1
|
Lao XY, Sun YL, Zhao ZJ, Liu J, Ruan XF. Pharmacological effects of betulinic acid and its protective mechanisms on the cardiovascular system. Fitoterapia 2025; 183:106561. [PMID: 40288588 DOI: 10.1016/j.fitote.2025.106561] [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: 02/11/2025] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Betulinic acid (BA), a pentacyclic triterpenoid saponin widely found in plants, has attracted attention for its diverse pharmacological activities. Recent studies highlight its cardioprotective potential, promoting its relevance in cardiovascular research. AIM OF THE REVIEW This review summarizes BA's physicochemical properties, structure-activity relationships, natural sources, and synthesis strategies. It further discusses its pharmacokinetics and toxicity to evaluate its drug development potential, with emphasis on cardioprotective effects and related signaling pathways. METHODS Literature was collected from databases such as PubMed and Web of Science, focusing on studies addressing BA's chemical characteristics, biological activities, pharmacokinetics, and cardiovascular relevance. RESULTS BA exerts cardioprotective effects via multiple signaling pathways, including NRF2, NF-κB, MAPK, and NFAT. These contribute to its antioxidant, anti-inflammatory, anti-apoptotic, and anti-proliferative actions, as well as its enhancement of endothelial function through nitric oxide signaling. BA also reduces lipid accumulation. Combined with favorable physicochemical properties and synthetic accessibility, these findings support BA as a promising multifunctional lead compound in cardiovascular pharmacology. CONCLUSION BA shows strong potential as a cardioprotective natural compound. Although further research is needed to validate its clinical efficacy and safety, its multi-target actions and structural versatility provide a solid basis for development in cardiovascular drug discovery.
Collapse
Affiliation(s)
- Xu Yuan Lao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuan Long Sun
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Zhe Jun Zhao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jie Liu
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao Fen Ruan
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Wang SY, Chen YS, Jin BY, Bilal A. The cGAS-STING pathway in atherosclerosis. Front Cardiovasc Med 2025; 12:1550930. [PMID: 40351606 PMCID: PMC12062000 DOI: 10.3389/fcvm.2025.1550930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Atherosclerosis (AS), a chronic inflammatory disease, remains a leading contributor to cardiovascular morbidity and mortality. Recent studies highlight the critical role of the cGAS-STING pathway-a key innate immune signaling cascade-in driving AS progression. This pathway is activated by cytoplasmic DNA from damaged cells, thereby triggering inflammation and accelerating plaque formation. While risk factors such as aging, obesity, smoking, hypertension, and diabetes are known to exacerbate AS, emerging evidence suggests that these factors may also enhance cGAS-STING pathway, which amplifies inflammatory responses. Targeting this pathway offers a promising therapeutic strategy to reduce the burden of cardiovascular diseases (CVD). In this review, we summarize the mechanisms of the cGAS-STING pathway, explore its role in AS, and evaluate potential inhibitors as future therapeutic candidates. By integrating current knowledge, we aim to provide insights for developing novel treatments to mitigate AS and CVD burden.
Collapse
Affiliation(s)
- Si-yu Wang
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Yu-shan Chen
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Bo-yuan Jin
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Ahmad Bilal
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
3
|
Alibrandi L, Lionetti V. Interspecies differences in mitochondria: Implications for cardiac and vascular translational research. Vascul Pharmacol 2025; 159:107476. [PMID: 40037508 DOI: 10.1016/j.vph.2025.107476] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/09/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Mitochondria are essential organelles that regulate cellular energy metabolism, redox balance, and signaling pathways related to proliferation, aging and survival. So far, significant interspecies differences exist in mitochondrial structure, function, and dynamics, which have critical implications for cardiovascular physiology and pharmacology. This review explores the main differences in mitochondrial properties across species of animals that are commonly used for translational research, emphasizing their cardiac and vascular relevance. By addressing key interspecies differences, including mitochondrial DNA (mtDNA) variation, bioenergetic profile, oxidative stress response, epigenetic regulation, mitochondrial biogenesis, and adaptive mechanisms, we aim to provide insights into the challenges and opportunities in translating preclinical findings to clinical applications. Understanding these interspecies differences is essential for optimizing the design and interpretation of preclinical studies and for developing effective mitochondrial-targeted therapies.
