target. J Cell Mol Med 2024; 28:e70293. [PMID: 39720896 DOI: 10.1111/jcmm.70293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 12/26/2024] Open
Abstract
Aortic dissection (AD) represents a critical condition characterised by a tear in the inner lining of the aorta, leading to the leakage of blood into the layers of the aortic wall, posing a significant risk to life. However, the pathogenesis is unclear. In this study, scRNA-seq was applied to cells derived from aortas of both AD and non-AD donors (control) to unveil the cellular landscape. ScRNA-seq data uncover significant cellular heterogeneity in AD aortas. Specifically, we observed an accumulation of CD4+ T cells, which contributed to inflammation and cell death, and abnormal collagen formation mediated by fibroblast cells in AD. Moreover, we revealed a greater prevalence of cell death, oxidative stress and senescence in AD aorta cells. Furthermore, we found a decrease in the percentage of vascular stem cells (VSCs), along with a repression in their ability to differentiate into contractile vascular smooth muscle cells (VSMCs). Finally, our data demonstrated that the PDGF signalling pathway was activated in AD. We found that PDGF activation could lead to VSMCs aberrant switch from contractile to synthetic phenotype, which could be ameliorated by PDGF inhibitor. This underscores the potential of the PDGF as a therapeutic target for AD. In summary, our study highlights the cellular heterogeneity and associated injuries within aortas affected by AD, including cell death, oxidative stress, senescence and dysregulation of signalling pathways influencing the aberrant phenotypic switch of VSMCs. These insights offer valuable contributions to understanding the molecular mechanisms underlying AD and present new avenues for therapeutic intervention in this condition.
Collapse
Affiliation(s)
- Yichi Han
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- Department of Critical Care Medicine, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Afliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Yongji Cui
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Juli Liu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Dingchen Wang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Guoxiang Zou
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Xin Qi
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Jinxiu Meng
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiaoran Huang
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Haiwei He
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Xin Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
28
|
Li A, Feng Z, Fu S, Ma Z, Zhang H, Zhao Z. Dissecting causal relationships between immune cells, blood metabolites, and aortic dissection: A mediation Mendelian randomization study. IJC HEART & VASCULATURE 2024; 55:101530. [PMID: 39483148 PMCID: PMC11525623 DOI: 10.1016/j.ijcha.2024.101530] [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: 07/24/2024] [Revised: 09/09/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024]
Abstract
Background There exists a robust correlation between the infiltration of immune cells and the pathogenesis of aortic dissection (AD). Moreover, blood metabolites serve as immunomodulatory agents within the organism, influencing the immune system's response and potentially playing a role in the development of AD. Nevertheless, the intricate genetic causal nexus between specific immune cells, blood metabolites, and AD remains partially elucidated. Objectives This study aims to elucidate the causal relationships between specific immune cell types and the risk of developing AD, mediated by blood metabolites, using Mendelian Randomization (MR) methods. Methods We undertook a comprehensive investigation of 731 immune cell types through the analysis of published genome-wide association studies (GWAS). Our methodology hinged on the application of two-sample Mendelian randomization (MR) and mediator MR analyses, prioritizing blood metabolites as potential intermediary factors and AD as the principal outcome of interest. The primary statistical method employed was inverse variance-weighted estimation, complemented by a variety of sensitivity analyses to reinforce our conclusions. The entirety of our statistical analyses was executed on the R software platform. Results Our analyses elucidated that three immune cell types exhibited a positive correlation with the incidence of AD, whereas two immune cell types were inversely associated with AD risk. Significantly, our mediation Mendelian randomization (MR) findings identified Benzoate as a pivotal mediator in the influence of CD19 on IgD - CD38br cells on AD, with a mediation proportion of 5.38 %. Additionally, N-acetylproline was determined to mediate the effect of CD24 on IgD- CD38- cells on AD, accounting for a mediation proportion of 13.70 %. Furthermore, Carnitine C5:1 was found to mediate the effect of CD28 on secreting T regulatory (Treg) cells on AD, with a mediation proportion of 17.80 %. Conclusions These findings offer a nuanced understanding of the pathophysiological mechanisms underlying AD, thereby advancing the precision medicine paradigm in the clinical management of AD.Abbreviations: AD: aortic dissection; AA: aortic aneurysm; GWAS: genome-wide association study; MR: Mendelian randomization; TSMR: two-step Mendelian randomization; Treg: secreting T regulatory cell; VSMC: vascular smooth muscle cell; MMP: matrix metalloproteinase; ROS: reactive oxygen species; IV: instrumental variable; SNP: single-nucleotide polymorphism; IVW: inverse variance weighted; LDSC: linkage disequilibrium score regression; OR: odds ratio; CI: confidence interval; LD: linkage disequilibrium; AC: absolute cell; MFI: median fluorescence intensity; MP: morphological parameter; RC: relative cell; CLSA: Canadian Longitudinal Study of Aging; Lp(a): Lipoprotein a; OxPL: oxidised phospholipid; NMDAR: N-methyl-d-aspartate glutamate receptor; STROBE-MR: Strengthening the Reporting of Observational Studies in Epidemiology using Mendelian Randomization.
Collapse
Affiliation(s)
- Ao Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China(Anhui Provincial Hospital), Anhui, Hefei 230001, China
| | - ZiAn Feng
- Graduate School, University of Science and Technology of China, Anhui, Hefei 230001, China
| | - ShiHao Fu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China(Anhui Provincial Hospital), Anhui, Hefei 230001, China
| | - ZhenXiao Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China(Anhui Provincial Hospital), Anhui, Hefei 230001, China
| | - HaiYang Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China(Anhui Provincial Hospital), Anhui, Hefei 230001, China
| | - ZhiWei Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China(Anhui Provincial Hospital), Anhui, Hefei 230001, China
| |
Collapse
|
29
|
Su J, Cheng F, Yuan W. Unraveling the cGAS/STING signaling mechanism: impact on glycerolipid metabolism and diseases. Front Med (Lausanne) 2024; 11:1512916. [PMID: 39669992 PMCID: PMC11634591 DOI: 10.3389/fmed.2024.1512916] [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: 10/17/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
The cyclic GMP-AMP synthase (cGAS) and its downstream effector, the stimulator of interferon genes (STING), are crucial components of the innate immune response, traditionally recognized for their role in detecting cytosolic DNA from pathogens and damaged host cells. However, recent research indicates that the cGAS-STING pathway also significantly impacts metabolic processes, particularly glycerolipid metabolism. Glycerolipids are essential for energy storage and cellular membrane integrity, and their dysregulation is linked to metabolic disorders such as obesity, insulin resistance, and non-alcoholic fatty liver disease (NAFLD). Both cGAS and STING are expressed in various metabolic tissues, suggesting a potential role in lipid homeostasis. Chronic activation of the cGAS-STING pathway may promote inflammatory states that exacerbate insulin resistance and lipid accumulation, forming a feedback loop of metabolic dysfunction. This review explores the emerging relationship between cGAS/STING signaling and glycerolipid metabolism, discussing the mechanisms through which this pathway influences lipid regulation and the potential for therapeutic interventions. By integrating insights from immunology and metabolism, we aim to provide a comprehensive understanding of how the cGAS-STING axis may serve as a novel target for addressing metabolic disorders and enhancing metabolic health outcomes.
