1
|
Zhong YL, Xu CQ, Li J, Liang ZQ, Wang MM, Ma C, Jia CL, Cao YB, Chen J. Mitochondrial dynamics and metabolism in macrophages for cardiovascular disease: A review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156620. [PMID: 40068296 DOI: 10.1016/j.phymed.2025.156620] [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: 12/16/2024] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Mitochondria regulate macrophage function, affecting cardiovascular diseases like atherosclerosis and heart failure. Their dynamics interact with macrophage cell death mechanisms, including apoptosis and necroptosis. PURPOSE This review explores how mitochondrial dynamics and metabolism influence macrophage inflammation and cell death in CVDs, highlighting therapeutic targets for enhancing macrophage resilience and reducing CVD pathology, while examining molecular pathways and pharmacological agents involved. STUDY DESIGN This is a narrative review that integrates findings from various studies on mitochondrial dynamics and metabolism in macrophages, their interactions with the endoplasmic reticulum (ER) and Golgi apparatus, and their implications for CVDs. The review also considers the potential therapeutic effects of pharmacological agents on these pathways. METHODS The review utilizes a comprehensive literature search to identify relevant studies on mitochondrial dynamics and metabolism in macrophages, their role in CVDs, and the effects of pharmacological agents on these pathways. The selected studies are analyzed and synthesized to provide insights into the complex relationships between mitochondria, the ER, and Golgi apparatus, and their implications for macrophage function and fate. RESULTS The review reveals that mitochondrial metabolism intertwines with cellular architecture and function, particularly through its intricate interactions with the ER and Golgi apparatus. Mitochondrial-associated membranes (MAMs) facilitate Ca2+ transfer from the ER to mitochondria, maintaining mitochondrial homeostasis during ER stress. The Golgi apparatus transports proteins crucial for inflammatory signaling, contributing to immune responses. Inflammation-induced metabolic reprogramming in macrophages, characterized by a shift from oxidative phosphorylation to glycolysis, underscores the multifaceted role of mitochondrial metabolism in regulating immune cell polarization and inflammatory outcomes. Notably, mitochondrial dysfunction, marked by heightened reactive oxygen species generation, fuels inflammatory cascades and promotes cell death, exacerbating CVD pathology. However, pharmacological agents such as Metformin, Nitazoxanide, and Galanin emerge as potential therapeutic modulators of these pathways, offering avenues for mitigating CVD progression. CONCLUSION This review highlights mitochondrial dynamics and metabolism in macrophage inflammation and cell death in CVDs, suggesting therapeutic targets to improve macrophage resilience and reduce pathology, with new pharmacological agents offering treatment opportunities.
Collapse
Affiliation(s)
- Yi-Lang Zhong
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Chen-Qin Xu
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Ji Li
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Zhi-Qiang Liang
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Miao-Miao Wang
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Chao Ma
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Cheng-Lin Jia
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yong-Bing Cao
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Jian Chen
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Anhui Province Rural Revitalization Collaborative Technical Service Center, Huangshan University, Huangshan 245041, China; Department of Public Health, International College, Krirk University, Bangkok, Thailand.