Collapse
Affiliation(s)
- Lisa Alibrandi
- TrancriLab, Laboratory of Basic and Applied Medical Research, Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Pisa, Italy
| | - Vincenzo Lionetti
- TrancriLab, Laboratory of Basic and Applied Medical Research, Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Pisa, Italy; UOSVD Anesthesia and Intensive Care, Fondazione Toscana G. Monasterio, Pisa, Italy.
| |
Collapse
|
4
|
Guan C, Chen SX, Huang CL, Du YP, Wang KH, Li PX, Liu SR, Liu ZY, Huang Z. Utilizing Integrated Bioinformatics Analysis to Explore Potential Alterations in Mitochondrial Function Within Immune Cells Associated with Thoracic Aortic Aneurysms. Bioengineering (Basel) 2025; 12:197. [PMID: 40001716 PMCID: PMC11852063 DOI: 10.3390/bioengineering12020197] [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: 10/28/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Thoracic aortic aneurysm (TAA) is a life-threatening peripheral vascular disease with a complex pathogenesis. Altered mitochondrial function in vascular smooth muscle cells has been implicated in TAA development. However, the link between mitochondrial functional status and immune cell behavior in TAA patients remains largely unexplored. In this study, we analyzed several bulk RNA-seq and snRNA-seq datasets of TAA from the NCBI-GEO and Genome Sequence Archive database, identifying differentially expressed mitochondrial-related genes (DE-MRGs). To assess mitochondrial function, we calculated a mitoscore to represent the overall expression level of MRGs. Our analysis revealed mitochondrial-mediated apoptosis occurring in M1 macrophages, while CD4 + T cells demonstrated the activation of quality control mechanisms, such as mitochondrial fission. Through LASSO regression and SVM-RFE, we identified key MRGs, including MUCB, ARRB2, FRG, and ALPL, which we further validated using TAA mouse models. Additionally, we found that DE-MRGs were closely linked to methionine metabolism. In conclusion, this study highlights mitochondrial dysfunction in immune cells associated with TAA, shedding light on potential mitochondrial roles in TAA pathogenesis.
Collapse
Affiliation(s)
- Chang Guan
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| | - Si-Xu Chen
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; (S.-X.C.); (C.-L.H.)
| | - Chun-Ling Huang
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; (S.-X.C.); (C.-L.H.)
| | - Yi-Peng Du
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| | - Kai-Hao Wang
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| | - Pei-Xin Li
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| | - Shen-Rong Liu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| | - Zhao-Yu Liu
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; (S.-X.C.); (C.-L.H.)
| | - Zheng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| |
Collapse
|
5
|
Liu Z, Huang N, Liu C, Wu C, Zhou L, Liu X, Lei H. Mitochondrial DNA in atherosclerosis research progress: a mini review. Front Immunol 2025; 16:1526390. [PMID: 39991161 PMCID: PMC11842404 DOI: 10.3389/fimmu.2025.1526390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/23/2025] [Indexed: 02/25/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease that primarily affects large and medium-sized arteries and is one of the leading causes of death worldwide. This article reviews the multifaceted role of mitochondrial DNA (mtDNA) in AS, including its structure, function, release, and relationship with inflammation. Damage and release of mtDNA are considered central drivers in the development of AS, as they participate in the progression of AS by activating inflammatory pathways and affecting lipid metabolism. Therefore, therapeutic strategies targeting mtDNA and its downstream effects may provide new avenues to address this global health challenge.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Nan Huang
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Chan Liu
- Department of Clinical Pharmacy, Liuyang People’s Hospital, Liuyang, China
| | - Can Wu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Ling Zhou
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Haibo Lei
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| |
Collapse
|
6
|
Liu Y, Yang Z, Na J, Chen X, Wang Z, Zheng L, Fan Y. In vitro stretch modulates mitochondrial dynamics and energy metabolism to induce smooth muscle differentiation in mesenchymal stem cells. FASEB J 2025; 39:e70354. [PMID: 39840656 DOI: 10.1096/fj.202402944r] [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: 11/23/2024] [Revised: 12/30/2024] [Accepted: 01/15/2025] [Indexed: 01/23/2025]