Collapse
Affiliation(s)
- Jie Su
- Department of Cardiology, Hospital of Jiangsu University, Zhenjiang, China
- The British Heart Foundation Centre of Excellence, St Thomas’ Hospital, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, The Rayne Institute, London, United Kingdom
| | - Fuyu Cheng
- Department of Cardiology, Hospital of Jiangsu University, Zhenjiang, China
- School of Engineering and Material Sciences, Digital Environment Research Institute, Queen Mary University of London, London, United Kingdom
| | - Wei Yuan
- Department of Cardiology, Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
30
|
Wang Y, Li G, Zeng F, Li K, Pan W, Zhang M, Yang H, Jin J. Association of gene polymorphism in ERG rs2836411 with anemia and susceptibility to aortic dissection. BMC Cardiovasc Disord 2024; 24:682. [PMID: 39604843 PMCID: PMC11600712 DOI: 10.1186/s12872-024-04345-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The erythroblast transformation-specific related gene (ERG) is expressed in hematopoietic stem and progenitor cells and endothelial cells. This study aimed to investigate if ERG rs2836411 is a novel genetic locus associated with anemia and aortic dissection (AD). METHOD A case-control trial was conducted to evaluate the association between ERG rs2836411 polymorphism, anemia, and AD risk. The ERG rs2836411 polymorphism was analyzed using Sanger dideoxy chain termination sequencing on genomic DNA extracted from whole blood. Serum erythropoietin (EPO) and interleukin-6 (IL-6) concentrations were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS 119 preoperative AD patients and 119 healthy controls were enrolled, with age and sex matched. Anemia was found independently associated with AD presence (Odds ratio (OR) 20.82, p < 0.001). Remarkably, T carriers (CT + TT) of ERG rs2836411 were associated with anemia in AD patients (OR 2.81, p = 0.013) but not in controls. After adjusting for conventional risk factors including age, sex, smoking and hypertension status, T carriers (CT + TT) were independently associated with AD presence (OR 2.20, p = 0.015), but were not associated if anemia was further adjusted. While EPO concentration was higher in AD patients and was associated with AD presence (OR 1.09, p = 0.006), no difference in EPO levels was observed between the ERG genotypes of AD patients. CONCLUSIONS T carriers (CT + TT) of ERG rs2836411 are independently associated with anemia and AD presence. The association between ERG rs2836411 polymorphism and susceptibility to AD may be mediated by anemia. Further studies are warranted to validate whether this association is causal.
Collapse
Affiliation(s)
- Ying Wang
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, China
| | - Gaoshan Li
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, China
| | - Fangzheng Zeng
- Department of Emergency Medicine, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, China
| | - Kunyan Li
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, China
| | - Wenxu Pan
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, China
| | - Mingle Zhang
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, China
| | - Hao Yang
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, China
| | - Jun Jin
- Department of Cardiology, The Second Affiliated Hospital (Xinqiao Hospital) of Army Medical University, Chongqing, China.
| |
Collapse
|
31
|
Sukketsiri W, Tipmanee V, Rungruang P, Higashihara M, Sumi T, Moriyama T, Zaima N. Octanoic Acid and Decanoic Acid Inhibit Tunicamycin-Induced ER Stress in Rat Aortic Smooth Muscle Cells. Adv Pharmacol Pharm Sci 2024; 2024:9076988. [PMID: 39628939 PMCID: PMC11614520 DOI: 10.1155/adpp/9076988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 11/08/2024] [Indexed: 12/06/2024] Open
Abstract
ER stress is a crucial factor in the progression of vascular cell diseases. Notably, octanoic acid (OA; C8:0) and decanoic acid (DA; C10:0), prominent components of medium-chain fatty acids (MCFAs), may provide potential health benefits. However, their effects on vascular smooth muscle cells (VSMCs) remain unknown. Given the link between ER stress and vascular cell pathological conditions, the primary goal of this research is to investigate the protective effects of OA and DA against ER stress induction in rat aortic smooth muscle cells (RASMCs). To achieve this objective, RASMCs were pretreated with OA and DA at concentrations of 250 and 500 μM for 24 h. Subsequently, the cells were exposed to 1 μg/mL of tunicamycin, an ER stress inducer, for an additional 24 h. Apoptosis was assessed using DAPI staining, while DCFH2-DA probe was used to measure ROS levels. Furthermore, the gene expression of ER stress markers, such as CHOP, GRP78, ATF4, and eIF2α, as well as contractile markers like αSMA and MYH11, was assessed using real-time reverse transcription polymerase chain reaction. Moreover, the αSMA protein level was measured using immunocytochemistry techniques. The study revealed that OA and DA significantly mitigated cell death caused by tunicamycin, decreased ROS production, and inhibited the gene expression of ER stress markers (CHOP, GRP78, and eIF2α). Notably, OA and DA also inhibited the expression of contractile genes (α-SMA and MYH11) and reduced the number of α-SMA-positive cells in tunicamycin-treated RASMCs. These findings indicate that OA and DA offer protection against ER stress-stimulated cell death and ROS generation in VSMCs, thereby supporting their potential therapeutic applications for safeguarding these cells.
Collapse
Affiliation(s)
- Wanida Sukketsiri
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Varomyalin Tipmanee
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Panlekha Rungruang
- Molecular Medicine Program, Multidisciplinary Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Mayo Higashihara
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara, Japan
| | - Tomoko Sumi
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara, Japan
| | - Tatsuya Moriyama
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara, Japan
- Agricultural Technology and Innovation Research Institute, Kindai University, Nara, Japan
| | - Nobuhiro Zaima
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara, Japan
- Agricultural Technology and Innovation Research Institute, Kindai University, Nara, Japan
| |
Collapse
|
32
|
Ma XR, Conley SD, Kosicki M, Bredikhin D, Cui R, Tran S, Sheth MU, Qiu WL, Chen S, Kundu S, Kang HY, Amgalan D, Munger CJ, Duan L, Dang K, Rubio OM, Kany S, Zamirpour S, DePaolo J, Padmanabhan A, Olgin J, Damrauer S, Andersson R, Gu M, Priest JR, Quertermous T, Qiu X, Rabinovitch M, Visel A, Pennacchio L, Kundaje A, Glass IA, Gifford CA, Pirruccello JP, Goodyer WR, Engreitz JM. Molecular convergence of risk variants for congenital heart defects leveraging a regulatory map of the human fetal heart. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.20.24317557. [PMID: 39606363 PMCID: PMC11601760 DOI: 10.1101/2024.11.20.24317557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Congenital heart defects (CHD) arise in part due to inherited genetic variants that alter genes and noncoding regulatory elements in the human genome. These variants are thought to act during fetal development to influence the formation of different heart structures. However, identifying the genes, pathways, and cell types that mediate these effects has been challenging due to the immense diversity of cell types involved in heart development as well as the superimposed complexities of interpreting noncoding sequences. As such, understanding the molecular functions of both noncoding and coding variants remains paramount to our fundamental understanding of cardiac development and CHD. Here, we created a gene regulation map of the healthy human fetal heart across developmental time, and applied it to interpret the functions of variants associated with CHD and quantitative cardiac traits. We collected single-cell multiomic data from 734,000 single cells sampled from 41 fetal hearts spanning post-conception weeks 6 to 22, enabling the construction of gene regulation maps in 90 cardiac cell types and states, including rare populations of cardiac conduction cells. Through an unbiased analysis of all 90 cell types, we find that both rare coding variants associated with CHD and common noncoding variants associated with valve traits converge to affect valvular interstitial cells (VICs). VICs are enriched for high expression of known CHD genes previously identified through mapping of rare coding variants. Eight CHD genes, as well as other genes in similar molecular pathways, are linked to common noncoding variants associated with other valve diseases or traits via enhancers in VICs. In addition, certain common noncoding variants impact enhancers with activities highly specific to particular subanatomic structures in the heart, illuminating how such variants can impact specific aspects of heart structure and function. Together, these results implicate new enhancers, genes, and cell types in the genetic etiology of CHD, identify molecular convergence of common noncoding and rare coding variants on VICs, and suggest a more expansive view of the cell types instrumental in genetic risk for CHD, beyond the working cardiomyocyte. This regulatory map of the human fetal heart will provide a foundational resource for understanding cardiac development, interpreting genetic variants associated with heart disease, and discovering targets for cell-type specific therapies.