| |
Collapse
|
2
|
Chang X, Li Z, Tian M, Deng Z, Zhu L, Li G. Rotenone activates the LKB1-AMPK-ULK1 signaling pathway to induce autophagy and apoptosis in rat thoracic aortic endothelial cells. BMC Pharmacol Toxicol 2024; 25:33. [PMID: 38783387 PMCID: PMC11118107 DOI: 10.1186/s40360-024-00755-5] [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: 05/05/2023] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND The specific mechanism by which rotenone impacts thoracic aortic autophagy and apoptosis is unknown. We aimed to investigate the regulatory effects of rotenone on autophagy and apoptosis in rat thoracic aortic endothelial cells (RTAEC) via activation of the LKB1-AMPK-ULK1 signaling pathway and to elucidate the molecular mechanisms of rotenone on autophagy and apoptosis in vascular endothelial cells. METHODS In vivo, 60 male SD rats were randomly selected and divided into 5 groups: control (Con), DMSO, 1, 2, and 4 mg/kg groups, respectively. After 28 days of treatment, histopathological and ultrastructural changes in each group were observed using HE and transmission electron microscopy; Autophagy, apoptosis, and LKB1-AMPK-ULK1 pathway-related proteins were detected by Western blot; Apoptosis levels in the thoracic aorta were detected by TUNEL. In vitro, RTAEC were cultured and divided into control (Con), DMSO, 20, 100, 500, and 1000 nM groups. After 24 h of intervention, autophagy, apoptosis, and LKB1-AMPK-ULK1 pathway-related factors were detected by Western blot and qRT-PCR; Flow cytometry to detect apoptosis levels; Autophagy was inhibited with 3-MA and CQ to detect apoptosis levels, and changes in autophagy, apoptosis, and downstream factors were detected by the AMPK inhibitor CC intervention. RESULTS Gavage in SD rats for 28 days, some degree of damage was observed in the thoracic aorta and heart of the rotenone group, as well as the appearance of autophagic vesicles was observed in the thoracic aorta. TUNEL analysis revealed higher apoptosis in the rotenone group's thoracic aorta; RTAEC cultured in vitro, after 24 h of rotenone intervention, showed increased ROS production and significantly decreased ATP production. The flow cytometry data suggested an increase in the number of apoptotic RTAEC. The thoracic aorta and RTAEC in the rotenone group displayed elevated levels of autophagy and apoptosis, and the LKB1-AMPK-ULK1 pathway proteins were activated and expressed at higher levels. Apoptosis and autophagy were both suppressed by the autophagy inhibitors 3-MA and CQ. The AMPK inhibitor CC reduced autophagy and apoptosis in RTAEC and suppressed the production of the AMPK downstream factors ULK1 and P-ULK1. CONCLUSIONS Rotenone may promote autophagy in the thoracic aorta and RTAEC by activating the LKB1-AMPK-ULK1 signaling pathway, thereby inducing apoptosis.
Collapse
Affiliation(s)
- Xiaoyu Chang
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China
| | - Zeyuan Li
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China
| | - Mi Tian
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China
| | - Ziwei Deng
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China
| | - Lingqin Zhu
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China.
| | - Guanghua Li
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China.
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
3
|
Wang Y, Wang T, Han Z, Wang R, Hu Y, Yang Z, Shen T, Zheng Y, Luo J, Ma Y, Luo Y, Jiao L. Explore the role of long noncoding RNAs and mRNAs in intracranial atherosclerotic stenosis: From the perspective of neutrophils. Brain Circ 2023; 9:240-250. [PMID: 38284107 PMCID: PMC10821680 DOI: 10.4103/bc.bc_63_23] [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: 07/29/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 01/30/2024] Open
Abstract
CONTEXT Circulating neutrophils and long noncoding RNAs (lncRNAs) play various roles in intracranial atherosclerotic stenosis (ICAS). OBJECTIVE Our study aimed to detect differentially expressed (DE) lncRNAs and mRNAs in circulating neutrophils and explore the pathogenesis of atherosclerosis from the perspective of neutrophils. METHODS Nineteen patients with ICAS and 15 healthy controls were enrolled. The peripheral blood of the participants was collected, and neutrophils were separated. The expression profiles of lncRNAs and mRNAs in neutrophils from five patients and five healthy controls were obtained, and DE lncRNAs and mRNAs were selected. Six lncRNAs were selected and validated using quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and ceRNA and lncRNA-RNA binding protein (RBP)-mRNA networks were constructed. Correlation analysis between lncRNAs and mRNAs was performed. Functional enrichment annotations were also performed. RESULTS Volcano plots and heat maps displayed the expression profiles and DE lncRNAs and mRNAs, respectively. The qRT-PCR results revealed that the four lncRNAs showed a tendency consistent with the expression profile, with statistical significance. The ceRNA network revealed three pairs of regulatory networks: lncRNA RP3-406A7.3-NAGLU, lncRNA HOTAIRM1-MVK/IL-25/GBF1/CNOT4/ANKK1/PLEKHG6, and lncRNA RP11-701H16.4-ZNF416. The lncRNA-RBP-mRNA network showed five pairs of regulatory networks: lncRNA RP11-701H16.4-TEK, lncRNA RP11-701H16.4-MED17, lncRNA SNHG19-NADH-ubiquinone oxidoreductase core subunit V1, lncRNA RP3-406A7.3-Angel1, and lncRNA HOTAIRM1-CARD16. CONCLUSIONS Our study identified and verified four lncRNAs in neutrophils derived from peripheral blood, which may explain the transcriptional alteration of neutrophils during the pathophysiological process of ICAS. Our results provide insights for research related to the pathogenic mechanisms and drug design of ICAS.