Abstract
The smooth muscle cells (SMCs) located in the vascular media layer are continuously subjected to cyclic stretching perpendicular to the vessel wall and play a crucial role in vascular wall remodeling and blood pressure regulation. Mesenchymal stem cells (MSCs) are promising tools to differentiate into SMCs. Mechanical stretch loading offers an opportunity to guide the MSC-SMC differentiation and mechanical adaption for function regeneration of blood vessels. This study shows that cyclic stretch induces the expression of SMC markers α-SMA and SM22 in MSCs. These cells exhibit contractile ability in vitro and facilitate angiogenesis in the Matrigel plug assay in vivo. The contraction of SMCs requires remodeling of their energy metabolism. However, the underlying mechanism in the differentiation of MSCs into SMCs remains to be revealed. Cyclic stretch training promotes glycolysis, oxidative phosphorylation, and mitochondrial fusion and modulates mitochondrial dynamics-related proteins (MFN1, MFN2, DRP1) expression, thereby contributing to MSCs differentiation. Yes-associated protein (YAP) affects mitochondrial dynamics, oxidative phosphorylation, and glycolysis to regulate stretch-mediated differentiation into SMCs. Additionally, Piezo-type mechanosensitive ion channel component 1 (Piezo1) impacts energy metabolism and MSCs differentiation by regulating intracellular Ca2+ levels and YAP nuclear localization. It indicates that YAP can integrate stretch force and energy metabolism signals to regulate the differentiation of MSCs into SMCs.
Collapse
Affiliation(s)
- Yu Liu
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Zhijie Yang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jing Na
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xinyuan Chen
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Ziyi Wang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Lisha Zheng
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
7
|
Lu X, Wang Y, Geng N, Zou Z, Feng X, Wang Y, Xu Z, Zhang N, Pu J. Dysregulated Mitochondrial Calcium Causes Spiral Artery Remodeling Failure in Preeclampsia. Hypertension 2024; 81:2368-2382. [PMID: 39291377 DOI: 10.1161/hypertensionaha.124.23046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Calcium deficiency in women is strongly linked to an increased risk of developing preeclampsia. Mitochondrial calcium ([Ca2+]m) homeostasis is essential to regulate vascular smooth muscle cell (VSMC) function. However, the role of [Ca2+]m in preeclampsia development remains largely unknown. METHODS To investigate this, human spiral arteries obtained from normotensive and preeclamptic women were collected for vascular function, RNA sequencing, and VSMC studies. N(ω)-nitro-L-arginine methyl ester-induced preeclampsia animal experiments were established to investigate the effects of intervening in [Ca2+]m to improve the outcome for preeclamptic mothers or their infants. RESULTS Our initial findings revealed compromised vessel function in spiral arteries derived from patients with preeclampsia, as evidenced by diminished vasoconstriction and vasodilation responses to angiotensin II and sodium nitroprusside, respectively. Moreover, the spiral artery VSMCs from patients with preeclampsia exhibited phenotypic transformation and proliferation associated with the disrupted regulatory mechanisms of [Ca2+]m uptake. Subsequent in vitro experiments employing gain- and loss-of-function approaches demonstrated that the mitochondrial Na+/Ca2+ exchanger played a role in promoting phenotypic switching and impaired mitochondrial functions in VSMCs. Furthermore, mtNCLX (mitochondrial Na+/Ca2+ exchanger) inhibitor CGP37157 significantly improved VSMC phenotypic changes and restored mitochondrial function in both patients with preeclampsia-derived VSMCs and the preeclampsia rat model. CONCLUSIONS This study provides comprehensive evidence supporting the disrupted regulatory mechanisms of [Ca2+]m uptake in VSMCs of spiral arteries of patients with preeclampsia and further elucidates its correlation with VSMC phenotypic switching and defective spiral artery remodeling. The findings suggest that targeting mtNCLX holds promise as a novel therapeutic approach for managing preeclampsia.