Collapse
Affiliation(s)
- X Rosa Ma
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Stephanie D Conley
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Michael Kosicki
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Danila Bredikhin
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ran Cui
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steven Tran
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Maya U Sheth
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Wei-Lin Qiu
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Sijie Chen
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Soumya Kundu
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Helen Y Kang
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Current address: PhD Program in Computational and Systems Biology, MIT, Cambridge, MA, USA
| | - Dulguun Amgalan
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
| | - Chad J Munger
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lauren Duan
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Katherine Dang
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Oriane Matthys Rubio
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Siavash Zamirpour
- School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - John DePaolo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arun Padmanabhan
- Gladstone Institutes, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco School of Medicine, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jeffrey Olgin
- Division of Cardiology, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Scott Damrauer
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robin Andersson
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - James R Priest
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Tenaya Therapeutics, South San Francisco, CA, USA
| | - Thomas Quertermous
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Xiaojie Qiu
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Stanford Bio-X, Stanford University, Stanford, CA, USA
| | - Marlene Rabinovitch
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- School of Natural Sciences, University of California, Merced, Merced, CA, USA
| | - Len Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA, 94720, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Ian A Glass
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Department of Pediatrics and Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Casey A Gifford
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Cardiology, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Bakar Computation Health Sciences Institute, University of California, San Francisco, CA, USA
| | - William R Goodyer
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Jesse M Engreitz
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
33
|
Tomida S, Ishima T, Nagai R, Aizawa K. T-Type Voltage-Gated Calcium Channels: Potential Regulators of Smooth Muscle Contractility. Int J Mol Sci 2024; 25:12420. [PMID: 39596484 PMCID: PMC11594734 DOI: 10.3390/ijms252212420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/16/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging evidence has indicated a possible link between attenuation of contractility in aortic smooth muscle cells and pathogenesis of aortic dissection, as revealed through comprehensive, multi-omic analyses of familial thoracic aortic aneurysm and dissection models. While L-type voltage-gated calcium channels have been extensively investigated for their roles in smooth muscle contraction, more recent investigations have suggested that downregulation of T-type voltage-gated calcium channels, rather than their L-type counterparts, may be more closely associated with impaired contractility observed in vascular smooth muscle cells. This review provides a detailed examination of T-type voltage-gated calcium channels, highlighting their structure, electrophysiology, biophysics, expression patterns, functional roles, and potential mechanisms through which their downregulation may contribute to reduced contractile function. Furthermore, the application of multi-omic approaches in investigating calcium channels is discussed.
Collapse
Affiliation(s)
- Shota Tomida
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan
- School of Medicine, Faculty of Medicine, Gunma University, Maebashi 371-8511, Japan
| | - Tamaki Ishima
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Ryozo Nagai
- Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Kenichi Aizawa
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan
- Clinical Pharmacology Center, Jichi Medical University Hospital, Shimotsuke 329-0498, Japan
- Division of Translational Research, Clinical Research Center, Jichi Medical University Hospital, Shimotsuke 329-0498, Japan
| |
Collapse
|
34
|
Yin Z, Zhang J, Zhao M, Liu J, Xu Y, Peng S, Pan W, Wei C, Zheng Z, Liu S, Qin JJ, Wan J, Wang M. EDIL3/Del-1 prevents aortic dissection through enhancing internalization and degradation of apoptotic vascular smooth muscle cells. Autophagy 2024; 20:2405-2425. [PMID: 38873925 PMCID: PMC11572282 DOI: 10.1080/15548627.2024.2367191] [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: 12/04/2023] [Revised: 05/28/2024] [Accepted: 06/08/2024] [Indexed: 06/15/2024] Open
Abstract
Thoracic aortic dissection (TAD) is a severe disease, characterized by numerous apoptotic vascular smooth muscle cells (VSMCs). EDIL3/Del-1 is a secreted protein involved in macrophage efferocytosis in acute inflammation. Here, we aimed to investigate whether EDIL3 promoted the internalization and degradation of apoptotic VSMCs during TAD. The levels of EDIL3 were decreased in the serum and aortic tissue from TAD mice. Global edil3 knockout (edil3-/-) mice and edil3-/- bone marrow chimeric mice exhibited a considerable exacerbation in β-aminopropionitrile monofumarate (BAPN)-induced TAD, accompanied with increased apoptotic VSMCs accumulating in the damaged aortic tissue. Two types of phagocytes, RAW264.7 cells and bone marrow-derived macrophages (BMDMs) were used for in vitro efferocytosis assay. edil3-deficient phagocytes exhibited inefficient internalization and degradation of apoptotic VSMCs. Instead, EDIL3 promoted the internalization phase through interacting with phosphatidylserine (PtdSer) on apoptotic VSMCs and binding to the macrophage ITGAV/αv-ITGB3/β3 integrin. In addition, EDIL3 accelerated the degradation phase through activating LC3-associated phagocytosis (LAP). Mechanically, following the engulfment, EDIL3 enhanced the activity of SMPD1/acid sphingomyelinase in the phagosome through blocking ITGAV-ITGB3 integrin, which facilitates phagosomal reactive oxygen species (ROS) production by NAPDH oxidase CYBB/NOX2. Furthermore, exogenous EDIL3 supplementation alleviated BAPN-induced TAD and promoted apoptotic cell clearance. EDIL3 may be a novel factor for the prevention and treatment of TAD.Abbreviations: BAPN: β-aminopropionitrile monofumarate; BMDM: bone marrow-derived macrophage; C12FDG: 5-dodecanoylaminofluorescein-di-β-D-galactopyranoside; CTRL: control; CYBB/NOX2: cytochrome b-245, beta polypeptide; DCFH-DA: 2',7'-dichlorofluorescin diacetate; EDIL3/Del-1: EGF-like repeats and discoidin I-like domains 3; EdU: 5-ethynyl-2'-deoxyuridine; EVG: elastic van Gieson; H&E: hematoxylin and eosin; IL: interleukin; LAP: LC3-associated phagocytosis; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; NAC: N-acetylcysteine; PtdSer: phosphatidylserine; rEDIL3: recombinant EDIL3; ROS: reactive oxygen species; SMPD1: sphingomyelin phosphodiesterase 1; TAD: thoracic aortic dissection; TEM: transmission electron microscopy; VSMC: vascular smooth muscle cell; WT: wild-type.
Collapse
Affiliation(s)
- Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Siqi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
35
|
Han X, Xu S, Hu K, Yu Y, Wang X, Qu C, Yang B, Liu X. Early growth response 1 exacerbates thoracic aortic aneurysm and dissection of mice by inducing the phenotypic switching of vascular smooth muscle cell through the activation of Krüppel-like factor 5. Acta Physiol (Oxf) 2024; 240:e14237. [PMID: 39345002 DOI: 10.1111/apha.14237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/15/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024]
Abstract
AIM Vascular smooth muscle cell (VSMC) phenotypic switching has been reported to regulate vascular function and thoracic aortic aneurysm and dissection (TAAD) progression. Early growth response 1 (Egr1) is associated with the differentiation of VSMCs. However, the mechanisms through which Egr1 participates in the regulation of VSMCs and progression of TAAD remain unknown. This study aimed to investigate the role of Egr1 in the phenotypic switching of VSMCs and the development of TAAD. METHODS Wild-type C57BL/6 and SMC-specific Egr1-knockout mice were used as experimental subjects and fed β-aminopropionitrile for 4 weeks to construct the TAAD model. Ultrasound and aortic staining were performed to examine the pathological features in thoracic aortic tissues. Transwell, wound healing, CCK8, and immunofluorescence assays detected the migration and proliferation of synthetic VSMCs. Egr1 was directly bound to the promoter of Krüppel-like factor 5 (KLF5) and promoted the expression of KLF5, which was validated by JASPAR database and dual-luciferase reporter assay. RESULTS Egr1 expression increased and was partially co-located with VSMCs in aortic tissues of mice with TAAD. SMC-specific Egr1 deficiency alleviated TAAD and inhibited the phenotypic switching of VSMC. Egr1 knockdown prevented the phenotypic switching of VSMCs and subsequently suppressed the migration and proliferation of synthetic VSMCs. The inhibitory effects of Egr1 deficiency on VSMCs were blunted once KLF5 was overexpressed. CONCLUSION Egr1 aggravated the development of TAAD by promoting the phenotypic switching of VSMCs via enhancing the transcriptional activation of KLF5. These results suggest that inhibition of SMC-specific Egr1 expression is a promising therapy for TAAD.