Collapse
Affiliation(s)
- Yilin Wang
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ziping Han
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Rongliang Wang
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yue Hu
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zhenhong Yang
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Tong Shen
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yangmin Zheng
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yan Ma
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yumin Luo
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Miao G, Zhao X, Chan SL, Zhang L, Li Y, Zhang Y, Zhang L, Wang B. Vascular smooth muscle cell c-Fos is critical for foam cell formation and atherosclerosis. Metabolism 2022; 132:155213. [PMID: 35513168 DOI: 10.1016/j.metabol.2022.155213] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Hyperlipidemia-induced vascular smooth muscle cell (VSMC)-derived foam cell formation is considered a crucial event in the development of atherosclerosis. Since c-Fos emerges as a key modulator of lipid metabolism, we investigated whether c-Fos plays a role in hyperlipidemia-induced VSMC-derived foam cell formation and atherosclerosis. APPROACH AND RESULTS c-Fos expression was observed in VSMCs in atherosclerotic plaques from patients and western diet-fed atherosclerosis-prone LDLR-/- and ApoE-/- mice by immunofluorescence staining. To ascertain c-Fos's function in atherosclerosis development, VSMC-specific c-Fos deficient mice in ApoE-/- background were established. Western diet-fed c-FosVSMCKOApoE-/- mice exhibited a significant reduction of atherosclerotic lesion formation as measured by hematoxylin and eosin staining, accompanied by decreased lipid deposition within aortic roots as determined by Oil red O staining. Primary rat VSMCs were isolated to examine the role of c-Fos in lipid uptake and foam cell formation. oxLDL stimulation resulted in VSMC-derived foam cell formation and elevated intracellular mitochondrial reactive oxygen species (mtROS), c-Fos and LOX-1 levels, whereas specific inhibition of mtROS, c-Fos or LOX-1 lessened lipid accumulation in oxLDL-stimulated VSMCs. Mechanistically, oxLDL acts through mtROS to enhance transcription activity of c-Fos to facilitate the expression of LOX-1, exerting a feedforward mechanism with oxLDL to increase lipid uptake and propel VSMC-derived foam cell formation and atherogenesis. CONCLUSION Our study demonstrates a fundamental role of mtROS/c-Fos/LOX-1 signaling pathway in promoting oxLDL uptake and VSMC-derived foam cell formation during atherosclerosis. c-Fos may represent a promising therapeutic target amenable to clinical translation in the future.