Collapse
Affiliation(s)
- Xiyuan Lu
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yifan Wang
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Na Geng
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Zhiguo Zou
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Xueqing Feng
- Department of Obstetrics Affiliated Hospital of Jining Medical University, China (X.F.)
| | - Yuehong Wang
- State Key Laboratory of Systems Medicine for Cancer (Yuehong Wang), School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, China
| | - Zhice Xu
- Wuxi Maternity and Child Health Care Hospital, China (Z.X.)
| | - Ning Zhang
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology (N.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital State Key Laboratory of Systems Medicine for Cancer (J.P.), School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, China
| |
Collapse
|
8
|
Zhang L, Chen Y, Pan Q, Fang S, Zhang Z, Wang J, Yang Y, Yang D, Sun X. Silencing of PCK1 mitigates the proliferation and migration of vascular smooth muscle cells and vascular intimal hyperplasia by suppressing STAT3/DRP1-mediated mitochondrial fission. Acta Biochim Biophys Sin (Shanghai) 2024; 57:633-645. [PMID: 39262325 PMCID: PMC12040600 DOI: 10.3724/abbs.2024154] [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: 05/20/2024] [Accepted: 07/30/2024] [Indexed: 09/13/2024] Open
Abstract
The pathological proliferation and migration of vascular smooth muscle cells (VSMCs) are key processes during vascular neointimal hyperplasia (NIH) and restenosis. Phosphoenolpyruvate carboxy kinase 1 (PCK1) is closely related to a variety of malignant proliferative diseases. However, the role of PCK1 in VSMCs has rarely been investigated. This study aims to examine the role of PCK1 in the proliferation and migration of VSMCs and vascular NIH after injury. In vivo, extensive NIH and increased expression of PCK1 within the neointima are observed in injured arteries. Interestingly, the administration of adeno-associated virus-9 (AAV-9) carrying Pck1 short hairpin RNA (sh Pck1) significantly attenuates NIH and stenosis of the vascular lumen. In vitro, Pck1 small interfering RNA (si Pck1)-induced PCK1 silencing inhibits VSMC proliferation and migration. Additionally, silencing of PCK1 leads to reduced expression of dynamin-related protein 1 (DRP1) and attenuated mitochondrial fission. Lentivirus-mediated DRP1 overexpression markedly reverses the inhibitory effects of PCK1 silencing on VSMC proliferation, migration, and mitochondrial fission. Finally, PCK1 inhibition attenuates the phosphorylation of signal transducer and activator of transcription 3 (STAT3). Activation of STAT3 abolishes the suppressive effects of PCK1 silencing on DRP1 expression, mitochondrial fission, proliferation, and migration in VSMCs. In conclusion, PCK1 inhibition attenuates the mitochondrial fission, proliferation, and migration of VSMCs by inhibiting the STAT3/DRP1 axis, thereby suppressing vascular NIH and restenosis.