Collapse
MESH Headings
- Animals
- Early Growth Response Protein 1/metabolism
- Early Growth Response Protein 1/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Mice
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Phenotype
- Male
- Myocytes, Smooth Muscle/metabolism
- Disease Models, Animal
- Cell Proliferation
Collapse
Affiliation(s)
- Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Shengnan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Ke Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xiukun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| |
Collapse
|
36
|
Wang S, Shi H, Cheng Y, Jiang L, Lou Y, Kumar M, Sun M, Shao X, Zhao X, Wang B. Akkermansia muciniphila alleviates abdominal aortic aneurysms via restoring CITED2 activated by EPAS1. Infect Immun 2024; 92:e0017224. [PMID: 39207146 PMCID: PMC11477905 DOI: 10.1128/iai.00172-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening cardiovascular disease that has been linked to gut microbiome dysbiosis. Therefore, this study aims to investigate the effects of Akkermansia muciniphila (Am) on AAA mice and the biomolecules involved. AAA mice were generated using angiotensin II (Ang II), and 16sRNA sequencing was used to identify an altered abundance of microbiota in the feces of AAA mice. Vascular smooth muscle cell (VSMC) markers and apoptosis, and macrophage infiltration in mouse aortic tissues were examined. The abundance of Am was reduced in AAA mouse feces, and endothelial PAS domain-containing protein 1 (EPAS1) was downregulated in AAA mice and VSMC induced with Ang II. Am delayed AAA progression in mice, which was blunted by knockdown of EPAS1. EPAS1 was bound to the Cbp/p300-interacting transactivator 2 (CITED2) promoter and promoted CITED2 transcription. CITED2 reduced VSMC apoptosis and delayed AAA progression. Moreover, EPAS1 inhibited macrophage inflammatory response by promoting CITED2 transcription. In conclusion, gut microbiome dysbiosis in AAA induces EPAS1-mediated dysregulation of CITED2 to promote macrophage inflammatory response and VSMC apoptosis.
Collapse
MESH Headings
- Animals
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Aortic Aneurysm, Abdominal/microbiology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Mice
- Akkermansia
- Gastrointestinal Microbiome
- Trans-Activators/metabolism
- Trans-Activators/genetics
- Male
- Disease Models, Animal
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/microbiology
- Muscle, Smooth, Vascular/pathology
- Apoptosis
- Angiotensin II/metabolism
- Myocytes, Smooth Muscle/metabolism
- Macrophages/metabolism
- Macrophages/microbiology
- Macrophages/immunology
- Mice, Inbred C57BL
- Dysbiosis/microbiology
Collapse
Affiliation(s)
- Siqing Wang
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hang Shi
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Cheng
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lei Jiang
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yang Lou
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Manish Kumar
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Mingfei Sun
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xianze Shao
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xuan Zhao
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Baichun Wang
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
37
|
Wang J, Ye W, Zou J, Yang P, Jin M, Zheng Z, Zhou C, Qiu W, Lu J, Li C, Guo S, Xu Y, Huang Z, Liu P, Liu Z. Targeting the smooth muscle cell Keap1-Nrf2-GSDMD-pyroptosis axis by cryptotanshinone prevents abdominal aortic aneurysm formation. Theranostics 2024; 14:6516-6542. [PMID: 39479449 PMCID: PMC11519792 DOI: 10.7150/thno.98400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024] Open
Abstract
Rationale: Abdominal aortic aneurysm (AAA) is an inflammatory, fatal aortic disease that currently lacks any effective drugs. Cryptotanshinone (CTS) is a prominent and inexpensive bioactive substance derived from Salvia miltiorrhiza Bunge, a well-known medicinal herb for treating cardiovascular diseases through its potent anti-inflammatory properties. Nevertheless, the therapeutic effect of CTS on AAA formation remains unknown. Methods: To investigate the therapeutic effect of CTS in AAA, variety of experimental approaches were employed, majorly including AAA mouse model establishment, real-time polymerase chain reaction (PCR), RNA sequencing, western blot, co-immunoprecipitation, scanning/transmission electron microscopy (SEM/TEM), enzyme-linked immunosorbent assay (ELISA), seahorse analysis, immunohistochemistry, and confocal imaging. Results: In this study, we demonstrated that CTS suppressed the formation of AAA in apolipoprotein E knock-out (ApoE-/-) mice infused with Ang II. A combination of network pharmacology and whole transcriptome sequencing analysis indicated that activation of the Keap1-Nrf2 pathway and regulation of programmed cell death in vascular smooth muscle cells (VSMCs) are closely linked to the anti-AAA effect of CTS. Mechanistically, CTS promoted the transcription of Nrf2 target genes, particularly Hmox-1, which prevented the activation of NLRP3 and GSDMD-initiated pyroptosis in VSMCs, thereby mitigating VSMC inflammation and maintaining the VSMC contractile phenotype. Subsequently, by utilizing molecular docking, together with the cellular thermal shift assay (CETSA) and isothermal titration calorimetry (ITC), a particular binding site was established between CTS and Keap1 at Arg415. To confirm the binding site, site-directed mutagenesis was performed, which intriguingly showed that the Arg415 mutation eliminated the binding between CTS and the Keap1-Nrf2 protein and abrogated the antioxidant and anti-pyroptosis effects of CTS. Furthermore, VSMC-specific Nrf2 knockdown in mice dramatically reversed the protective action of CTS in AAA and the inhibitory effect of CTS on VSMC pyroptosis. Conclusion: Naturally derived CTS exhibits promising efficacy as a treatment drug for AAA through its targeting of the Keap1-Nrf2-GSDMD-pyroptosis axis in VSMCs.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Angiotensin II/metabolism
- Angiotensin II/pharmacology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/pathology
- Disease Models, Animal
- Kelch-Like ECH-Associated Protein 1/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- NF-E2-Related Factor 2/metabolism
- Phenanthrenes/pharmacology
- Pyroptosis/drug effects
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Jiaojiao Wang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Weile Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Jiami Zou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Pinglian Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Mei Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Zhihua Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Chunhong Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Wanlu Qiu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Jing Lu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chengzhi Li
- Department of Interventional Radiology and Vascular Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Shuai Guo
- School of Basic Medical Sciences, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yiming Xu
- School of Basic Medical Sciences, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zunnan Huang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Peiqing Liu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| |
Collapse
|
38
|
Wang W, Liu Q, Yang Q, Fu S, Zheng D, Su Y, Xu J, Wang Y, Piao H, Liu K. 3D-printing hydrogel programmed released exosomes to restore aortic medial degeneration through inhibiting VSMC ferroptosis in aortic dissection. J Nanobiotechnology 2024; 22:600. [PMID: 39367412 PMCID: PMC11453022 DOI: 10.1186/s12951-024-02821-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/30/2024] [Indexed: 10/06/2024] Open
Abstract
Aortic dissection (AD) is a devastating disease with a high mortality rate. Exosomes derived from mesenchymal stem cells (exo-MSCs) offer a promising strategy to restore aortic medial degeneration and combat ferroptosis in AD. However, their rapid degradation in the circulatory system and low treatment efficiency limit their clinical application. Methylacrylated gelatin (Gelma) was reported as a matrix material to achieve controlled release of exosomes. Herein, exo-MSCs-embedded in Gelma hydrogels (Gelma-exos) using ultraviolet light and three-dimensional (3D) printing technology. These Gelma-exos provide a sustained release of exo-MSCs as Gelma gradually degrades, helping to restore aortic medial degeneration and prevent ferroptosis. The sustained release of exosomes can inhibit the phenotypic switch of vascular smooth muscle cells (VSMCs) to a proliferative state, and curb their proliferation and migration. Additionally, the 3D-printed Gelma-exos demonstrated the ability to inhibit ferroptosis in vitro, in vivo and ex vivo experiments. In conclusion, our Gelma-exos, combined with 3D-printed technology, offer an alternative treatment approach for repairing aortic medial degeneration and ferroptosis in AD, potentially reducing the incidence of aortic dissection rupture.