Collapse
Affiliation(s)
- Guolin Miao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, China
| | - Xi Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Siu-Lung Chan
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yaohua Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuke Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| | - Beibei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
5
|
Cai Y, Li Z. Mathematical modeling of plaque progression and associated microenvironment: How far from predicting the fate of atherosclerosis? COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2021; 211:106435. [PMID: 34619601 DOI: 10.1016/j.cmpb.2021.106435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/16/2021] [Indexed: 06/13/2023]
Abstract
Mathematical modeling contributes to pathophysiological research of atherosclerosis by helping to elucidate mechanisms and by providing quantitative predictions that can be validated. In turn, the complexity of atherosclerosis is well suited to quantitative approaches as it provides challenges and opportunities for new developments of modeling. In this review, we summarize the current 'state of the art' on the mathematical modeling of the effects of biomechanical factors and microenvironmental factors on the plaque progression, and its potential help in prediction of plaque development. We begin with models that describe the biomechanical environment inside and outside the plaque and its influence on its growth and rupture. We then discuss mathematical models that describe the dynamic evolution of plaque microenvironmental factors, such as lipid deposition, inflammation, smooth muscle cells migration and intraplaque hemorrhage, followed by studies on plaque growth and progression using these modelling approaches. Moreover, we present several key questions for future research. Mathematical models can complement experimental and clinical studies, but also challenge current paradigms, redefine our understanding of mechanisms driving plaque vulnerability and propose future potential direction in therapy for cardiovascular disease.
Collapse
Affiliation(s)
- Yan Cai
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Zhiyong Li
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China; School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4001, Australia
| |
Collapse
|
6
|
Li J, Chen J, Zhang F, Li J, An S, Cheng M, Li J. LncRNA CDKN2B-AS1 hinders the proliferation and facilitates apoptosis of ox-LDL-induced vascular smooth muscle cells via the ceRNA network of CDKN2B-AS1/miR-126-5p/PTPN7. Int J Cardiol 2021; 340:79-87. [PMID: 34384839 DOI: 10.1016/j.ijcard.2021.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/27/2021] [Accepted: 08/04/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The patterns of lncRNA CDKN2B-AS1 in coronary heart disease (CHD) have been extensively studied. This study investigated the competing endogenous RNA (ceRNA) network of CDKN2B-AS1 in coronary atherosclerosis (CAS). METHODS Microarray analyses were performed to screen out the CHD-related lncRNAs (CDKN2B-AS1) and the downstream microRNAs (miR-126-5p). The expression of CDKN2B-AS1 in serum of patients with CHD and healthy volunteers was detected. Vascular smooth muscle cells (VSMCs) were treated with oxidized low density lipoprotein (ox-LDL) to establish the cell model. Then pcDNA-CDKN2B-AS1 and/or miR-126-5p mimic were transfected into ox-LDL-treated VSMCs to estimate cell proliferation, apoptosis and inflammation. The ceRNA network of CDKN2B-AS1 along with the possible pathway in CHD was testified. RESULTS CDKN2B-AS1 expression was low in patients with CHD and ox-LDL-treated VSMCs. Upon CDKN2B-AS1 overexpression, TNF-α, NF-κB and IL-1β levels in VSMCs were decreased, the proliferation of VSMCs was inhibited and the apoptosis rate was increased. Overexpression of miR-126-5p could reverse these trends. CDKN2B-AS1 as a ceRNA competitively bound to miR-126-5p to upregulate PTPN7. CDKN2B-AS1 inhibited VSMC proliferation and accelerated apoptosis by inhibiting the PI3K-Akt pathway. CONCLUSION LncRNA CDKN2B-AS1 upregulates PTPN7 by absorbing miR-126-5p and inhibits the PI3K-Akt pathway, thus hindering the proliferation and accelerating apoptosis of VSMCs induced by ox-LDL, thus being a therapeutic approach for CAS.