Collapse
MESH Headings
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/genetics
- Cell Proliferation/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Animals
- Cell Movement/genetics
- Mitochondrial Dynamics/genetics
- Dynamins/metabolism
- Dynamins/genetics
- Hyperplasia/metabolism
- Hyperplasia/genetics
- Hyperplasia/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Male
- Neointima/pathology
- Neointima/genetics
- Neointima/metabolism
- Gene Silencing
- Rats
- Rats, Sprague-Dawley
- Cells, Cultured
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Li Zhang
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Yingmei Chen
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Quanrong Pan
- Department of General Practicethe General Hospital of Western Theater CommandChengdu610083China
| | - Shizheng Fang
- Department of Critical Care Medicinethe General Hospital of Western Theater CommandChengdu610083China
| | - Zhongjian Zhang
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Jia Wang
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Yongjian Yang
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Dachun Yang
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| | - Xiongshan Sun
- Department of Cardiologythe General Hospital of Western Theater CommandChengdu610083China
| |
Collapse
|
9
|
Sun J, Shao Y, Pei L, Zhu Q, Yu X, Yao W. AKAP1 alleviates VSMC phenotypic modulation and neointima formation by inhibiting Drp1-dependent mitochondrial fission. Biomed Pharmacother 2024; 176:116858. [PMID: 38850669 DOI: 10.1016/j.biopha.2024.116858] [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: 12/18/2023] [Revised: 05/26/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
The roles and mechanisms of A-kinase anchoring protein 1 (AKAP1) in vascular smooth muscle cell (VSMC) phenotypic modulation and neointima formation are currently unknown. AKAP1 is a mitochondrial PKA-anchored protein and maintains mitochondrial homeostasis. This study aimed to investigate how AKAP1/PKA signaling plays a protective role in inhibiting VSMC phenotypic transformation and neointima formation by regulating mitochondrial fission. The results showed that both PDGF-BB treatment and balloon injury reduced the transcription, expression, and mitochondrial anchoring of AKAP1. In vitro, the overexpression of AKAP1 significantly inhibited PDGF-BB mediated VSMC proliferation and migration, whereas AKAP1 knockdown further aggravated VSMC phenotypic transformation. Additionally, in the balloon injury model in vivo, AKAP1 overexpression reduced neointima formation, the muscle fiber area ratio, and rat VSMC proliferation and migration. Furthermore, PDGF-BB and balloon injury inhibited Drp1 phosphorylation at Ser637 and promoted Drp1 activity and mitochondrial midzone fission; AKAP1 overexpression reversed these effects. AKAP1 overexpression also inhibited the distribution of mitochondria at the plasma membrane and the reduction of PKARIIβ expression induced by PDGF-BB, as evidenced by an increase in mitochondria-plasma membrane distance as well as PKARIIβ protein levels. Moreover, the PKA agonist promoted Drp1 phosphorylation (Ser637) and inhibited PDGF-BB-mediated mitochondrial fission, cell proliferation, and migration. The PKA antagonist reversed the increase in Drp1 phosphorylation (Ser637) and the decline in mitochondrial midzone fission and VSMC phenotypic transformation caused by AKAP1 overexpression. The results of this study reveal that AKAP1 protects VSMCs against phenotypic modulation by improving Drp1 phosphorylation at Ser637 through PKA and inhibiting mitochondrial fission, thereby preventing neointima formation.
Collapse
MESH Headings
- Animals
- Male
- Rats
- A Kinase Anchor Proteins/metabolism
- A Kinase Anchor Proteins/genetics
- Becaplermin/pharmacology
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Dynamins/metabolism
- Mitochondrial Dynamics/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Neointima/metabolism
- Neointima/pathology
- Phenotype
- Phosphorylation
- Rats, Sprague-Dawley
- Signal Transduction
Collapse
Affiliation(s)
- Jingwen Sun
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Yuting Shao
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Lele Pei
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Qingyu Zhu
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Xiaoqiang Yu
- Department of Vascular Surgery, The First People's Hospital of Nantong, Nantong 226001, China
| | - Wenjuan Yao
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China.
| |
Collapse
|
10
|
Ren H, Hu W, Jiang T, Yao Q, Qi Y, Huang K. Mechanical stress induced mitochondrial dysfunction in cardiovascular diseases: Novel mechanisms and therapeutic targets. Biomed Pharmacother 2024; 174:116545. [PMID: 38603884 DOI: 10.1016/j.biopha.2024.116545] [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: 02/05/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality worldwide. Others and our studies have shown that mechanical stresses (forces) including shear stress and cyclic stretch, occur in various pathological conditions, play significant roles in the development and progression of CVDs. Mitochondria regulate the physiological processes of cardiac and vascular cells mainly through adenosine triphosphate (ATP) production, calcium flux and redox control while promote cell death through electron transport complex (ETC) related cellular stress response. Mounting evidence reveal that mechanical stress-induced mitochondrial dysfunction plays a vital role in the pathogenesis of many CVDs including heart failure and atherosclerosis. This review summarized mitochondrial functions in cardiovascular system under physiological mechanical stress and mitochondrial dysfunction under pathological mechanical stress in CVDs (graphical abstract). The study of mitochondrial dysfunction under mechanical stress can further our understanding of the underlying mechanisms, identify potential therapeutic targets, and aid the development of novel treatments of CVDs.