Collapse
Affiliation(s)
- Weitie Wang
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Yatai Street 4026, Changchun, 130041, Jilin, China
| | - Qing Liu
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiwei Yang
- China Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Songning Fu
- The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dongdong Zheng
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Yatai Street 4026, Changchun, 130041, Jilin, China
| | - Yale Su
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Yatai Street 4026, Changchun, 130041, Jilin, China
| | - Jinyu Xu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Yatai Street 4026, Changchun, 130041, Jilin, China
| | - Yong Wang
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Yatai Street 4026, Changchun, 130041, Jilin, China
| | - Hulin Piao
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Yatai Street 4026, Changchun, 130041, Jilin, China
| | - Kexiang Liu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Yatai Street 4026, Changchun, 130041, Jilin, China.
| |
Collapse
|
39
|
Li D, Wang L, Jiang B, Miao Y, Li X. An evidence update to explore molecular targets and protective mechanisms of apigenin against abdominal aortic aneurysms based on network pharmacology and experimental validation. Mol Divers 2024; 28:2913-2929. [PMID: 37653360 DOI: 10.1007/s11030-023-10723-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Abdominal aortic aneurysms (AAA) is a life-threatening disease and the incidence of AAA is still on the rise in recent years. Numerous studies suggest that dietary moderate consumption of polyphenol exerts beneficial effects on cardiovascular disease. Apigenin (API) is a promising dietary polyphenol and possesses potent beneficial effects on our body. Although our previous study revealed protective effects of API on experimental AAA formation, up till now few studies were carried out to further investigate its involved molecular mechanisms. In the present study, network pharmacology combined molecular docking and experimental validation was used to explore API-related therapeutic targets and mechanisms in the treatment of AAA. Firstly, we collected 202 API-related therapeutic targets and 2475 AAA-related pathogenetic targets. After removing duplicates, a total of 68 potential therapeutic targets were obtained. Moreover, 5 targets with high degree including TNF, ACTB, INS, JUN, and MMP9 were identified as core targets of API for treating AAA. In addition, functional enrichment analysis indicated that API exerted pharmacological effects in AAA by affecting versatile mechanisms, including apoptosis, inflammation, blood fluid dynamics, and immune modulation. Molecular docking results further supported that API had strong affinity with the above core targets. Furthermore, protein level of core targets and related pathways were evaluated in a Cacl2-induced AAA model by using western blot and immunohistochemistry. The experimental validation results demonstrated that API significantly attenuated phosphorylation of JUN and protein level of predicted core targets. Taken together, based on network pharmacological and experimental validation, our study systematically explored associated core targets and potential therapeutic pathways of API for AAA treatment, which could supply valuable insights and theoretical basis for AAA treatment.
Collapse
Affiliation(s)
- Dongyu Li
- Department of General Surgery & VIP In-Patient Ward, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Lei Wang
- Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Nanjingbei 155 Street, Shenyang, 110001, Liaoning Province, China
| | - Bo Jiang
- Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Nanjingbei 155 Street, Shenyang, 110001, Liaoning Province, China
| | - Yuxi Miao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China
| | - Xuan Li
- Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Nanjingbei 155 Street, Shenyang, 110001, Liaoning Province, China.
| |
Collapse
|
40
|
El-Nashar H, Sabry M, Tseng YT, Francis N, Latif N, Parker KH, Moore JE, Yacoub MH. Multiscale structure and function of the aortic valve apparatus. Physiol Rev 2024; 104:1487-1532. [PMID: 37732828 PMCID: PMC11495199 DOI: 10.1152/physrev.00038.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/22/2023] Open
Abstract
Whereas studying the aortic valve in isolation has facilitated the development of life-saving procedures and technologies, the dynamic interplay of the aortic valve and its surrounding structures is vital to preserving their function across the wide range of conditions encountered in an active lifestyle. Our view is that these structures should be viewed as an integrated functional unit, here referred to as the aortic valve apparatus (AVA). The coupling of the aortic valve and root, left ventricular outflow tract, and blood circulation is crucial for AVA's functions: unidirectional flow out of the left ventricle, coronary perfusion, reservoir function, and support of left ventricular function. In this review, we explore the multiscale biological and physical phenomena that underlie the simultaneous fulfillment of these functions. A brief overview of the tools used to investigate the AVA, such as medical imaging modalities, experimental methods, and computational modeling, specifically fluid-structure interaction (FSI) simulations, is included. Some pathologies affecting the AVA are explored, and insights are provided on treatments and interventions that aim to maintain quality of life. The concepts explained in this article support the idea of AVA being an integrated functional unit and help identify unanswered research questions. Incorporating phenomena through the molecular, micro, meso, and whole tissue scales is crucial for understanding the sophisticated normal functions and diseases of the AVA.
Collapse
Affiliation(s)
- Hussam El-Nashar
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Malak Sabry
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Department of Biomedical Engineering, King's College London, London, United Kingdom
| | - Yuan-Tsan Tseng
- Heart Science Centre, Magdi Yacoub Institute, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nadine Francis
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Najma Latif
- Heart Science Centre, Magdi Yacoub Institute, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kim H Parker
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - James E Moore
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Magdi H Yacoub
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Heart Science Centre, Magdi Yacoub Institute, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
41
|
Rombouts KB, van Merrienboer TAR, Henneman AA, Knol JC, Pham TV, Piersma SR, Jimenez CR, Bogunovic N, van der Velden J, Yeung KK. Insight in the (Phospho)proteome of Vascular Smooth Muscle Cells Derived From Patients With Abdominal Aortic Aneurysm Reveals Novel Disease Mechanisms. Arterioscler Thromb Vasc Biol 2024; 44:2226-2243. [PMID: 39206541 DOI: 10.1161/atvbaha.124.321087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is characterized by weakening and dilatation of the aortic wall in the abdomen. The aim of this study was to gain insight into cell-specific mechanisms involved in AAA pathophysiology by analyzing the (phospho)proteome of vascular smooth muscle cells derived from patients with AAA compared with those of healthy donors. METHODS A (phospho)proteomics analysis based on tandem mass spectrometry was performed on vascular smooth muscle cells derived from patients with AAA (n=24) and healthy, control individuals (C-SMC, n=8). Following protein identification and quantification using MaxQuant, integrative inferred kinase activity analysis was used to calculate kinase activity scores. RESULTS Expression differences between vascular smooth muscle cells derived from patients with AAA and healthy, control individuals were predominantly found in proteins involved in ECM (extracellular matrix) remodeling (THSD4 [thrombospondin type-1 domain-containing protein 4] and ADAMTS1 [A disintegrin and metalloproteinase with thrombospondin motifs 1]), energy metabolism (GYS1 [glycogen synthase 1] and PCK2 [phosphoenolpyruvate carboxykinase 2, mitochondrial]), and contractility (CACNA2D1 [calcium voltage-dependent channel subunit α-2/δ-1] and TPM1 [tropomyosin α-1 chain]). Phosphorylation patterns on proteins related to actin cytoskeleton organization dominated the phosphoproteome of vascular smooth muscle cells derived from patients with AAA . Besides, phosphorylation changes on proteins related to energy metabolism (GYS1), contractility (PARVA [α-parvin], PPP1R12A [protein phosphatase 1 regulatory subunit 12A], and CALD1 [caldesmon 1]), and intracellular communication (GJA1 [gap junction α-1 protein]) were seen. Kinase activity of NUAK1 (NUAK family SNF1-like kinase 1), FYN (tyrosine-protein kinase Fyn), MAPK7 (mitogen-activated protein kinase 7), and STK10 (serine/threonine kinase 10) was different in vascular smooth muscle cells derived from patients with AAA compared with those from healthy, control individuals. CONCLUSIONS This study revealed changes in expression and phosphorylation levels of proteins involved in various processes responsible for AAA progression and development (eg, energy metabolism, ECM remodeling, actin cytoskeleton organization, contractility, intracellular communication, and cell adhesion). These newly identified proteins, phosphosites, and related kinases provide further insight into the underlying mechanism of vascular smooth muscle cell dysfunction within the aneurysmal wall. Our omics data thereby offer the opportunity to study the relevance, either as drug target or biomarker, of these proteins in AAA development.