Collapse
Affiliation(s)
- Jie Li
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Jia Chen
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Fan Zhang
- Department of Cardiac Vascular Surgery, Linfen City Center Hospital, Linfen 041000, Shanxi, China
| | - Jianfeng Li
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Shoukuan An
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Ming Cheng
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China.
| | - Junquan Li
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
7
|
Zhu Y, Han XQ, Sun XJ, Yang R, Ma WQ, Liu NF. Lactate accelerates vascular calcification through NR4A1-regulated mitochondrial fission and BNIP3-related mitophagy. Apoptosis 2021; 25:321-340. [PMID: 31993850 DOI: 10.1007/s10495-020-01592-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Arterial media calcification is related to mitochondrial dysfunction. Protective mitophagy delays the progression of vascular calcification. We previously reported that lactate accelerates osteoblastic phenotype transition of VSMC through BNIP3-mediated mitophagy suppression. In this study, we investigated the specific links between lactate, mitochondrial homeostasis, and vascular calcification. Ex vivo, alizarin S red and von Kossa staining in addition to measurement of calcium content, RUNX2, and BMP-2 protein levels revealed that lactate accelerated arterial media calcification. We demonstrated that lactate induced mitochondrial fission and apoptosis in aortas, whereas mitophagy was suppressed. In VSMCs, lactate increased NR4A1 expression, leading to activation of DNA-PKcs and p53. Lactate induced Drp1 migration to the mitochondria and enhanced mitochondrial fission through NR4A1. Western blot analysis of LC3-II and p62 and mRFP-GFP-LC3 adenovirus detection showed that NR4A1 knockdown was involved in enhanced autophagy flux. Furthermore, NR4A1 inhibited BNIP3-related mitophagy, which was confirmed by TOMM20 and BNIP3 protein levels, and LC3-II co-localization with TOMM20. The excessive fission and deficient mitophagy damaged mitochondrial structure and impaired respiratory function, determined by mPTP opening rate, mitochondrial membrane potential, mitochondrial morphology under TEM, ATP production, and OCR, which was reversed by NR4A1 silencing. Mechanistically, lactate enhanced fission but halted mitophagy via activation of the NR4A1/DNA-PKcs/p53 pathway, evoking apoptosis, finally accelerating osteoblastic phenotype transition of VSMC and calcium deposition. This study suggests that the NR4A1/DNA-PKcs/p53 pathway is involved in the mechanism by which lactate accelerates vascular calcification, partly through excessive Drp-mediated mitochondrial fission and BNIP3-related mitophagy deficiency.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
| | - Xi-Qiong Han
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
| | - Xue-Jiao Sun
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
| | - Rui Yang
- Pharmaceutical Department, Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
| | - Wen-Qi Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
8
|
Ma WQ, Sun XJ, Zhu Y, Liu NF. PDK4 promotes vascular calcification by interfering with autophagic activity and metabolic reprogramming. Cell Death Dis 2020; 11:991. [PMID: 33203874 PMCID: PMC7673024 DOI: 10.1038/s41419-020-03162-w] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023]
Abstract
Pyruvate dehydrogenase kinase 4 (PDK4) is an important mitochondrial matrix enzyme in cellular energy regulation. Previous studies suggested that PDK4 is increased in the calcified vessels of patients with atherosclerosis and is closely associated with mitochondrial function, but the precise regulatory mechanisms remain largely unknown. This study aims to investigate the role of PDK4 in vascular calcification and the molecular mechanisms involved. Using a variety of complementary techniques, we found impaired autophagic activity in the process of vascular smooth muscle cells (VSMCs) calcification, whereas knocking down PDK4 had the opposite effect. PDK4 drives the metabolic reprogramming of VSMCs towards a Warburg effect, and the inhibition of PDK4 abrogates VSMCs calcification. Mechanistically, PDK4 disturbs the integrity of the mitochondria-associated endoplasmic reticulum membrane, concomitantly impairing mitochondrial respiratory capacity, which contributes to a decrease in lysosomal degradation by inhibiting the V-ATPase and lactate dehydrogenase B interaction. PDK4 also inhibits the nuclear translocation of the transcription factor EB, thus inhibiting lysosomal function. These changes result in the interruption of autophagic flux, which accelerates calcium deposition in VSMCs. In addition, glycolysis serves as a metabolic adaptation to improve VSMCs oxidative stress resistance, whereas inhibition of glycolysis by 2-deoxy-D-glucose induces the apoptosis of VSMCs and increases the calcium deposition in VSMCs. Our results suggest that PDK4 plays a key role in vascular calcification through autophagy inhibition and metabolic reprogramming.