Collapse
Affiliation(s)
- He Ren
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Weiyi Hu
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China
| | - Tao Jiang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Qingping Yao
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China
| | - Yingxin Qi
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China
| | - Kai Huang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China.
| |
Collapse
|
11
|
Ping L, Zhi-Ming L, Bi-Shan Z, Lei Z, Bo Y, Yi-Chun Z, Ming-Jie W. S-propargyl-cysteine promotes the stability of atherosclerotic plaque via maintaining vascular muscle contractile phenotype. Front Cell Dev Biol 2024; 11:1291170. [PMID: 38328305 PMCID: PMC10847265 DOI: 10.3389/fcell.2023.1291170] [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: 09/08/2023] [Accepted: 12/22/2023] [Indexed: 02/09/2024] Open
Abstract
Introduction: Plaque rupture in atherosclerosis contributes to various acute cardiovascular events. As a new sulfide-containing donor, S-propargyl-cysteine (SPRC) has been reported to play a beneficial role in cardioprotection, potentially through its anti-inflammatory, anti-oxidative and anti-atherogenic activities. Our previous study observed an increase in eNOS phosphorylation in endothelial cells. However, it remains unclear whether SPRC influences vascular smooth muscle cells (VSMCs) within the plaque and if this effect contributes to plaque stabilization. Methods: An atherosclerotic unstable plaque mouse model was established by subjecting ApoE-/- mice to tandem stenosis of the right carotid artery along with a Western diet. Daily SPRC administration was conducted for 13 weeks. Plaque morphology and stability were assessed using MRI scanning and histopathological staining. In our in vitro studies, we stimulated human artery vascular smooth muscle cells (HAVSMCs) with platelet-derived growth factor-BB (PDGF-BB), both with and without 100 μM SPRC treatment. Cell phenotype was assessed using both Western blot and Real-time PCR. Cell proliferation was assessed using the BrdU cell proliferation kit and immunofluorescence of Ki-67, while cell migration was measured using scratch wound healing and transwell assay. MiR-143-3p overexpression and knockdown experiments were used to investigate whether it mediates the effect of SPRC on VSMC phenotype. Results and Discussion: SPRC treatment reduced plasma lipid levels, increased collagen content and decreased cell apoptosis in atherosclerotic plaques, indicating improved plaque stability. Both in vivo and in vitro studies elucidated the role of SPRC in preserving the contractile phenotype of VSMCs through up-regulation of miR-143-3p expression. Furthermore, SPRC suppressed the pro-proliferation and pro-migration effects of PDGF-BB on HAVSMCs. Overall, these findings suggest that the inhibitory effect of SPRC on phenotype switch from contractile to synthetic VSMCs may contribute to its beneficial role in enhancing plaque stability.
Collapse
Affiliation(s)
- Li Ping
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, The Innovative Research Team of High-level Local Universities in Shanghai, Fudan University, Shanghai, China
| | - Li Zhi-Ming
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, The Innovative Research Team of High-level Local Universities in Shanghai, Fudan University, Shanghai, China
| | - Zhang Bi-Shan
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, The Innovative Research Team of High-level Local Universities in Shanghai, Fudan University, Shanghai, China
| | - Zhu Lei
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Bo
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhu Yi-Chun
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, The Innovative Research Team of High-level Local Universities in Shanghai, Fudan University, Shanghai, China
| | - Wang Ming-Jie
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, The Innovative Research Team of High-level Local Universities in Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Mailloux RJ, Treberg J, Grayson C, Agellon LB, Sies H. Mitochondrial function and phenotype are defined by bioenergetics. Nat Metab 2023; 5:1641. [PMID: 37605058 DOI: 10.1038/s42255-023-00885-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Affiliation(s)
- Ryan J Mailloux
- The School of Human Nutrition, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada.
| | - Jason Treberg
- Department of Biological Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Cathryn Grayson
- The School of Human Nutrition, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Luis B Agellon
- The School of Human Nutrition, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Faculty of Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| |
Collapse
|