Collapse
MESH Headings
- Humans
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Proteomics/methods
- Male
- Aged
- Cells, Cultured
- Phosphorylation
- Case-Control Studies
- Proteome
- Female
- Vascular Remodeling
- Middle Aged
- Phosphoproteins/metabolism
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Energy Metabolism
- Tandem Mass Spectrometry
- Signal Transduction
Collapse
Affiliation(s)
- Karlijn B Rombouts
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location Vrije Universiteit (VU) Medical Center and Academic Medical Centre (AMC), the Netherlands (K.B.R., T.A.R.v.M., N.B., K.K.Y.)
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, the Netherlands (K.B.R., T.A.R.v.M., N.B., J.v.d.V., K.K.Y.)
| | - Tara A R van Merrienboer
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location Vrije Universiteit (VU) Medical Center and Academic Medical Centre (AMC), the Netherlands (K.B.R., T.A.R.v.M., N.B., K.K.Y.)
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, the Netherlands (K.B.R., T.A.R.v.M., N.B., J.v.d.V., K.K.Y.)
| | - Alex A Henneman
- Department of Laboratory Medical Oncology, OncoProteomics Laboratory, Amsterdam University Medical Centers, Location VU Medical Center, Cancer Center Amsterdam, the Netherlands (A.A.H., J.C.K., T.V.P., S.R.P., C.R.J.)
| | - Jaco C Knol
- Department of Laboratory Medical Oncology, OncoProteomics Laboratory, Amsterdam University Medical Centers, Location VU Medical Center, Cancer Center Amsterdam, the Netherlands (A.A.H., J.C.K., T.V.P., S.R.P., C.R.J.)
| | - Thang V Pham
- Department of Laboratory Medical Oncology, OncoProteomics Laboratory, Amsterdam University Medical Centers, Location VU Medical Center, Cancer Center Amsterdam, the Netherlands (A.A.H., J.C.K., T.V.P., S.R.P., C.R.J.)
| | - Sander R Piersma
- Department of Laboratory Medical Oncology, OncoProteomics Laboratory, Amsterdam University Medical Centers, Location VU Medical Center, Cancer Center Amsterdam, the Netherlands (A.A.H., J.C.K., T.V.P., S.R.P., C.R.J.)
| | - Connie R Jimenez
- Department of Laboratory Medical Oncology, OncoProteomics Laboratory, Amsterdam University Medical Centers, Location VU Medical Center, Cancer Center Amsterdam, the Netherlands (A.A.H., J.C.K., T.V.P., S.R.P., C.R.J.)
| | - Natalija Bogunovic
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location Vrije Universiteit (VU) Medical Center and Academic Medical Centre (AMC), the Netherlands (K.B.R., T.A.R.v.M., N.B., K.K.Y.)
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, the Netherlands (K.B.R., T.A.R.v.M., N.B., J.v.d.V., K.K.Y.)
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, the Netherlands (K.B.R., T.A.R.v.M., N.B., J.v.d.V., K.K.Y.)
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location Vrije Universiteit (VU) Medical Center and Academic Medical Centre (AMC), the Netherlands (K.B.R., T.A.R.v.M., N.B., K.K.Y.)
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, the Netherlands (K.B.R., T.A.R.v.M., N.B., J.v.d.V., K.K.Y.)
| |
Collapse
|
42
|
Giudici A, Szafron JM, Ramachandra AB, Spronck B. Instability in Computational Models of Vascular Smooth Muscle Cell Contraction. Ann Biomed Eng 2024; 52:2403-2416. [PMID: 38949730 PMCID: PMC11329416 DOI: 10.1007/s10439-024-03532-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/30/2024] [Indexed: 07/02/2024]
Abstract
PURPOSE Through their contractile and synthetic capacity, vascular smooth muscle cells (VSMCs) can regulate the stiffness and resistance of the circulation. To model the contraction of blood vessels, an active stress component can be added to the (passive) Cauchy stress tensor. Different constitutive formulations have been proposed to describe this active stress component. Notably, however, measuring biomechanical behaviour of contracted blood vessels ex vivo presents several experimental challenges, which complicate the acquisition of comprehensive datasets to inform complex active stress models. In this work, we examine formulations for use with limited experimental contraction data as well as those developed to capture more comprehensive datasets. METHODS First, we prove analytically that a subset of constitutive active stress formulations exhibits unstable behaviours (i.e., a non-unique diameter solution for a given pressure) in certain parameter ranges, particularly for large contractile deformations. Second, using experimental literature data, we present two case studies where these formulations are used to capture the contractile response of VSMCs in the presence of (1) limited and (2) extensive contraction data. RESULTS We show how limited contraction data complicates selecting an appropriate active stress model for vascular applications, potentially resulting in unrealistic modelled behaviours. CONCLUSION Our data provide a useful reference for selecting an active stress model which balances the trade-off between accuracy and available biomechanical information. Whilst complex physiologically motivated models' superior accuracy is recommended whenever active biomechanics can be extensively characterised experimentally, a constant 2nd Piola-Kirchhoff active stress model balances well accuracy and applicability with sparse contractile data.
Collapse
Affiliation(s)
- Alessandro Giudici
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 40, Room C5.578A, Maastricht, 6229 ER, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Jason M Szafron
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | | | - Bart Spronck
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 40, Room C5.578A, Maastricht, 6229 ER, The Netherlands.
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
43
|
He W, Zheng Q, Zou T, Yan W, Gao X, Wang C, Xiong Y. Angiopoietin-like 4 facilitates human aortic smooth muscle cell phenotype switch and dysfunctions through the PI3K/Akt signaling in aortic dissection. Adv Med Sci 2024; 69:474-483. [PMID: 39326736 DOI: 10.1016/j.advms.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/17/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
PURPOSE Vascular smooth muscle cell (VSMC) phenotype switch and dysfunctions have been reported to participate in aortic dissection (AD) progression. This study was aimed to investigate the role of angiopoietin-like 4 (ANGPTL4) in regulating VSMCs phenotype switch. MATERIALS AND METHODS Key genes were analyzed in AD using public datasets, and it was found that the central differential gene ANGPTL4 was up-regulated in AD. The KEGG signaling pathway annotation was performed to validate the associated pathways, and the expression of ANGPTL4 was verified using multiple datasets and clinical samples. Furthermore, the specific functions of ANGPTL4 on platelet-derived growth factor-BB (PDGF-BB)-treated human aortic smooth muscle cell (HASMC) phenotypes were investigated. The dynamic effects of ANGPTL4 and core signaling antagonists on HASMC phenotypes were examined. RESULTS Hub gene ANGPTL4 was significantly up-regulated in AD. ANGPTL4 was linked to the PI3K/Akt signaling, angiogenesis, and neovascularization and remodeling. ANGPTL4 overexpression further enhanced PDGF-BB effects on HASMC phenotypes, including promoted cell viability and migration, decreased contractile VSMC markers α-SMA and SM22α, elevated ECM degradation markers MMP-2 and MMP-9, and promoted phosphorylation of PI3K and Akt. ANGPTL4 knockdown partially abolished PDGF-BB-induced contractile/synthetic VSMCs imbalance and HASMC dysfunctions. Furthermore, in ANGPTL4-overexpressing HASMCs pre-treated with PDGF-BB, the PI3K/Akt signaling inhibitor LY294002 also partially eliminated the effects caused by the PDGF-BB treatment and ANGPTL4 overexpression. CONCLUSIONS ANGPTL4 is significantly up-regulated in AD. ANGPTL4 overexpression further enhanced PDGF-BB effects on HASMC phenotype switch and dysfunctions, which might be involved in the PI3K/Akt signaling.