Collapse
Affiliation(s)
- Wen-Qi Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, P.R. China
| | - Xue-Jiao Sun
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, P.R. China
| | - Yi Zhu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, P.R. China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, P.R. China.
| |
Collapse
|
9
|
Sun C, Fu Y, Gu X, Xi X, Peng X, Wang C, Sun Q, Wang X, Qian F, Qin Z, Qu W, Piao M, Zhong S, Liu S, Zhang M, Fang S, Tian J, Li C, Maegdefessel L, Tian J, Yu B. Macrophage-Enriched lncRNA RAPIA: A Novel Therapeutic Target for Atherosclerosis. Arterioscler Thromb Vasc Biol 2020; 40:1464-1478. [PMID: 32268789 DOI: 10.1161/atvbaha.119.313749] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Despite the current antiatherosclerotic and antithrombotic therapies, the incidence of advanced atherosclerosis-associated clinical events remains high. Whether long noncoding RNAs (lncRNAs) affect the progression of atherosclerosis and whether they are potential targets for the treatment of advanced atherosclerosis are poorly understood. Approach and Results: The progression of atherosclerotic lesions was accompanied by dynamic alterations in lncRNA expression, as revealed by RNA sequencing and quantitative polymerase chain reaction. Among the dynamically changing lncRNAs, we identified a novel lncRNA, lncRNA Associated with the Progression and Intervention of Atherosclerosis (RAPIA), that was highly expressed in advanced atherosclerotic lesions and in macrophages. Inhibition of RAPIA in vivo not only repressed the progression of atherosclerosis but also exerted atheroprotective effects similar to those of atorvastatin on advanced atherosclerotic plaques that had already formed. In vitro assays demonstrated that RAPIA promoted proliferation and reduced apoptosis of macrophages. A molecular sponge interaction between RAPIA and microRNA-183-5p was demonstrated by dual-luciferase reporter and RNA immunoprecipitation assays. Rescue assays indicated that RAPIA functioned at least in part by targeting the microRNA-183-5p/ITGB1 (integrin β1) pathway in macrophages. In addition, the transcription factor FoxO1 (forkhead box O1) could bind to the RAPIA promoter region and facilitate the expression of RAPIA. CONCLUSIONS The progression of atherosclerotic lesions was accompanied by dynamic changes in the expression of lncRNAs. Inhibition of the pivotal lncRNA RAPIA may be a novel preventive and therapeutic strategy for advanced atherosclerosis, especially in patients resistant or intolerant to statins.
Collapse
Affiliation(s)
- Changbin Sun
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Yahong Fu
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Xia Gu
- Department of Cardiology, Heilongjiang Provincial Hospital, Harbin, China (X.G.)
| | - Xiangwen Xi
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Xiang Peng
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Chuhan Wang
- Department of Pathology, Harbin Medical University, China (C.W.)
| | - Qi Sun
- Key Laboratory of Heilongjiang Province for Cancer Prevention and Control, Mudanjiang Medical University, China (Q.S.)
| | - Xueyu Wang
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Fengcui Qian
- School of Medical Informatics, Daqing Campus, Harbin Medical University, China (F.Q., C.L.)
| | - Zhifeng Qin
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Wenbo Qu
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Minghui Piao
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Shan Zhong
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Shengliang Liu
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Maomao Zhang
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Shaohong Fang
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Jiangtian Tian
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Chunquan Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, China (F.Q., C.L.)
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Jinwei Tian
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| | - Bo Yu
- From the Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China (C.S., Y.F., X.X., X.P., X.W., Z.Q., W.Q., M.P., S.Z., S.L., M.Z., S.F., Jiangtian Tian, Jinwei Tian, B.Y.)
| |
Collapse
|