Collapse
Affiliation(s)
- Wei He
- Department of Extracorporeal Life Support Center of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Quan Zheng
- Department of Extracorporeal Life Support Center of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tingfang Zou
- Department of Extracorporeal Life Support Center of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Yan
- Department of Extracorporeal Life Support Center of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xue Gao
- Department of Extracorporeal Life Support Center of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunle Wang
- Department of Extracorporeal Life Support Center of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yaoyao Xiong
- Department of Extracorporeal Life Support Center of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
44
|
Mo F, Wang C, Li S, Li Z, Xiao C, Zhang Y, Hu C, Wang E, Lin P, Yuan T, Zuo Z, Fu W, Chen X, Ren L, Wang L. A Dual-Targeting, Multi-Faceted Biocompatible Nanodrug Optimizes the Microenvironment to Ameliorate Abdominal Aortic Aneurysm. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405761. [PMID: 38923441 DOI: 10.1002/adma.202405761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/07/2024] [Indexed: 06/28/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a highly lethal cardiovascular disease that currently lacks effective pharmacological treatment given the complex pathophysiology of the disease. Here, single-cell RNA-sequencing data from patients with AAA and a mouse model are analyzed, which reveals pivotal pathological changes, including the M1-like polarization of macrophages and the loss of contractile function in smooth muscle cells (SMCs). Both cell types express the integrin αvβ3, allowing for their dual targeting with a single rationally designed molecule. To this end, a biocompatible nanodrug, which is termed EVMS@R-HNC, that consists of the multifunctional drug everolimus (EVMS) encapsulated by the hepatitis B virus core protein modifies to contain the RGD sequence to specifically bind to integrin αvβ3 is designed. Both in vitro and in vivo results show that EVMS@R-HNC can target macrophages as well as SMCs. Upon binding of the nanodrug, the EVMS is released intracellularly where it exhibits multiple functions, including inhibiting M1 macrophage polarization, thereby suppressing the self-propagating inflammatory cascade and immune microenvironment imbalance, while preserving the normal contractile function of SMCs. Collectively, these results suggest that EVMS@R-HNC presents a highly promising therapeutic approach for the management of AAA.
Collapse
Affiliation(s)
- Fandi Mo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Chufan Wang
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China
| | - Shiyi Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Zheyun Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Cheng Xiao
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China
| | - Yuchong Zhang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Chengkai Hu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Enci Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Peng Lin
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Tong Yuan
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Ziang Zuo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Lei Ren
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| |
Collapse
|
45
|
Li C, Liu L, Li S, Liu YS. N 6-Methyladenosine in Vascular Aging and Related Diseases: Clinical Perspectives. Aging Dis 2024; 15:1447-1473. [PMID: 37815911 PMCID: PMC11272212 DOI: 10.14336/ad.2023.0924-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023] Open
Abstract
Aging leads to progressive deterioration of the structure and function of arteries, which eventually contributes to the development of vascular aging-related diseases. N6-methyladenosine (m6A) is the most prevalent modification in eukaryotic RNAs. This reversible m6A RNA modification is dynamically regulated by writers, erasers, and readers, playing a critical role in various physiological and pathological conditions by affecting almost all stages of the RNA life cycle. Recent studies have highlighted the involvement of m6A in vascular aging and related diseases, shedding light on its potential clinical significance. In this paper, we comprehensively discuss the current understanding of m6A in vascular aging and its clinical implications. We discuss the molecular insights into m6A and its association with clinical realities, emphasizing its significance in unraveling the mechanisms underlying vascular aging. Furthermore, we explore the possibility of m6A and its regulators as clinical indicators for early diagnosis and prognosis prediction and investigate the therapeutic potential of m6A-associated anti-aging approaches. We also examine the challenges and future directions in this field and highlight the necessity of integrating m6A knowledge into patient-centered care. Finally, we emphasize the need for multidisciplinary collaboration to advance the field of m6A research and its clinical application.
Collapse
Affiliation(s)
- Chen Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - Le Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - Shuang Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| |
Collapse
|
46
|
Xu S, Han X, Wang X, Yu Y, Qu C, Liu X, Yang B. The role of oxidative stress in aortic dissection: a potential therapeutic target. Front Cardiovasc Med 2024; 11:1410477. [PMID: 39070552 PMCID: PMC11272543 DOI: 10.3389/fcvm.2024.1410477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
The incidence of aortic dissection (AD) is steadily increasing, driven by the rising prevalence of chronic conditions such as hypertension and the global aging of the population. Oxidative stress emerges as a pivotal pathophysiological mechanism contributing to the progression of AD. Oxidative stress triggers apoptosis in vascular smooth muscle cells, reshapes the extracellular matrix (ECM), and governs ECM degradation and remodeling, subsequently impacting aortic compliance. Furthermore, oxidative stress not only facilitates the infiltration of macrophages and mononuclear lymphocytes but also disrupts the integral structure and functionality of endothelial cells, thereby inducing endothelial cell dysfunction and furthering the degeneration of the middle layer of the aortic wall. Investigating antioxidants holds promise as a therapeutic avenue for addressing AD.
Collapse
Affiliation(s)
- Shengnan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Xiukun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Yi Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| |
Collapse
|
47
|
Wang Q, Lv H, Ainiwan M, Yesitayi G, Abudesimu A, Siti D, Aizitiaili A, Ma X. Untargeted metabolomics identifies indole-3-propionic acid to relieve Ang II-induced endothelial dysfunction in aortic dissection. Mol Cell Biochem 2024; 479:1767-1786. [PMID: 38485805 DOI: 10.1007/s11010-024-04961-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/07/2024] [Indexed: 07/18/2024]
Abstract
Indole-3-propionic acid (IPA), a gut microbiota-derived metabolite of tryptophan, has been proven to fulfill an essential function in cardiovascular disease (CVD) and nerve regeneration disease. However, the role of IPA in aortic dissection (AD) has not been revealed. We aimed to investigate the role of IPA in the pathogenesis of AD and the underlying mechanisms of IPA in endothelial dysfunction. Untargeted metabolomics has been employed to screen the plasma metabolic profile of AD patients in comparison with healthy individuals. Network pharmacology provides insights into the potential molecular mechanisms underlying IPA. 3-aminopropionitrile fumarate (BAPN) and angiotensin II (Ang II) were administered to induce AD in mice, while human umbilical vein endothelial cells (HUVECs) were employed for in vitro validation of the signaling pathways predicted by network pharmacology. A total of 224 potentially differential plasma metabolites were identified in the AD patients, with 110 up-regulated metabolites and 114 down-regulated metabolites. IPA was the most significantly decreased metabolite involved in tryptophan metabolism. Bcl2, caspase3, and AKT1 were predicted as the target genes of IPA by network pharmacology and molecular docking. IPA suppressed Ang II-induced apoptosis, intracellular ROS generation, inflammation, and endothelial tight junction (TJ) loss. Animal experiments demonstrated that administration of IPA alleviated the occurrence and severity of AD in mice. Taken together, we identified a previously unexplored association between tryptophan metabolite IPA and AD, providing a novel perspective on the underlying mechanism through which IPA mitigates endothelial dysfunction to protect against AD.
Collapse
Affiliation(s)
- Qi Wang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China
| | - Hui Lv
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China
| | - Mierxiati Ainiwan
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China
| | - Gulinazi Yesitayi
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China
| | - Asiya Abudesimu
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China
| | - Dilixiati Siti
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China
| | - Aliya Aizitiaili
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China
| | - Xiang Ma
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Ürümqi, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China.
| |
Collapse
|
48
|
Paredes F, Williams HC, Liu X, Holden C, Bogan B, Wang Y, Crotty KM, Yeligar SM, Elorza AA, Lin Z, Rezvan A, San Martin A. The mitochondrial protease ClpP is a druggable target that controls VSMC phenotype by a SIRT1-dependent mechanism. Redox Biol 2024; 73:103203. [PMID: 38823208 PMCID: PMC11169483 DOI: 10.1016/j.redox.2024.103203] [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: 02/19/2024] [Revised: 05/12/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024] Open
Abstract
Vascular smooth muscle cells (VSMCs), known for their remarkable lifelong phenotypic plasticity, play a pivotal role in vascular pathologies through their ability to transition between different phenotypes. Our group discovered that the deficiency of the mitochondrial protein Poldip2 induces VSMC differentiation both in vivo and in vitro. Further comprehensive biochemical investigations revealed Poldip2's specific interaction with the mitochondrial ATPase caseinolytic protease chaperone subunit X (CLPX), which is the regulatory subunit for the caseinolytic protease proteolytic subunit (ClpP) that forms part of the ClpXP complex - a proteasome-like protease evolutionarily conserved from bacteria to humans. This interaction limits the protease's activity, and reduced Poldip2 levels lead to ClpXP complex activation. This finding prompted the hypothesis that ClpXP complex activity within the mitochondria may regulate the VSMC phenotype. Employing gain-of-function and loss-of-function strategies, we demonstrated that ClpXP activity significantly influences the VSMC phenotype. Notably, both genetic and pharmacological activation of ClpXP inhibits VSMC plasticity and fosters a quiescent, differentiated, and anti-inflammatory VSMC phenotype. The pharmacological activation of ClpP using TIC10, currently in phase III clinical trials for cancer, successfully replicates this phenotype both in vitro and in vivo and markedly reduces aneurysm development in a mouse model of elastase-induced aortic aneurysms. Our mechanistic exploration indicates that ClpP activation regulates the VSMC phenotype by modifying the cellular NAD+/NADH ratio and activating Sirtuin 1. Our findings reveal the crucial role of mitochondrial proteostasis in the regulation of the VSMC phenotype and propose the ClpP protease as a novel, actionable target for manipulating the VSMC phenotype.
Collapse
Affiliation(s)
- Felipe Paredes
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Holly C Williams
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Xuesong Liu
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States; Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Claire Holden
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Bethany Bogan
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Yu Wang
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Kathryn M Crotty
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States; Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Samantha M Yeligar
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States; Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Alvaro A Elorza
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Zhiyong Lin
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Amir Rezvan
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Alejandra San Martin
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States; Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
| |
Collapse
|
49
|
Song Y, Deng M, Qiu Y, Cui Y, Zhang B, Xin J, Feng L, Mu X, Cui J, Li H, Sun Y, Yi W. Bergenin alleviates proliferative arterial diseases by modulating glucose metabolism in vascular smooth muscle cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155592. [PMID: 38608597 DOI: 10.1016/j.phymed.2024.155592] [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: 10/23/2023] [Revised: 02/06/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Vascular smooth muscle cell (VSMC) proliferation and phenotypic switching are key mechanisms in the development of proliferative arterial diseases. Notably, reprogramming of the glucose metabolism pattern in VSMCs plays an important role in this process. PURPOSE The aim of this study is to investigate the therapeutic potential and the mechanism underlying the effect of bergenin, an active compound found in Bergenia, in proliferative arterial diseases. METHODS The effect of bergenin on proliferative arterial disease was evaluated using platelet-derived growth factor (PDGF)-stimulated VSMCs and a mouse model of carotid artery ligation. VSMC proliferation and phenotypic switching were evaluated in vitro using cell counting kit-8, 5-ethynyl-2-deoxyuridine incorporation, scratch, and transwell assays. Carotid artery neointimal hyperplasia was evaluated in vivo using hematoxylin and eosin staining and immunofluorescence. The expression of proliferation and VSMC contractile phenotype markers was evaluated using PCR and western blotting. RESULTS Bergenin treatment inhibited PDGF-induced VSMC proliferation and phenotypic switching and reduced neointimal hyperplasia in the carotid artery ligation model. Additionally, bergenin partially reversed the PDGF-induced Warburg-like glucose metabolism pattern in VSMCs. RNA-sequencing data revealed that bergenin treatment significantly upregulated Ndufs2, an essential subunit of mitochondrial complex I. Ndufs2 knockdown attenuated the inhibitory effect of bergenin on PDGF-induced VSMC proliferation and phenotypic switching, and suppressed neointimal hyperplasia in vivo. Conversely, Ndufs2 overexpression enhanced the protective effect of bergenin. Moreover, Ndufs2 knockdown abrogated the effects of bergenin on the regulation of glucose metabolism in VSMCs. CONCLUSION These findings suggest that bergenin is effective in alleviating proliferative arterial diseases. The reversal of the Warburg-like glucose metabolism pattern in VSMCs during proliferation and phenotypic switching may underlie this therapeutic mechanism.
Collapse
Affiliation(s)
- Yujie Song
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China
| | - Meng Deng
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China
| | - Yufeng Qiu
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China
| | - Yang Cui
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China
| | - Bing Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China
| | - Jialin Xin
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China
| | - Lele Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China
| | - Xingdou Mu
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China
| | - Jun Cui
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China
| | - Hong Li
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China
| | - Yang Sun
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China.
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710000, China,.
| |
Collapse
|
50
|
Huang X, Deng H. Construction of the circRNA-miRNA-mRNA axis based on ferroptosis-related gene AKR1C1 to explore the potential pathogenesis of abdominal aortic aneurysm. Medicine (Baltimore) 2024; 103:e38749. [PMID: 38941402 PMCID: PMC11466172 DOI: 10.1097/md.0000000000038749] [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: 01/10/2024] [Accepted: 06/07/2024] [Indexed: 06/30/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a cardiovascular disease that seriously threatens human health and brings huge economic burden. At present, its pathogenesis remains unclear and its treatment is limited to surgical treatment. With the deepening and analysis of studies on the mechanism of ferroptosis, a new idea has been provided for the clinical management of AAA patients, including diagnosis, treatment and prevention. Therefore, this paper aims to construct a competitive endogenous RNA (ceRNA) regulatory axis based on ferroptosis to preliminarily explore the pathogenesis and potential therapeutic targets of AAA. We obtained upregulated and downregulated ferroptosis-related DEGs (FRGs) from GSE144431 dataset and 60 known ferroptosis-related genes. Pearson correlation analysis was used to find aldoketone reductase 1C (AKR1C1) in AAA samples. Enrichment analysis of these genes was performed via Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Correlation test between immune cells and AKR1C1 was investigated through single-sample gene set enrichment analysis (ssGSEA). The AKR1C1-miRNA pairs were predicted by the TargetScan database and miRWalk database. Circular RNA (CircRNA)-miRNA pairs were selected by the CircInteractome database. Overlapping miRNA between circRNA-miRNA and AKR1C1-miRNA pairs was visualized by Venn diagram. Finally, the circRNA-miRNA-mRNA axis was constructed by searching for upstream circRNA and downstream mRNA of overlapping miRNA. Only one downregulated AKR1C1 gene was found in GSE144431 and 60 ferroptosis-related genes. Functional Enrichment and Pathway Analysis of AKR1C1-related genes were further explored, and it was observed that they were mainly enriched in "response to oxidative stress," "glutathione biosynthetic process" and "nonribosomal peptide biosynthetic process," "Ferroptosis," "Glutathione metabolism" and "Chemical carcinogenesis-reactive oxygen species." They were also found to be significantly associated with most immune cells, including Activated Dendritic cells, CD56dim Natural killer cells, Gamma Delta T cells, Immature B cells, Plasmacytoid dendritic cell, Type 2 T helper cell, Activated CD4 T cell and Type 1 T helper cell. Has_circ_0005073-miRNA-543 and AKR1C1-miRNA-543 were identified by Online Database analysis. Therefore, we have established the has_circ_0005073/miRNA-543/AKR1C1 axis in AAA. We found AKR1C1 was differentially expressed between normal and AAA groups. Based on AKR1C1, we constructed the has_circ_0005073/miRNA-543/AKR1C1 axis to analyze AAA.
Collapse
Affiliation(s)
- Xuehua Huang
- Department of Neonatology, the First Hospital of China Medical University, Shenyang, China
| | - Huanhuan Deng
- Department of Nephrology, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|